1
|
Tiley JB, Beaudoin JJ, Derebail VK, Murphy WA, Park CC, Veeder JA, Tran L, Beers JL, Jia W, Stewart PW, Brouwer KLR. Altered bile acid and coproporphyrin-I disposition in patients with autosomal dominant polycystic kidney disease. Br J Clin Pharmacol 2024. [PMID: 39317666 DOI: 10.1111/bcp.16221] [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: 12/17/2023] [Revised: 06/22/2024] [Accepted: 07/30/2024] [Indexed: 09/26/2024] Open
Abstract
AIMS Serum, liver and urinary bile acids are increased, and hepatic transport protein levels are decreased in a non-clinical model of polycystic kidney disease. Similar changes in patients with autosomal dominant polycystic kidney disease (ADPKD) may predispose them to drug-induced liver injury (DILI) and hepatic drug-drug interactions (DDIs). Systemic coproporphyrin-I (CP-I), an endogenous biomarker for hepatic OATP1B function and MRP2 substrate, is used to evaluate OATP1B-mediated DDI risk in humans. In this clinical observational cohort-comparison study, bile acid profiles and CP-I concentrations in healthy volunteers and patients with ADPKD were compared. METHODS Serum and urine samples from healthy volunteers (n = 16) and patients with ADPKD (n = 8) were collected. Serum bile acids, and serum and urine CP-I concentrations, were quantified by ultra-performance liquid chromatography tandem mass spectrometry (UPLC-MS/MS). RESULTS Patients with ADPKD exhibited increased serum concentrations of total (1.3-fold) and taurine-conjugated (2.8-fold) bile acids compared to healthy volunteers. Specifically, serum concentrations of six bile acids known to be more hydrophobic/hepatotoxic (glycochenodeoxycholate, taurochenodeoxycholate, taurodeoxycholate, lithocholate, glycolithocholate and taurolithocholate) were increased (1.5-, 2.9-, 2.8-, 1.6-, 1.7- and 2.7-fold, respectively) in patients with ADPKD. Furthermore, serum CP-I concentrations were elevated and the renal clearance of CP-I was reduced in patients with ADPKD compared to healthy volunteers. CONCLUSIONS Increased exposure to bile acids may increase susceptibility to DILI in some patients with ADPKD. Furthermore, the observed increase in serum CP-I concentrations could be attributed, in part, to impaired OATP1B function in patients with ADPKD, which could increase the risk of DDIs involving OATP1B substrates compared to healthy volunteers.
Collapse
Affiliation(s)
- Jacqueline B Tiley
- Division of Pharmacotherapy and Experimental Therapeutics, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - 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
| | - Vimal K Derebail
- UNC Kidney Center, Division of Nephrology and Hypertension, UNC School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - William A Murphy
- Division of Pharmacotherapy and Experimental Therapeutics, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Christine C Park
- Division of Pharmacotherapy and Experimental Therapeutics, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Justin A Veeder
- Division of Pharmacotherapy and Experimental Therapeutics, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Lana Tran
- Division of Pharmacotherapy and Experimental Therapeutics, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Jessica L Beers
- Division of Pharmacotherapy and Experimental Therapeutics, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Wei Jia
- University of Hawaii Cancer Center, Honolulu, Hawaii, USA
| | - Paul W Stewart
- Department of Biostatistics, UNC Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - 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
|
2
|
Łuczykowski K, Warmuzińska N, Jaroch K, Kollmann D, Selzner M, Bojko B. Recent solid-phase microextraction-based analytical approaches for the profiling of biliary bile acids in pre-transplant assessments of liver grafts subjected to normothermic ex vivo liver perfusion. Anal Chim Acta 2024; 1318:342954. [PMID: 39067930 DOI: 10.1016/j.aca.2024.342954] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2024] [Revised: 07/05/2024] [Accepted: 07/06/2024] [Indexed: 07/30/2024]
Abstract
BACKGROUND Liver transplantation is the definitive treatment for end-stage liver failure, but the scarcity of donor organs remains a significant challenge. Leveraging organs from extended criteria donors (ECD) offers a potential avenue to address worldwide shortages, though these organs are more susceptible to post-reperfusion injury. This study explores the use of normothermic ex vivo liver perfusion (NEVLP) as a method for organ preservation - an approach that sustains liver metabolism and facilitates pre-transplant assessments of organ viability via bile analysis. The focal point of this study revolves on the development of analytical methods for determining the bile acid profile throughout the peritransplantation period as a potential indicator of liver function and viability. RESULTS The study optimized and validated a high-throughput analytical method to quantify selected bile acids in bile samples using a thin-film microextraction-liquid chromatography-mass spectrometry (TFME-LC-MS) platform. Furthermore, it introduced a solid-phase microextraction-microfluidic open interface-mass spectrometry (SPME-MOI-MS) method for rapid direct analysis of bile acid isobar groups. In the animal study, discernible variations in the concentrations of specific bile acids were observed between donors after circulatory death (DCD) and heart-beating donors (HBD), particularly following normothermic perfusion and reperfusion. Noteworthy fluctuations in individual bile acid concentrations were observed throughout the entire organ transplantation process, with taurocholic acid (TCA), glycocholic acid (GCA), and glycochenodeoxycholic acid (GCDCA) emerging as promising indicators of organ quality. The efficacy of the SPME-MOI-MS platform in corroborating these trends highlights its potential for real-time bile acid analysis during liver transplantation procedures. SIGNIFICANCE Our findings underscore the efficacy of NEVLP in tandem with advanced bile acid analysis methods as a reliable strategy for pre-transplant assessments of organ viability, potentially increasing the use of ECD organs and reducing organ shortages. The ability to monitor bile acid profiles in real-time provides crucial insights into liver function and ischemic injury, making significant strides in improving transplant outcomes and patient survival rates.
Collapse
Affiliation(s)
- Kamil Łuczykowski
- Department of Pharmacodynamics and Molecular Pharmacology, Faculty of Pharmacy, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Toruń, Bydgoszcz, Poland
| | - Natalia Warmuzińska
- Department of Pharmacodynamics and Molecular Pharmacology, Faculty of Pharmacy, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Toruń, Bydgoszcz, Poland
| | - Karol Jaroch
- Department of Pharmacodynamics and Molecular Pharmacology, Faculty of Pharmacy, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Toruń, Bydgoszcz, Poland
| | - Dagmar Kollmann
- Department of Surgery, Ajmera Transplant Centre, Toronto General Hospital, University Health Network, Toronto, ON M5G 2C4, Canada; Department of General Surgery, Medical University of Vienna, 1090 Vienna, Austria
| | - Markus Selzner
- Department of Surgery, Ajmera Transplant Centre, Toronto General Hospital, University Health Network, Toronto, ON M5G 2C4, Canada
| | - Barbara Bojko
- Department of Pharmacodynamics and Molecular Pharmacology, Faculty of Pharmacy, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Toruń, Bydgoszcz, Poland.
| |
Collapse
|
3
|
Schwarcz S, Kovács P, Nyerges P, Ujlaki G, Sipos A, Uray K, Bai P, Mikó E. The bacterial metabolite, lithocholic acid, has antineoplastic effects in pancreatic adenocarcinoma. Cell Death Discov 2024; 10:248. [PMID: 38782891 PMCID: PMC11116504 DOI: 10.1038/s41420-024-02023-1] [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: 09/06/2023] [Revised: 05/08/2024] [Accepted: 05/10/2024] [Indexed: 05/25/2024] Open
Abstract
Lithocholic acid (LCA) is a secondary bile acid. LCA enters the circulation after bacterial synthesis in the gastrointestinal tract, reaches distantly located cancer cells, and influences their behavior. LCA was considered carcinogenic, but recent studies demonstrated that LCA has antitumor effects. We assessed the possible role of LCA in pancreatic adenocarcinoma. At the serum reference concentration, LCA induced a multi-pronged antineoplastic program in pancreatic adenocarcinoma cells. LCA inhibited cancer cell proliferation and induced mesenchymal-to-epithelial (MET) transition that reduced cell invasion capacity. LCA induced oxidative/nitrosative stress by decreasing the expression of nuclear factor, erythroid 2-like 2 (NRF2) and inducing inducible nitric oxide synthase (iNOS). The oxidative/nitrosative stress increased protein nitration and lipid peroxidation. Suppression of oxidative stress by glutathione (GSH) or pegylated catalase (pegCAT) blunted LCA-induced MET. Antioxidant genes were overexpressed in pancreatic adenocarcinoma and decreased antioxidant levels correlated with better survival of pancreatic adenocarcinoma patients. Furthermore, LCA treatment decreased the proportions of cancer stem cells. Finally, LCA induced total and ATP-linked mitochondrial oxidation and fatty acid oxidation. LCA exerted effects through the farnesoid X receptor (FXR), vitamin D receptor (VDR), and constitutive androstane receptor (CAR). LCA did not interfere with cytostatic agents used in the chemotherapy of pancreatic adenocarcinoma. Taken together, LCA is a non-toxic compound and has antineoplastic effects in pancreatic adenocarcinoma.
Collapse
Affiliation(s)
- Szandra Schwarcz
- Department of Medical Chemistry, Faculty of Medicine, University of Debrecen, Debrecen, 4032, Hungary
| | - Patrik Kovács
- Department of Medical Chemistry, Faculty of Medicine, University of Debrecen, Debrecen, 4032, Hungary
| | - Petra Nyerges
- Department of Medical Chemistry, Faculty of Medicine, University of Debrecen, Debrecen, 4032, Hungary
| | - Gyula Ujlaki
- Department of Medical Chemistry, Faculty of Medicine, University of Debrecen, Debrecen, 4032, Hungary
- HUN-REN-UD Cell Biology and Signaling Research Group, Debrecen, 4032, Hungary
| | - Adrienn Sipos
- Department of Medical Chemistry, Faculty of Medicine, University of Debrecen, Debrecen, 4032, Hungary
- HUN-REN-UD Cell Biology and Signaling Research Group, Debrecen, 4032, Hungary
| | - Karen Uray
- Department of Medical Chemistry, Faculty of Medicine, University of Debrecen, Debrecen, 4032, Hungary
| | - Péter Bai
- Department of Medical Chemistry, Faculty of Medicine, University of Debrecen, Debrecen, 4032, Hungary
- HUN-REN-UD Cell Biology and Signaling Research Group, Debrecen, 4032, Hungary
- MTA-DE Lendület Laboratory of Cellular Metabolism, Debrecen, 4032, Hungary
- Research Center for Molecular Medicine, Faculty of Medicine, University of Debrecen, Debrecen, 4032, Hungary
| | - Edit Mikó
- Department of Medical Chemistry, Faculty of Medicine, University of Debrecen, Debrecen, 4032, Hungary.
- MTA-DE Lendület Laboratory of Cellular Metabolism, Debrecen, 4032, Hungary.
| |
Collapse
|
4
|
Dekker SEI, Bierau J, Giera M, Blomberg N, Drenth JPH, Mayboroda OA, de Fijter JW, Soonawala D. Serum bile acids associate with liver volume in polycystic liver disease and decrease upon treatment with lanreotide. Eur J Clin Invest 2024; 54:e14147. [PMID: 38071418 DOI: 10.1111/eci.14147] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/04/2023] [Revised: 11/27/2023] [Accepted: 11/29/2023] [Indexed: 03/13/2024]
Abstract
BACKGROUND Polycystic liver disease (PLD) is a common extrarenal manifestation of autosomal dominant polycystic kidney disease (ADPKD). Bile acids may play a role in PLD pathogenesis. We performed a post-hoc exploratory analysis of bile acids in ADPKD patients, who had participated in a trial on the effect of a somatostatin analogue. Our hypothesis was that serum bile acid levels increase in PLD, and that lanreotide, which reduces liver growth, may also reduce bile acid levels. Furthermore, in PLD, urinary excretion of bile acids might contribute to renal disease. METHODS With liquid chromatography-mass spectrometry, 11 bile acids in serum and 6 in urine were quantified in 105 PLD ADPKD patients and 52 age-, sex-, mutation- and eGFR-matched non-PLD ADPKD patients. Sampling was done at baseline and after 120 weeks of either lanreotide or standard care. RESULTS Baseline serum levels of taurine- and glycine-conjugated bile acids were higher in patients with larger livers. In PLD patients, multiple bile acids decreased upon treatment with lanreotide but remained stable in untreated subjects. Changes over time did not correlate with changes in liver volume. Urine bile acid levels did not change and did not correlate with renal disease progression. CONCLUSION In ADPKD patients with PLD, baseline serum bile acids were associated with liver volume. Lanreotide reduced bile acid levels and has previously been shown to reduce liver volume. However, in this study, the decrease in bile acids was not associated with the change in liver volume.
Collapse
Affiliation(s)
- Shosha E I Dekker
- Department of Nephrology, Leiden University Medical Center, Leiden, the Netherlands
| | - Jörgen Bierau
- Department of Clinical Genetics, Erasmus Medical Center, Rotterdam, the Netherlands
| | - Martin Giera
- Center for Proteomics and Metabolomics, Leiden University Medical Center, Leiden, the Netherlands
| | - Niek Blomberg
- Center for Proteomics and Metabolomics, Leiden University Medical Center, Leiden, the Netherlands
| | - Joost P H Drenth
- Department of Gastroenterology and Hepatology, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Oleg A Mayboroda
- Center for Proteomics and Metabolomics, Leiden University Medical Center, Leiden, the Netherlands
| | - Johan W de Fijter
- Department of Nephrology, Leiden University Medical Center, Leiden, the Netherlands
| | - Darius Soonawala
- Department of Nephrology, Leiden University Medical Center, Leiden, the Netherlands
- Department of Internal Medicine, Haga Teaching Hospital, The Hague, the Netherlands
| |
Collapse
|
5
|
Li M, Zhou Y, Zhu H, Xu LM, Ping J. Danhongqing formula alleviates cholestatic liver fibrosis by downregulating long non-coding RNA H19 derived from cholangiocytes and inhibiting hepatic stellate cell activation. JOURNAL OF INTEGRATIVE MEDICINE 2024; 22:188-198. [PMID: 38472011 DOI: 10.1016/j.joim.2024.03.006] [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: 10/31/2023] [Accepted: 02/18/2024] [Indexed: 03/14/2024]
Abstract
OBJECTIVE This study explores the mechanism of action of Danhongqing formula (DHQ), a compound-based Chinese medicine formula, in the treatment of cholestatic liver fibrosis. METHODS In vivo experiments were conducted using 8-week-old multidrug resistance protein 2 knockout (Mdr2-/-) mice as an animal model of cholestatic liver fibrosis. DHQ was administered orally for 8 weeks, and its impact on cholestatic liver fibrosis was evaluated by assessing liver function, liver histopathology, and the expression of liver fibrosis-related proteins. Real-time polymerase chain reaction, Western blot, immunohistochemistry and other methods were used to observe the effects of DHQ on long non-coding RNA H19 (H19) and signal transducer and activator of transcription 3 (STAT3) phosphorylation in the liver tissue of Mdr2-/- mice. In addition, cholangiocytes and hepatic stellate cells (HSCs) were cultured in vitro to measure the effects of bile acids on cholangiocyte injury and H19 expression. Cholangiocytes overexpressing H19 were constructed, and a conditioned medium containing H19 was collected to measure its effects on STAT3 protein expression and cell activation. The intervention effect of DHQ on these processes was also investigated. HSCs overexpressing H19 were constructed to measure the impact of H19 on cell activation and assess the intervention effect of DHQ. RESULTS DHQ alleviated liver injury, ductular reaction, and fibrosis in Mdr2-/- mice, and inhibited H19 expression, STAT3 expression and STAT3 phosphorylation. This formula also reduced hydrophobic bile acid-induced cholangiocyte injury and the upregulation of H19, inhibited the activation of HSCs induced by cholangiocyte-derived conditioned medium, and decreased the expression of activation markers in HSCs. The overexpression of H19 in a human HSC line confirmed that H19 promoted STAT3 phosphorylation and HSC activation, and DHQ was able to successfully inhibit these effects. CONCLUSION DHQ effectively alleviated spontaneous cholestatic liver fibrosis in Mdr2-/- mice by inhibiting H19 upregulation in cholangiocytes and preventing the inhibition of STAT3 phosphorylation in HSC, thereby suppressing cell activation. Please cite this article as: Li M, Zhou Y, Zhu H, Xu LM, Ping J. Danhongqing formula alleviates cholestatic liver fibrosis by downregulating long non-coding RNA H19 derived from cholangiocytes and inhibiting hepatic stellate cell activation. J Integr Med. 2024; 22(2): 188-198.
Collapse
Affiliation(s)
- Meng Li
- Institute of Liver Diseases, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Yang Zhou
- Preventive Treatment Center, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 200437, China
| | - Hui Zhu
- Department of Gastroenterology, Suzhou Traditional Chinese Medicine Hospital, Suzhou 215000, Jiangsu Province, China
| | - Lie-Ming Xu
- Institute of Liver Diseases, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China; Shanghai Key Laboratory of Traditional Chinese Medicine, Shanghai 201203, China; Key Laboratory of Liver and Kidney Diseases (Ministry of Education), Shanghai 201203, China.
| | - Jian Ping
- Institute of Liver Diseases, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China; Shanghai Key Laboratory of Traditional Chinese Medicine, Shanghai 201203, China; Key Laboratory of Liver and Kidney Diseases (Ministry of Education), Shanghai 201203, China.
| |
Collapse
|
6
|
Islam D, Israr I, Taleb MAB, Rao A, Yosief R, Sultana R, Sampaziotis F, Tysoe OC, Trauner M, Karpen SJ, Ghanekar A, Kamath BM. A novel model to study mechanisms of cholestasis in human cholangiocytes reveals a role for the SIPR2 pathway. Hepatol Commun 2024; 8:e0389. [PMID: 38407207 PMCID: PMC10898671 DOI: 10.1097/hc9.0000000000000389] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Accepted: 12/09/2023] [Indexed: 02/27/2024] Open
Abstract
BACKGROUND Ductular reactivity is central to the pathophysiology of cholangiopathies. Mechanisms underlying the reactive phenotype activation by exogenous inflammatory mediators and bile acids are poorly understood. METHODS Using human extrahepatic cholangiocyte organoids (ECOs) we developed an injury model emulating the cholestatic microenvironment with exposure to inflammatory mediators and various pathogenic bile acids. Moreover, we explored roles for the bile acid activated Sphingosine-1-phosphate receptor 2 (S1PR2) and potential beneficial effects of therapeutic bile acids UDCA and norUDCA. RESULTS Synergistic exposure to bile acids (taurocholic acid, glycocholic acid, glycochenodeoxycholic acid) and TNF-α for 24 hours induced a reactive state as measured by ECO diameter, proliferation, lactate dehydrogenase activity and reactive phenotype markers. While NorUDCA and UDCA treatments given 8 hours after injury induction both suppressed reactive phenotype activation and most injury parameters, proliferation was improved by NorUDCA only. Extrahepatic cholangiocyte organoid stimulation with S1PR2 agonist sphingosine-1-phosphate reproduced the cholangiocyte reactive state and upregulated S1PR2 downstream mediators; these effects were suppressed by S1PR2 antagonist JET-013 (JET), downstream mediator extracellular signal-regulated kinase 1/2 inhibitor, and by norUDCA or UDCA treatments. JET also partially suppressed reactive phenotype after bile acid injury. CONCLUSIONS We developed a novel model to study the reactive cholangiocyte state in response to pathological stimuli in cholestasis and demonstrated a contributory role of S1PR2 signaling in both injury and NorUDCA/UDCA treatments. This model is a valuable tool to further explore the pathophysiology of human cholangiopathies.
Collapse
Affiliation(s)
- Diana Islam
- Development & Stem Cell Biology program, Peter Gilligan Centre for Research and Learning, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Izza Israr
- Development & Stem Cell Biology program, Peter Gilligan Centre for Research and Learning, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Mohamed A. B. Taleb
- Development & Stem Cell Biology program, Peter Gilligan Centre for Research and Learning, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Aditya Rao
- Development & Stem Cell Biology program, Peter Gilligan Centre for Research and Learning, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Robel Yosief
- Development & Stem Cell Biology program, Peter Gilligan Centre for Research and Learning, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Rukhsar Sultana
- Development & Stem Cell Biology program, Peter Gilligan Centre for Research and Learning, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Fotios Sampaziotis
- Wellcome–MRC Cambridge Stem Cell Institute, Department of Medicine, University of Cambridge, Cambridge, Cambridgeshire, UK
- Department of Medicine, University of Cambridge, Cambridge, Cambridgeshire, UK
| | - Olivia C. Tysoe
- Wellcome–MRC Cambridge Stem Cell Institute, Department of Medicine, University of Cambridge, Cambridge, Cambridgeshire, UK
| | - Michael Trauner
- Hans Popper Laboratory of Molecular Hepatology, Department of Internal Medicine III, Division of Gastroenterology and Hepatology, Medical University of Vienna, Vienna, Austria
| | - Saul J. Karpen
- Division of Pediatric Gastroenterology, Department of Pediatrics, Hepatology, and Nutrition, Children’s Healthcare of Atlanta and Emory University School of Medicine, Atlanta, Georgia, USA
| | - Anand Ghanekar
- Division of General Surgery, Department of Surgery, University Health Network & The Hospital for Sick Children, University of Toronto, Toronto, Ontario, Canada
| | - Binita M. Kamath
- Development & Stem Cell Biology program, Peter Gilligan Centre for Research and Learning, The Hospital for Sick Children, Toronto, Ontario, Canada
- Division of Gastroenterology, Hepatology and Nutrition, Department of Pediatrics, The Hospital for Sick Children and the University of Toronto, Toronto, Canada
| |
Collapse
|
7
|
Marciano LPA, Costa LF, Cardoso NS, Freire J, Feltrim F, Oliveira GS, Paula FBA, Silvério ACP, Martins I. Biomonitoring and risk assessment of human exposure to triazole fungicides. Regul Toxicol Pharmacol 2024; 147:105565. [PMID: 38185363 DOI: 10.1016/j.yrtph.2024.105565] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2023] [Revised: 12/23/2023] [Accepted: 01/04/2024] [Indexed: 01/09/2024]
Abstract
Risk assessment and biomarkers were evaluated in volunteers exposed to triazole fungicides in southern Minas Gerais, Brazil. Volunteers were divided into two groups: occupationally and environmentally exposed to pesticides (n = 140) and those unexposed (n = 50) from urban areas. Urine samples were analyzed by GC-MS for triazoles, and samples from men and women in the exposed group were quantified. Groups were further stratified by sex to evaluate the biomarkers results. Oxidative stress was indicated by biomarker analysis for occupationally exposed men with elevated malondialdehyde levels and reduced superoxide dismutase and catalase activity (p < 0.0001). Bile acid levels were also elevated in the exposed group (p < 0.0001). Biomarkers in this study suggest recent, reversible changes due to pesticide exposure. Liver enzyme levels showed no significant differences. The highest Estimated Daily Intake for epoxiconazole ranged from 0.534 to 6.31 μg/kg-bw/day for men and 0.657-8.77 μg/kg-bw/day for women in the exposed group. Considering the highest detected urinary triazole value, the calculated Hazard Quotient for epoxiconazole was 0.789 for men and 1.1 for women. Results indicate a health risk associated with environmental triazole exposure, highlighting the importance of biomonitoring in risk assessment to prevent intoxication and assist in mitigating adverse health effects from chronic pesticide exposure.
Collapse
Affiliation(s)
- Luiz P A Marciano
- Laboratory of Toxicant and Drug Analyses, Department of Clinical and Toxicological Analysis, Gabriel Monteiro da Silva St. 700, Federal University of Alfenas - Unifal-MG, 37130-000, Alfenas, MG, Brazil.
| | - Luiz F Costa
- Laboratory of Toxicant and Drug Analyses, Department of Clinical and Toxicological Analysis, Gabriel Monteiro da Silva St. 700, Federal University of Alfenas - Unifal-MG, 37130-000, Alfenas, MG, Brazil.
| | - Naiane S Cardoso
- Clinical and Experimental Analysis Laboratory, Department of Clinical and Toxicological Analysis, Gabriel Monteiro da Silva St. 700, Federal University of Alfenas - Unifal-MG, 37130-000, Alfenas, MG, Brazil.
| | - Josiane Freire
- Laboratory of Toxicant and Drug Analyses, Department of Clinical and Toxicological Analysis, Gabriel Monteiro da Silva St. 700, Federal University of Alfenas - Unifal-MG, 37130-000, Alfenas, MG, Brazil.
| | - Fernando Feltrim
- Laboratory of Toxicant and Drug Analyses, Department of Clinical and Toxicological Analysis, Gabriel Monteiro da Silva St. 700, Federal University of Alfenas - Unifal-MG, 37130-000, Alfenas, MG, Brazil.
| | - Geovana S Oliveira
- Laboratory of Toxicant and Drug Analyses, Department of Clinical and Toxicological Analysis, Gabriel Monteiro da Silva St. 700, Federal University of Alfenas - Unifal-MG, 37130-000, Alfenas, MG, Brazil.
| | - Fernanda B A Paula
- Clinical and Experimental Analysis Laboratory, Department of Clinical and Toxicological Analysis, Gabriel Monteiro da Silva St. 700, Federal University of Alfenas - Unifal-MG, 37130-000, Alfenas, MG, Brazil.
| | | | - Isarita Martins
- Laboratory of Toxicant and Drug Analyses, Department of Clinical and Toxicological Analysis, Gabriel Monteiro da Silva St. 700, Federal University of Alfenas - Unifal-MG, 37130-000, Alfenas, MG, Brazil.
| |
Collapse
|
8
|
Kong L, Ma J, Dong L, Zhu C, Zhang J, Li J. Metformin exerts anti-liver fibrosis effect based on the regulation of gut microbiota homeostasis and multi-target synergy. Heliyon 2024; 10:e24610. [PMID: 38288020 PMCID: PMC10823097 DOI: 10.1016/j.heliyon.2024.e24610] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2023] [Revised: 01/09/2024] [Accepted: 01/11/2024] [Indexed: 01/31/2024] Open
Abstract
Liver fibrosis can progress to cirrhosis if left untreated. Therefore, identifying effective antifibrotic drugs is crucial. This study aimed to investigate the role and potential mechanism of metformin in treating hepatic fibrosis based on the synergistic effect of multiple targets and the "intestine-liver axis" theory. A CCl4-induced liver fibrosis mouse model was established. We measured liver function, liver fibrosis indicators, oxidative stress and inflammation indices. Hematoxylin and eosin and Masson's trichrome staining were used to detect collagen deposition. The expression of apoptotic proteins, TGF-β/Smads and TIMP-1/MMPs was assessed. 16S rRNA and untargeted metabolomics (liquid chromatography-mass spectrometry) were used to assess mouse intestinal flora and metabolites, performing a comprehensive correlation analysis. Metformin improved the general status and liver function and decreased liver collagen deposition in CCl4-induced liver fibrotic mice. Compared with the control group, IL-6, TNF-α and COX-2 serum levels in the liver fibrosis group increased. Although not significantly different, the serum inflammatory marker levels in the metformin group were lower than those in the model group. Metformin decreased serum MDA and increased serum SOD activity, which increased and decreased, respectively, in the model group. Furthermore, metformin inhibited liver cell apoptosis, TGF-β1 expression and TIMP-1, while promoting Smad7 expression, MMP-1 and MMP-2 in fibrotic mice. 16S rRNA analysis indicated that metformin significantly ameliorated the Bacteroides, Helicobacter, Parabacteroides and Parasutterella imbalance. We identified 385 differential metabolites between the metformin and model groups. Prevotella abundance significantly decreased in the metformin group and positively correlated with decreased taurocholic acid levels. Metformin potentially reverses liver fibrosis by inhibiting inflammation, mitigating oxidative stress damage and suppressing hepatocyte apoptosis via intestinal flora metabolite regulation. Metformin also regulates the TGF-β/Smads and TIMP-1/MMPs signalling pathways. This study provides a theoretical basis for the clinical use of metformin in patients with liver fibrosis.
Collapse
Affiliation(s)
- Lianhua Kong
- Department of Infectious Disease, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, Jiangsu, China
| | - Juncong Ma
- Department of Emergency. Lian Shui People's Hospital, Huai'an, 223400, Jiangsu, China
| | - Li Dong
- Department of Infectious Disease, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, Jiangsu, China
| | - Chuanlong Zhu
- Department of Infectious Disease, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, Jiangsu, China
| | - Jie Zhang
- Department of Endocrinology, The Affiliated Huai'an Hospital of Xuzhou Medical University, Huai'an, 223002, Jiangsu, China
| | - Jun Li
- Department of Infectious Disease, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, Jiangsu, China
| |
Collapse
|
9
|
Lu Q, Zhu Y, Wang C, Zhang R, Miao Y, Chai Y, Jiang Z, Yu Q. Obeticholic acid protects against lithocholic acid-induced exogenous cell apoptosis during cholestatic liver injury. Life Sci 2024; 337:122355. [PMID: 38104861 DOI: 10.1016/j.lfs.2023.122355] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2023] [Revised: 12/02/2023] [Accepted: 12/12/2023] [Indexed: 12/19/2023]
Abstract
AIMS Lithocholic acid (LCA)-induced cholestasis was accompanied by the occurrence of apoptosis, which indicated that anti-apoptosis was a therapeutic strategy for primary biliary cholangitis (PBC). As an agonist of (Farnesoid X receptor) FXR, we supposed that the hepatoprotection of Obeticholic acid (OCA) against cholestatic liver injury is related to anti-apoptosis beside of the bile acids (BAs) regulation. Herein, we explored the non-metabolic regulating mechanism of OCA for resisting LCA-induced cholestatic liver injury via anti-apoptosis. MAIN METHODS LCA-induced cholestatic liver injury mice were pretreated with OCA to evaluate its hepatoprotective effect and mechanism. Biochemical and pathological indicators were used to detect the protective effect of OCA on LCA-induced cholestatic liver injury. The bile acids (BAs) profile in serum was detected by LC-MS/MS. Hepatocyte BAs metabolism, apoptosis and inflammation related genes and proteins alteration were investigated by biochemical determination. KEY FINDINGS OCA improved LCA-induced cholestasis and hepatic apoptosis in mice. The BA profile in serum was changed by OCA mainly manifested as a reduction of taurine-conjugated bile acids, which was due to the upregulation of FXR-related bile acid efflux transporters bile salt export pump (BSEP), multi-drug resistant associated protein 2 (MRP2), MRP3 and multi-drug resistance 3 (MDR3). Apoptosis related proteins cleaved caspase-3, cleaved caspase-8 and cleaved PARP were obviously reduced after OCA treatment. SIGNIFICANCE OCA improved LCA-induced cholestatic liver injury via FXR-induced exogenous cell apoptosis, which will provide new evidence for the application of OCA to ameliorate PBC in clinical.
Collapse
Affiliation(s)
- Qian Lu
- New Drug Screening Center, Jiangsu Center for Pharmacodynamics Research and Evaluation, China Pharmaceutical University, Nanjing 210009, China
| | - Yangping Zhu
- New Drug Screening Center, Jiangsu Center for Pharmacodynamics Research and Evaluation, China Pharmaceutical University, Nanjing 210009, China
| | - Changling Wang
- New Drug Screening Center, Jiangsu Center for Pharmacodynamics Research and Evaluation, China Pharmaceutical University, Nanjing 210009, China
| | - Rongmi Zhang
- New Drug Screening Center, Jiangsu Center for Pharmacodynamics Research and Evaluation, China Pharmaceutical University, Nanjing 210009, China
| | - Yingying Miao
- New Drug Screening Center, Jiangsu Center for Pharmacodynamics Research and Evaluation, China Pharmaceutical University, Nanjing 210009, China
| | - Yuanyuan Chai
- New Drug Screening Center, Jiangsu Center for Pharmacodynamics Research and Evaluation, China Pharmaceutical University, Nanjing 210009, China
| | - Zhenzhou Jiang
- New Drug Screening Center, Jiangsu Center for Pharmacodynamics Research and Evaluation, China Pharmaceutical University, Nanjing 210009, China; Key Laboratory of Drug Quality Control and Pharmacovigilance, Ministry of Education, China Pharmaceutical University, Nanjing 210009, China; Jiangsu Key Laboratory of Druggability of Biopharmaceuticals, Nanjing 210009, China.
| | - Qinwei Yu
- New Drug Screening Center, Jiangsu Center for Pharmacodynamics Research and Evaluation, China Pharmaceutical University, Nanjing 210009, China.
| |
Collapse
|
10
|
Zhang SS, Du J, Cui N, Yang X, Zhang L, Zhang WX, Yue M, Wu YX, Yang T, Zhang XA, Yang ZD, Lv HD, Lu QB, Liu W. Clinical efficacy of immunoglobulin on the treatment of severe fever with thrombocytopenia syndrome: a retrospective cohort study. EBioMedicine 2023; 96:104807. [PMID: 37738834 PMCID: PMC10520313 DOI: 10.1016/j.ebiom.2023.104807] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2023] [Revised: 08/29/2023] [Accepted: 09/05/2023] [Indexed: 09/24/2023] Open
Abstract
BACKGROUND Optimal treatment strategy for severe fever with thrombocytopenia syndrome (SFTS) remained unknown. We aimed to evaluate the efficacy of intravenous immunoglobulin (IVIG) on SFTS. METHODS A retrospective cohort study was conducted based on medical records of the laboratory-confirmed SFTS patients hospitalized during 2010-2020 in the 154th hospital, China. A 1:1 propensity score matching with age, sex, the interval from symptom onset to admission, presence of chronic viral hepatitis, diabetes and disease severity was performed between Non-IVIG group (supportive therapy) and IVIG group (IVIG plus supportive therapy). The matching variables were adjusted to compare the case fatality rates (CFRs), viral load and laboratory parameters between the two groups. Risk ratio (RR) and 95% confidence interval (CI) were reported. FINDINGS Totally 2219 SFTS patients were recruited. CFRs were significantly higher in 1051 patients in IVIG group than 1168 patients in Non-IVIG group (19.0% vs. 4.6%, RR = 4.30, 95% CI 3.12-5.93). The difference remained significant after matching (17.2% vs. 5.1%, RR = 4.02, 95% CI 2.71-5.97). The CFR of IVIG group was significantly higher in all age groups, two IVIG therapy delay groups and two therapy duration groups compared to that of Non-IVIG group (all P < 0.05). IVIG therapy was related to higher viral loads and reduced counts of lymphocytes, T cells, CD4+ T cells and natural killer cells in the blood (all P < 0.05). INTERPRETATION No obvious efficacy of IVIG in saving life or improving outcome of SFTS was observed. Caution is needed for clinical physicians to continue prescribing IVIG for SFTS patients. FUNDING Natural Science Foundation of China.
Collapse
Affiliation(s)
- Shan-Shan Zhang
- Department of Laboratorial Science and Technology & Vaccine Research Center, School of Public Health, Peking University, Beijing, China; Center for Infectious Disease and Policy Research & Global Health and Infectious Diseases Group, Peking University, Beijing, China
| | - Juan Du
- Center for Infectious Disease and Policy Research & Global Health and Infectious Diseases Group, Peking University, Beijing, China
| | - Ning Cui
- The 154th Hospital, Xinyang, China
| | - Xin Yang
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing, China
| | | | - Wan-Xue Zhang
- Center for Infectious Disease and Policy Research & Global Health and Infectious Diseases Group, Peking University, Beijing, China
| | - Ming Yue
- Department of Infectious Diseases, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China; Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, China
| | - Yong-Xiang Wu
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing, China
| | - Tong Yang
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing, China
| | - Xiao-Ai Zhang
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing, China
| | | | | | - Qing-Bin Lu
- Department of Laboratorial Science and Technology & Vaccine Research Center, School of Public Health, Peking University, Beijing, China; Center for Infectious Disease and Policy Research & Global Health and Infectious Diseases Group, Peking University, Beijing, China; Key Laboratory of Epidemiology of Major Diseases (Peking University), Ministry of Education, Beijing, China.
| | - Wei Liu
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing, China; Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, China; School of Public Health, Anhui Medical University, Hefei, China.
| |
Collapse
|
11
|
Muranaka H, Hendifar A, Osipov A, Moshayedi N, Placencio-Hickok V, Tatonetti N, Stotland A, Parker S, Van Eyk J, Pandol SJ, Bhowmick NA, Gong J. Plasma Metabolomics Predicts Chemotherapy Response in Advanced Pancreatic Cancer. Cancers (Basel) 2023; 15:3020. [PMID: 37296982 PMCID: PMC10252041 DOI: 10.3390/cancers15113020] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2023] [Revised: 05/26/2023] [Accepted: 05/30/2023] [Indexed: 06/12/2023] Open
Abstract
Pancreatic cancer (PC) is one of the deadliest cancers. Developing biomarkers for chemotherapeutic response prediction is crucial for improving the dismal prognosis of advanced-PC patients (pts). To evaluate the potential of plasma metabolites as predictors of the response to chemotherapy for PC patients, we analyzed plasma metabolites using high-performance liquid chromatography-mass spectrometry from 31 cachectic, advanced-PC subjects enrolled into the PANCAX-1 (NCT02400398) prospective trial to receive a jejunal tube peptide-based diet for 12 weeks and who were planned for palliative chemotherapy. Overall, there were statistically significant differences in the levels of intermediates of multiple metabolic pathways in pts with a partial response (PR)/stable disease (SD) vs. progressive disease (PD) to chemotherapy. When stratified by the chemotherapy regimen, PD after 5-fluorouracil-based chemotherapy (e.g., FOLFIRINOX) was associated with decreased levels of amino acids (AAs). For gemcitabine-based chemotherapy (e.g., gemcitabine/nab-paclitaxel), PD was associated with increased levels of intermediates of glycolysis, the TCA cycle, nucleoside synthesis, and bile acid metabolism. These results demonstrate the feasibility of plasma metabolomics in a prospective cohort of advanced-PC patients for assessing the effect of enteral feeding as their primary source of nutrition. Metabolic signatures unique to FOLFIRINOX or gemcitabine/nab-paclitaxel may be predictive of a patient's response and warrant further study.
Collapse
Affiliation(s)
- Hayato Muranaka
- Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA; (H.M.); (A.H.); (A.O.); (N.M.); (V.P.-H.); (S.J.P.); (N.A.B.)
- Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Andrew Hendifar
- Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA; (H.M.); (A.H.); (A.O.); (N.M.); (V.P.-H.); (S.J.P.); (N.A.B.)
- Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Arsen Osipov
- Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA; (H.M.); (A.H.); (A.O.); (N.M.); (V.P.-H.); (S.J.P.); (N.A.B.)
- Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Natalie Moshayedi
- Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA; (H.M.); (A.H.); (A.O.); (N.M.); (V.P.-H.); (S.J.P.); (N.A.B.)
- Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Veronica Placencio-Hickok
- Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA; (H.M.); (A.H.); (A.O.); (N.M.); (V.P.-H.); (S.J.P.); (N.A.B.)
- Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Nicholas Tatonetti
- Department of Computational Biomedicine, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA;
| | - Aleksandr Stotland
- Advanced Clinical Biosystems Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA; (A.S.); (S.P.); (J.V.E.)
| | - Sarah Parker
- Advanced Clinical Biosystems Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA; (A.S.); (S.P.); (J.V.E.)
| | - Jennifer Van Eyk
- Advanced Clinical Biosystems Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA; (A.S.); (S.P.); (J.V.E.)
| | - Stephen J. Pandol
- Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA; (H.M.); (A.H.); (A.O.); (N.M.); (V.P.-H.); (S.J.P.); (N.A.B.)
- Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Neil A. Bhowmick
- Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA; (H.M.); (A.H.); (A.O.); (N.M.); (V.P.-H.); (S.J.P.); (N.A.B.)
- Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
- Department of Research, VA Greater Los Angeles Healthcare System, Los Angeles, CA 90073, USA
| | - Jun Gong
- Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA; (H.M.); (A.H.); (A.O.); (N.M.); (V.P.-H.); (S.J.P.); (N.A.B.)
- Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| |
Collapse
|
12
|
Schwarcz S, Kovács P, Kovács T, Ujlaki G, Nyerges P, Uray K, Bai P, Mikó E. The pro- and antineoplastic effects of deoxycholic acid in pancreatic adenocarcinoma cell models. Mol Biol Rep 2023; 50:5273-5282. [PMID: 37145211 DOI: 10.1007/s11033-023-08453-x] [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: 12/09/2022] [Accepted: 04/12/2023] [Indexed: 05/06/2023]
Abstract
BACKGROUND Commensal bacteria secrete metabolites that reach distant cancer cells through the circulation and influence cancer behavior. Deoxycholic acid (DCA), a hormone-like metabolite, is a secondary bile acid specifically synthesized by intestinal microbes. DCA may have both pro- and antineoplastic effects in cancers. METHODS AND RESULTS The pancreatic adenocarcinoma cell lines, Capan-2 and BxPC-3, were treated with 0.7 µM DCA, which corresponds to the reference concentration of DCA in human serum. DCA influenced the expression of epithelial to mesenchymal transition (EMT)-related genes, significantly decreased the expression level of the mesenchymal markers, transcription factor 7- like 2 (TCF7L2), snail family transcriptional repressor 2 (SLUG), CLAUDIN-1, and increased the expression of the epithelial genes, zona occludens 1 (ZO-1) and E-CADHERIN, as shown by real-time PCR and Western blotting. Consequently, DCA reduced the invasion capacity of pancreatic adenocarcinoma cells in Boyden chamber experiments. DCA induced the protein expression of oxidative/nitrosative stress markers. Moreover, DCA reduced aldehyde dehydrogenase 1 (ALDH1) activity in an Aldefluor assay and ALDH1 protein level, suggesting that DCA reduced stemness in pancreatic adenocarcinoma. In Seahorse experiments, DCA induced all fractions of mitochondrial respiration and glycolytic flux. The ratio of mitochondrial oxidation and glycolysis did not change after DCA treatment, suggesting that cells became hypermetabolic. CONCLUSION DCA induced antineoplastic effects in pancreatic adenocarcinoma cells by inhibiting EMT, reducing cancer stemness, and inducing oxidative/nitrosative stress and procarcinogenic effects such as hypermetabolic bioenergetics.
Collapse
Affiliation(s)
- Szandra Schwarcz
- Department of Medical Chemistry, University of Debrecen, Egyetem Tér 1., Debrecen, 4032, Hungary
| | - Patrik Kovács
- Department of Medical Chemistry, University of Debrecen, Egyetem Tér 1., Debrecen, 4032, Hungary
| | - Tünde Kovács
- Department of Medical Chemistry, University of Debrecen, Egyetem Tér 1., Debrecen, 4032, Hungary
- MTA-DE Lendület Laboratory of Cellular Metabolism, Debrecen, 4032, Hungary
| | - Gyula Ujlaki
- Department of Medical Chemistry, University of Debrecen, Egyetem Tér 1., Debrecen, 4032, Hungary
- MTA-DE Cell Biology and Signaling Research Group ELKH, Debrecen, 4032, Hungary
| | - Petra Nyerges
- Department of Medical Chemistry, University of Debrecen, Egyetem Tér 1., Debrecen, 4032, Hungary
| | - Karen Uray
- Department of Medical Chemistry, University of Debrecen, Egyetem Tér 1., Debrecen, 4032, Hungary
| | - Péter Bai
- Department of Medical Chemistry, University of Debrecen, Egyetem Tér 1., Debrecen, 4032, Hungary.
- MTA-DE Lendület Laboratory of Cellular Metabolism, Debrecen, 4032, Hungary.
- MTA-DE Cell Biology and Signaling Research Group ELKH, Debrecen, 4032, Hungary.
- Research Center for Molecular Medicine, Faculty of Medicine, University of Debrecen, Debrecen, 4032, Hungary.
| | - Edit Mikó
- Department of Medical Chemistry, University of Debrecen, Egyetem Tér 1., Debrecen, 4032, Hungary.
- MTA-DE Lendület Laboratory of Cellular Metabolism, Debrecen, 4032, Hungary.
| |
Collapse
|
13
|
Minowa K, Rodriguez-Agudo D, Suzuki M, Muto Y, Hirai S, Wang Y, Su L, Zhou H, Chen Q, Lesnefsky EJ, Mitamura K, Ikegawa S, Takei H, Nittono H, Fuchs M, Pandak WM, Kakiyama G. Insulin dysregulation drives mitochondrial cholesterol metabolite accumulation: initiating hepatic toxicity in nonalcoholic fatty liver disease. J Lipid Res 2023; 64:100363. [PMID: 36966904 PMCID: PMC10182330 DOI: 10.1016/j.jlr.2023.100363] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2023] [Revised: 03/09/2023] [Accepted: 03/21/2023] [Indexed: 04/07/2023] Open
Abstract
CYP7B1 catalyzes mitochondria-derived cholesterol metabolites such as (25R)26-hydroxycholesterol (26HC) and 3β-hydroxy-5-cholesten-(25R)26-oic acid (3βHCA) and facilitates their conversion to bile acids. Disruption of 26HC/3βHCA metabolism in the absence of CYP7B1 leads to neonatal liver failure. Disrupted 26HC/3βHCA metabolism with reduced hepatic CYP7B1 expression is also found in nonalcoholic steatohepatitis (NASH). The current study aimed to understand the regulatory mechanism of mitochondrial cholesterol metabolites and their contribution to onset of NASH. We used Cyp7b1-/- mice fed a normal diet (ND), Western diet (WD), or high-cholesterol diet (HCD). Serum and liver cholesterol metabolites as well as hepatic gene expressions were comprehensively analyzed. Interestingly, 26HC/3βHCA levels were maintained at basal levels in ND-fed Cyp7b1-/- mice livers by the reduced cholesterol transport to mitochondria, and the upregulated glucuronidation and sulfation. However, WD-fed Cyp7b1-/- mice developed insulin resistance (IR) with subsequent 26HC/3βHCA accumulation due to overwhelmed glucuronidation/sulfation with facilitated mitochondrial cholesterol transport. Meanwhile, Cyp7b1-/- mice fed an HCD did not develop IR or subsequent evidence of liver toxicity. HCD-fed mice livers revealed marked cholesterol accumulation but no 26HC/3βHCA accumulation. The results suggest 26HC/3βHCA-induced cytotoxicity occurs when increased cholesterol transport into mitochondria is coupled to decreased 26HC/3βHCA metabolism driven with IR. Supportive evidence for cholesterol metabolite-driven hepatotoxicity is provided in a diet-induced nonalcoholic fatty liver mouse model and by human specimen analyses. This study uncovers an insulin-mediated regulatory pathway that drives the formation and accumulation of toxic cholesterol metabolites within the hepatocyte mitochondria, mechanistically connecting IR to cholesterol metabolite-induced hepatocyte toxicity which drives nonalcoholic fatty liver disease.
Collapse
Affiliation(s)
- Kei Minowa
- Department of Internal Medicine, Virginia Commonwealth University School of Medicine, Richmond, VA, USA; Department of Pediatrics, Juntendo University Faculty of Medicine, Tokyo, Japan
| | - Daniel Rodriguez-Agudo
- Department of Internal Medicine, Virginia Commonwealth University School of Medicine, Richmond, VA, USA; Research Services, Central Virginia Veterans Affairs Healthcare System, Richmond, VA, USA
| | - Mitsuyoshi Suzuki
- Department of Pediatrics, Juntendo University Faculty of Medicine, Tokyo, Japan
| | - Yamato Muto
- Department of Pediatrics, Juntendo University Faculty of Medicine, Tokyo, Japan
| | - Saeko Hirai
- Department of Pediatrics, Juntendo University Faculty of Medicine, Tokyo, Japan
| | - Yaping Wang
- Department of Internal Medicine, Virginia Commonwealth University School of Medicine, Richmond, VA, USA; Research Services, Central Virginia Veterans Affairs Healthcare System, Richmond, VA, USA
| | - Lianyong Su
- Research Services, Central Virginia Veterans Affairs Healthcare System, Richmond, VA, USA; Department of Microbiology and Immunology, Virginia Commonwealth University School of Medicine, Richmond, VA, USA
| | - Huiping Zhou
- Research Services, Central Virginia Veterans Affairs Healthcare System, Richmond, VA, USA; Department of Microbiology and Immunology, Virginia Commonwealth University School of Medicine, Richmond, VA, USA
| | - Qun Chen
- Department of Internal Medicine, Virginia Commonwealth University School of Medicine, Richmond, VA, USA; Research Services, Central Virginia Veterans Affairs Healthcare System, Richmond, VA, USA
| | - Edward J Lesnefsky
- Department of Internal Medicine, Virginia Commonwealth University School of Medicine, Richmond, VA, USA; Research Services, Central Virginia Veterans Affairs Healthcare System, Richmond, VA, USA
| | - Kuniko Mitamura
- Department of Pharmaceutical Sciences, Kindai University, Osaka, Japan
| | - Shigeo Ikegawa
- Division of Research and Development, Genmaikoso Co. Ltd., Sapporo, Hokkaido, Japan
| | - Hajime Takei
- Junshin Clinic Bile Acid Institute, Tokyo, Japan
| | | | - Michael Fuchs
- Department of Internal Medicine, Virginia Commonwealth University School of Medicine, Richmond, VA, USA; Research Services, Central Virginia Veterans Affairs Healthcare System, Richmond, VA, USA
| | - William M Pandak
- Department of Internal Medicine, Virginia Commonwealth University School of Medicine, Richmond, VA, USA; Research Services, Central Virginia Veterans Affairs Healthcare System, Richmond, VA, USA; Massey Cancer Center, Virginia Commonwealth University, Richmond, VA, USA
| | - Genta Kakiyama
- Department of Internal Medicine, Virginia Commonwealth University School of Medicine, Richmond, VA, USA; Research Services, Central Virginia Veterans Affairs Healthcare System, Richmond, VA, USA.
| |
Collapse
|
14
|
Hoff J, Xiong L, Kammann T, Neugebauer S, Micheel JM, Gaßler N, Bauer M, Press AT. RIPK3 promoter hypermethylation in hepatocytes protects from bile acid-induced inflammation and necroptosis. Cell Death Dis 2023; 14:275. [PMID: 37072399 PMCID: PMC10113265 DOI: 10.1038/s41419-023-05794-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Revised: 03/24/2023] [Accepted: 04/04/2023] [Indexed: 04/20/2023]
Abstract
Necroptosis facilitates cell death in a controlled manner and is employed by many cell types following injury. It plays a significant role in various liver diseases, albeit the cell-type-specific regulation of necroptosis in the liver and especially hepatocytes, has not yet been conceptualized. We demonstrate that DNA methylation suppresses RIPK3 expression in human hepatocytes and HepG2 cells. In diseases leading to cholestasis, the RIPK3 expression is induced in mice and humans in a cell-type-specific manner. Overexpression of RIPK3 in HepG2 cells leads to RIPK3 activation by phosphorylation and cell death, further modulated by different bile acids. Additionally, bile acids and RIPK3 activation further facilitate JNK phosphorylation, IL-8 expression, and its release. This suggests that hepatocytes suppress RIPK3 expression to protect themselves from necroptosis and cytokine release induced by bile acid and RIPK3. In chronic liver diseases associated with cholestasis, induction of RIPK3 expression may be an early event signaling danger and repair through releasing IL-8.
Collapse
Affiliation(s)
- Jessica Hoff
- Department of Anesthesiology and Intensive Care Medicine, Nanophysiology Group, Jena University Hospital, Jena, 07747, Germany
- Center for Sepsis Control and Care, Jena University Hospital, Jena, 07743, Germany
| | - Ling Xiong
- Department of Anesthesiology and Intensive Care Medicine, Nanophysiology Group, Jena University Hospital, Jena, 07747, Germany
- Center for Sepsis Control and Care, Jena University Hospital, Jena, 07743, Germany
| | - Tobias Kammann
- Department of Anesthesiology and Intensive Care Medicine, Nanophysiology Group, Jena University Hospital, Jena, 07747, Germany
- Center for Sepsis Control and Care, Jena University Hospital, Jena, 07743, Germany
| | - Sophie Neugebauer
- Center for Sepsis Control and Care, Jena University Hospital, Jena, 07743, Germany
- Department of Clinical Chemistry and Laboratory Diagnostics, Jena University Hospital, Jena, 07747, Germany
| | - Julia M Micheel
- Department of Anesthesiology and Intensive Care Medicine, Nanophysiology Group, Jena University Hospital, Jena, 07747, Germany
- Center for Sepsis Control and Care, Jena University Hospital, Jena, 07743, Germany
| | | | - Michael Bauer
- Department of Anesthesiology and Intensive Care Medicine, Nanophysiology Group, Jena University Hospital, Jena, 07747, Germany
- Center for Sepsis Control and Care, Jena University Hospital, Jena, 07743, Germany
| | - Adrian T Press
- Department of Anesthesiology and Intensive Care Medicine, Nanophysiology Group, Jena University Hospital, Jena, 07747, Germany.
- Center for Sepsis Control and Care, Jena University Hospital, Jena, 07743, Germany.
- Faculty of Medicine, Friedrich Schiller University Jena, Jena, 07747, Germany.
| |
Collapse
|
15
|
Harnisch LO, Mihaylov D, Bein T, Apfelbacher C, Moerer O, Quintel M. A reduced glycine-to-taurine ratio of conjugated serum bile acids signifies an adaptive mechanism and is an early marker of outcome in acute respiratory distress syndrome. Intern Emerg Med 2023; 18:607-615. [PMID: 36378472 DOI: 10.1007/s11739-022-03152-0] [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: 08/19/2022] [Accepted: 11/03/2022] [Indexed: 11/16/2022]
Abstract
The accumulation of Bile Acids (BA) in serum is a common finding in critically ill patients and has been found in patients with Acute Respiratory Distress Syndrome (ARDS), where liver and biliary function could be essentially affected by the underlying disease process and subsequent therapeutic measures. We hypothesized that the glycine-to-taurine conjugation ratio (G/T-ratio) is predictive of outcome in ARDS patients and would support our previously published hypothesis that the BA profile reflects a (mal-) adaptive response of bile acid production when suffering from a disease or syndrome such as ARDS. In 70 patients with ARDS, we determined conjugated BA fractions from protein precipitated serum samples using a LC-MS/MS method and calculated the G/T-ratios, which were then compared with a healthy control group. In patients with ARDS, the G/T-ratio was markedly lower compared to the control group, due to an increase in taurine-conjugated BA. The G/T ratio was lowest on the day of diagnosis and increased steadily during the following days (control = 3.80 (2.28-4.44); day 0 = 1.79 (1.31-3.86); day 3 = 2.91 (1.71-5.68); day 5 = 2.28 (1.25-7.85), significant increases were found between day 0 and day 3 (p = 0.019) and between day 0 and day 5 (p = 0.031). G/T-ratio was significantly correlated with SAPS II score on day 0 (p = 0.009) and day 3 (p = 0.036) and with survival (p = 0.006). Regarding survival, the receiver-operator characteristic revealed an area-under-the-curve of 0.713 (CI 0.578-0.848), the Youden index revealed a G/T-ratio cut-off level of 2.835 (sensitivity 78.4%, specificity 63.2%). Our findings further support our previously published hypothesis that alterations in BA profiles represent adaptive mechanisms in states of severe disease. Our current study adds the finding of an increase in taurine-conjugated BA expressed by a decrease in the G/T-ratio of conjugated BA in serum. The G/T-ratio on day 3 using a threshold G/T-ratio of 2.8 was even associated with survival (p = 0.006); these results are yet to be confirmed by subsequent studies.
Collapse
Affiliation(s)
- Lars-Olav Harnisch
- Department of Anaesthesiology, University of Göttingen Medical Center, Robert-Koch-Street 40, 37099, Göttingen, Germany.
| | - Diana Mihaylov
- Institute of Clinical Chemistry and Laboratory Medicine of the University Hospital Jena, Am Klinikum 1, 07747, Jena, Germany
| | - Thomas Bein
- University of Regensburg, Franz-Josef-Strauß-Allee 11, 93053, Regensburg, Germany
| | - Christian Apfelbacher
- Institute for Social Medicine and Health Economics, University of Magdeburg, Leipziger Street 44, 39120, Magdeburg, Germany
| | - Onnen Moerer
- Department of Anaesthesiology, University of Göttingen Medical Center, Robert-Koch-Street 40, 37099, Göttingen, Germany
| | - Michael Quintel
- Department of Anaesthesiology, University of Göttingen Medical Center, Robert-Koch-Street 40, 37099, Göttingen, Germany
| |
Collapse
|
16
|
Eshiaty SA, Abdelsattar S, Sweed D, Abdel-Aziz SA, Elfert A, Elsaid H. The value of blood and urine metabolomics in differential diagnosis of cholestasis in infants. EGYPTIAN LIVER JOURNAL 2023. [DOI: 10.1186/s43066-023-00244-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/11/2023] Open
Abstract
Abstract
Background
Early detection of biliary atresia (BA) is a great challenge providing the main useful way to improve its clinical consequence. Promising metabolomics provides an effective method for determining innovative biomarkers and biochemical ways for improving early diagnosis. This study aimed to determine the benefit of serum and urinary potential bile acid metabolites in the differentiation of BA from non-biliary atresia (non-BA) cases using tandem mass spectrometry (MS/MS). Fourteen bile acids metabolites were measured quantitively by MS/MS in serum and urine samples from 102 cholestatic infants and 102 control infants, in addition to the assay of the total serum bile acid enzymatically.
Results
After the diagnostic clinical and laboratory workflow, cholestatic infants were divided into BA (37 infants) and non-BA (65 infants) subgroups. Remarkably on analysis of serum individual bile acid concentrations, there were significant differences between cholestatic BA and non-BA regarding serum (glycocenodeoxycholic acid (GCDCA), taurochenodeoxycholic acid (TCDCA), taurocholic acid (TCA), and GCDCA/chenodeoxycholic acid (CDCA) ratio) (p < 0.001, for all), while there was no significant difference between the two groups regarding serum level of (cholic acid (CA), glycocholic (GCA), or TCDCA/CDCA ratio). There were no significant differences in either the urinary individual bile acids or urinary primary bile acids (conjugated or unconjugated) between BA and non-BA. Further principal component analysis (PCA) analysis was done and receiver operating characteristic (ROC) analysis was performed using score plots of the positive factors in the first two principal components PC1 (CA, GCA, GCDCA, TCA, TCDCA) and PC2 (CA, CDCA, lithocholic (LCA), ursodeoxycholic acid (UDCA)) for establishing the differences between the two diseased groups and revealed that the area under the curve (AUC) for PC1 was (0.770) higher than AUC for PC2 (0.583) indicating that the positive components of PC1 may be potential biomarkers for differentiation between the two cholestatic groups.
Conclusions
Metabolomics of serum bile acid levels using tandem mass spectrometry might change the paradigm differentiating BA from non-BA saving patients from unnecessary invasive procedures.
Collapse
|
17
|
Beaudoin JJ, Yang K, Adiwidjaja J, Taneja G, Watkins PB, Siler SQ, Howell BA, Woodhead JL. Investigating bile acid-mediated cholestatic drug-induced liver injury using a mechanistic model of multidrug resistance protein 3 (MDR3) inhibition. Front Pharmacol 2023; 13:1085621. [PMID: 36733378 PMCID: PMC9887159 DOI: 10.3389/fphar.2022.1085621] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Accepted: 12/23/2022] [Indexed: 01/18/2023] Open
Abstract
Inhibition of the canalicular phospholipid floppase multidrug resistance protein 3 (MDR3) has been implicated in cholestatic drug-induced liver injury (DILI), which is clinically characterized by disrupted bile flow and damage to the biliary epithelium. Reduction in phospholipid excretion, as a consequence of MDR3 inhibition, decreases the formation of mixed micelles consisting of bile acids and phospholipids in the bile duct, resulting in a surplus of free bile acids that can damage the bile duct epithelial cells, i.e., cholangiocytes. Cholangiocytes may compensate for biliary increases in bile acid monomers via the cholehepatic shunt pathway or bicarbonate secretion, thereby influencing viability or progression to toxicity. To address the unmet need to predict drug-induced bile duct injury in humans, DILIsym, a quantitative systems toxicology model of DILI, was extended by representing key features of the bile duct, cholangiocyte functionality, bile acid and phospholipid disposition, and cholestatic hepatotoxicity. A virtual, healthy representative subject and population (n = 285) were calibrated and validated utilizing a variety of clinical data. Sensitivity analyses were performed for 1) the cholehepatic shunt pathway, 2) biliary bicarbonate concentrations and 3) modes of MDR3 inhibition. Simulations showed that an increase in shunting may decrease the biliary bile acid burden, but raise the hepatocellular concentrations of bile acids. Elevating the biliary concentration of bicarbonate may decrease bile acid shunting, but increase bile flow rate. In contrast to competitive inhibition, simulations demonstrated that non-competitive and mixed inhibition of MDR3 had a profound impact on phospholipid efflux, elevations in the biliary bile acid-to-phospholipid ratio, cholangiocyte toxicity, and adaptation pathways. The model with its extended bile acid homeostasis representation was furthermore able to predict DILI liability for compounds with previously studied interactions with bile acid transport. The cholestatic liver injury submodel in DILIsym accounts for several processes pertinent to bile duct viability and toxicity and hence, is useful for predictions of MDR3 inhibition-mediated cholestatic DILI in humans.
Collapse
Affiliation(s)
- James J. Beaudoin
- DILIsym Services Division, Simulations Plus Inc., Research Triangle Park, NC, United States
| | - Kyunghee Yang
- DILIsym Services Division, Simulations Plus Inc., Research Triangle Park, NC, United States
| | - Jeffry Adiwidjaja
- DILIsym Services Division, Simulations Plus Inc., Research Triangle Park, NC, United States
- Division of Pharmacotherapy and Experimental Therapeutics, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Guncha Taneja
- DILIsym Services Division, Simulations Plus Inc., Research Triangle Park, NC, United States
| | - Paul B. Watkins
- Division of Pharmacotherapy and Experimental Therapeutics, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Scott Q. Siler
- DILIsym Services Division, Simulations Plus Inc., Research Triangle Park, NC, United States
| | - Brett A. Howell
- DILIsym Services Division, Simulations Plus Inc., Research Triangle Park, NC, United States
| | - Jeffrey L. Woodhead
- DILIsym Services Division, Simulations Plus Inc., Research Triangle Park, NC, United States
| |
Collapse
|
18
|
Martinez-Gili L, Pechlivanis A, McDonald JA, Begum S, Badrock J, Dyson JK, Jones R, Hirschfield G, Ryder SD, Sandford R, Rushbrook S, Thorburn D, Taylor-Robinson SD, Crossey MM, Marchesi JR, Mells G, Holmes E, Jones D. Bacterial and metabolic phenotypes associated with inadequate response to ursodeoxycholic acid treatment in primary biliary cholangitis. Gut Microbes 2023; 15:2208501. [PMID: 37191344 PMCID: PMC10190197 DOI: 10.1080/19490976.2023.2208501] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Revised: 04/06/2023] [Accepted: 04/21/2023] [Indexed: 05/17/2023] Open
Abstract
Primary biliary cholangitis (PBC) is a chronic cholestatic liver disease with ursodeoxycholic acid (UDCA) as first-line treatment. Poor response to UDCA is associated with a higher risk of progressing to cirrhosis, but the underlying mechanisms are unclear. UDCA modulates the composition of primary and bacterial-derived bile acids (BAs). We characterized the phenotypic response to UDCA based on BA and bacterial profiles of PBC patients treated with UDCA. Patients from the UK-PBC cohort (n = 419) treated with UDCA for a minimum of 12-months were assessed using the Barcelona dynamic response criteria. BAs from serum, urine, and feces were analyzed using Ultra-High-Performance Liquid Chromatography-Mass Spectrometry and fecal bacterial composition measured using 16S rRNA gene sequencing. We identified 191 non-responders, 212 responders, and a subgroup of responders with persistently elevated liver biomarkers (n = 16). Responders had higher fecal secondary and tertiary BAs than non-responders and lower urinary bile acid abundances, with the exception of 12-dehydrocholic acid, which was higher in responders. The sub-group of responders with poor liver function showed lower alpha-diversity evenness, lower abundance of fecal secondary and tertiary BAs than the other groups and lower levels of phyla with BA-deconjugation capacity (Actinobacteriota/Actinomycetota, Desulfobacterota, Verrucomicrobiota) compared to responders. UDCA dynamic response was associated with an increased capacity to generate oxo-/epimerized secondary BAs. 12-dehydrocholic acid is a potential biomarker of treatment response. Lower alpha-diversity and lower abundance of bacteria with BA deconjugation capacity might be associated with an incomplete response to treatment in some patients.
Collapse
Affiliation(s)
- Laura Martinez-Gili
- Division of Systems Medicine, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, UK
- Division of Digestive Diseases, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, UK
| | - Alexandros Pechlivanis
- Division of Systems Medicine, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, UK
- Biomic_Auth, Bioanalysis and Omics Laboratory, Center for Interdisciplinary Research and Innovation (CIRI-AUTH), Balkan Centre, Thessaloniki, Greece
| | - Julie A.K. McDonald
- Division of Digestive Diseases, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, UK
| | - Sofina Begum
- Division of Digestive Diseases, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, UK
| | - Jonathan Badrock
- Academic Department of Medical Genetics, Cambridge University, Cambridge, UK
| | - Jessica K. Dyson
- Liver Unit, Freeman Hospital, Newcastle Upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne, UK
- Institute of Translational and Clinical Research, Newcastle University, Newcastle upon Tyne, UK
| | - Rebecca Jones
- Leeds Liver Unit, St James’s University Hospital, Leeds, UK
| | - Gideon Hirschfield
- Center for Liver and Gastroenterology Research and National Institute for Health Research (NIHR) Birmingham Biomedical Research Centre, University of Birmingham, Birmingham, UK
| | - Stephen D. Ryder
- NIHR Biomedical Research Centre at Nottingham University Hospitals NHS Trust, University of Nottingham, Nottingham, UK
| | - Richard Sandford
- Academic Department of Medical Genetics, Cambridge University, Cambridge, UK
| | - Simon Rushbrook
- Department of Gastroenterology, Norfolk and Norwich University Hospital, Norwich, UK
| | - Douglas Thorburn
- UCL Royal Free Campus, Royal Free Hospital, University College London Institute of Liver and Digestive Health, London, UK
| | | | - Mary M.E. Crossey
- Division of Digestive Diseases, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, UK
| | - Julian R. Marchesi
- Division of Digestive Diseases, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, UK
| | - George Mells
- Academic Department of Medical Genetics, Cambridge University, Cambridge, UK
- Department of Hepatology, Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
| | - Elaine Holmes
- Division of Digestive Diseases, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, UK
- Center for Computational & Systems Medicine, Murdoch University, Perth, Australia
| | - David Jones
- Liver Unit, Freeman Hospital, Newcastle Upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne, UK
- Institute of Translational and Clinical Research, Newcastle University, Newcastle upon Tyne, UK
| |
Collapse
|
19
|
Yuan Z, Wang J, Zhang H, Chai Y, Xu Y, Miao Y, Yuan Z, Zhang L, Jiang Z, Yu Q. Glycocholic acid aggravates liver fibrosis by promoting the up-regulation of connective tissue growth factor in hepatocytes. Cell Signal 2023; 101:110508. [PMID: 36341984 DOI: 10.1016/j.cellsig.2022.110508] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2022] [Revised: 10/18/2022] [Accepted: 10/26/2022] [Indexed: 11/25/2022]
Abstract
AIMS The precise role of bile acid in the progression of liver fibrosis has yet to be elucidated. In this study, common bile duct ligation was used as an in vivo mouse model for the evaluation of bile acids that promote liver connective tissue growth factor expression. MAIN METHODS Primary rat and mice hepatocytes, as well as primary rat hepatic stellate and HepaRG cells were evaluated as in vitro models for promoting the expression of connective tissue growth factor by bile acids. KEY FINDINGS Compared with taurochenodeoxycholic acid, glycochenodeoxycholic acid, and taurocholic acid, glycocholic acid (GCA) most strongly promoted the secretion of connective tissue growth factor in mouse primary hepatocytes, rat primary hepatocytes and HepaRGs. GCA did not directly promote the activation of hepatic stellate cells. The administration of GCA in mice with ligated bile ducts promotes the progression of liver fibrosis, which may promote the yes-associated protein of hepatocytes into the nucleus, resulting in the hepatocytes secreting more connective tissue growth factor for hepatic stellate cell activation. In conclusion, our data showed that GCA can induce the expression of connective tissue growth factor in hepatocytes by promoting the nuclear translocation of yes-associated protein, thereby activating hepatic stellate cells. SIGNIFICANCE Our findings help to elucidate the contribution of GCA to the progression of hepatic fibrosis in cholestatic disease and aid the clinical monitoring of cholestatic liver fibrosis development.
Collapse
Affiliation(s)
- Zihang Yuan
- New drug screening center, Jiangsu Center for Pharmacodynamics Research and Evaluation, China Pharmaceutical University, Nanjing 210009, China
| | - Jie Wang
- New drug screening center, Jiangsu Center for Pharmacodynamics Research and Evaluation, China Pharmaceutical University, Nanjing 210009, China
| | - Haoran Zhang
- New drug screening center, Jiangsu Center for Pharmacodynamics Research and Evaluation, China Pharmaceutical University, Nanjing 210009, China
| | - Yuanyuan Chai
- New drug screening center, Jiangsu Center for Pharmacodynamics Research and Evaluation, China Pharmaceutical University, Nanjing 210009, China
| | - Yunxia Xu
- New drug screening center, Jiangsu Center for Pharmacodynamics Research and Evaluation, China Pharmaceutical University, Nanjing 210009, China
| | - Yingying Miao
- New drug screening center, Jiangsu Center for Pharmacodynamics Research and Evaluation, China Pharmaceutical University, Nanjing 210009, China
| | - Ziqiao Yuan
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China
| | - Luyong Zhang
- New drug screening center, Jiangsu Center for Pharmacodynamics Research and Evaluation, China Pharmaceutical University, Nanjing 210009, China; Center for Drug Research and Development, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Zhenzhou Jiang
- New drug screening center, Jiangsu Center for Pharmacodynamics Research and Evaluation, China Pharmaceutical University, Nanjing 210009, China; Key Laboratory of Drug Quality Control and Pharmacovigilance, Ministry of Education, China Pharmaceutical University, Nanjing 210009, China; State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China.
| | - Qinwei Yu
- New drug screening center, Jiangsu Center for Pharmacodynamics Research and Evaluation, China Pharmaceutical University, Nanjing 210009, China.
| |
Collapse
|
20
|
Ghallab A, Hassan R, Hofmann U, Friebel A, Hobloss Z, Brackhagen L, Begher-Tibbe B, Myllys M, Reinders J, Overbeck N, Sezgin S, Zühlke S, Seddek AL, Murad W, Brecklinghaus T, Kappenberg F, Rahnenführer J, González D, Goldring C, Copple IM, Marchan R, Longerich T, Vucur M, Luedde T, Urban S, Canbay A, Schreiter T, Trauner M, Akakpo JY, Olyaee M, Curry SC, Sowa JP, Jaeschke H, Hoehme S, Hengstler JG. Interruption of bile acid uptake by hepatocytes after acetaminophen overdose ameliorates hepatotoxicity. J Hepatol 2022; 77:71-83. [PMID: 35131407 PMCID: PMC9209783 DOI: 10.1016/j.jhep.2022.01.020] [Citation(s) in RCA: 43] [Impact Index Per Article: 21.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/24/2021] [Revised: 01/17/2022] [Accepted: 01/23/2022] [Indexed: 12/12/2022]
Abstract
BACKGROUND & AIMS Acetaminophen (APAP) overdose remains a frequent cause of acute liver failure, which is generally accompanied by increased levels of serum bile acids (BAs). However, the pathophysiological role of BAs remains elusive. Herein, we investigated the role of BAs in APAP-induced hepatotoxicity. METHODS We performed intravital imaging to investigate BA transport in mice, quantified endogenous BA concentrations in the serum of mice and patients with APAP overdose, analyzed liver tissue and bile by mass spectrometry and MALDI-mass spectrometry imaging, assessed the integrity of the blood-bile barrier and the role of oxidative stress by immunostaining of tight junction proteins and intravital imaging of fluorescent markers, identified the intracellular cytotoxic concentrations of BAs, and performed interventions to block BA uptake from blood into hepatocytes. RESULTS Prior to the onset of cell death, APAP overdose causes massive oxidative stress in the pericentral lobular zone, which coincided with a breach of the blood-bile barrier. Consequently, BAs leak from the bile canaliculi into the sinusoidal blood, which is then followed by their uptake into hepatocytes via the basolateral membrane, their secretion into canaliculi and repeated cycling. This, what we termed 'futile cycling' of BAs, led to increased intracellular BA concentrations that were high enough to cause hepatocyte death. Importantly, however, the interruption of BA re-uptake by pharmacological NTCP blockage using Myrcludex B and Oatp knockout strongly reduced APAP-induced hepatotoxicity. CONCLUSIONS APAP overdose induces a breach of the blood-bile barrier which leads to futile BA cycling that causes hepatocyte death. Prevention of BA cycling may represent a therapeutic option after APAP intoxication. LAY SUMMARY Only one drug, N-acetylcysteine, is approved for the treatment of acetaminophen overdose and it is only effective when given within ∼8 hours after ingestion. We identified a mechanism by which acetaminophen overdose causes an increase in bile acid concentrations (to above toxic thresholds) in hepatocytes. Blocking this mechanism prevented acetaminophen-induced hepatotoxicity in mice and evidence from patients suggests that this therapy may be effective for longer periods after ingestion compared to N-acetylcysteine.
Collapse
Affiliation(s)
- Ahmed Ghallab
- Leibniz Research Centre for Working Environment and Human Factors, Technical University Dortmund, Ardeystr. 67, 44139, Dortmund, Germany; Department of Forensic Medicine and Toxicology, Faculty of Veterinary Medicine, South Valley University, 83523, Qena, Egypt.
| | - Reham Hassan
- Leibniz Research Centre for Working Environment and Human Factors, Technical University Dortmund, Ardeystr. 67, 44139, Dortmund, Germany,Department of Forensic Medicine and Toxicology, Faculty of Veterinary Medicine, South Valley University, 83523, Qena, Egypt
| | - Ute Hofmann
- Dr. Margarete Fischer-Bosch Institute of Clinical Pharmacology and University of Tübingen, Auerbachstr. 112, 70376 Stuttgart, Germany
| | - Adrian Friebel
- Institute of Computer Science & Saxonian Incubator for Clinical Research (SIKT), University of Leipzig, Haertelstraße 16-18, 04107, Leipzig, Germany
| | - Zaynab Hobloss
- Leibniz Research Centre for Working Environment and Human Factors, Technical University Dortmund, Ardeystr. 67, 44139, Dortmund, Germany
| | - Lisa Brackhagen
- Leibniz Research Centre for Working Environment and Human Factors, Technical University Dortmund, Ardeystr. 67, 44139, Dortmund, Germany
| | - Brigitte Begher-Tibbe
- Leibniz Research Centre for Working Environment and Human Factors, Technical University Dortmund, Ardeystr. 67, 44139, Dortmund, Germany
| | - Maiju Myllys
- Leibniz Research Centre for Working Environment and Human Factors, Technical University Dortmund, Ardeystr. 67, 44139, Dortmund, Germany
| | - Joerg Reinders
- Leibniz Research Centre for Working Environment and Human Factors, Technical University Dortmund, Ardeystr. 67, 44139, Dortmund, Germany
| | - Nina Overbeck
- Leibniz Research Centre for Working Environment and Human Factors, Technical University Dortmund, Ardeystr. 67, 44139, Dortmund, Germany
| | - Selahaddin Sezgin
- Faculty of Chemistry and Chemical Biology, TU Dortmund, Dortmund, Germany
| | - Sebastian Zühlke
- Center for Mass Spectrometry (CMS), Faculty of Chemistry and Chemical Biology, TU Dortmund University, Dortmund, Germany
| | - Abdel-latif Seddek
- Department of Forensic Medicine and Toxicology, Faculty of Veterinary Medicine, South Valley University, 83523, Qena, Egypt
| | - Walaa Murad
- Histology Department, Faculty of Medicine, South Valley University, 83523 Qena, Egypt
| | - Tim Brecklinghaus
- Leibniz Research Centre for Working Environment and Human Factors, Technical University Dortmund, Ardeystr. 67, 44139, Dortmund, Germany
| | | | - Jörg Rahnenführer
- Department of Statistics, TU Dortmund University, 44227, Dortmund, Germany
| | - Daniela González
- Leibniz Research Centre for Working Environment and Human Factors, Technical University Dortmund, Ardeystr. 67, 44139, Dortmund, Germany
| | - Christopher Goldring
- Department of Pharmacology and Therapeutics, MRC Centre of Drug Safety Science, University of Liverpool, The Sherrington Building, Ashton Street, Liverpool, L69 3GE, UK
| | - Ian M. Copple
- Department of Pharmacology and Therapeutics, MRC Centre of Drug Safety Science, University of Liverpool, The Sherrington Building, Ashton Street, Liverpool, L69 3GE, UK
| | - Rosemarie Marchan
- Leibniz Research Centre for Working Environment and Human Factors, Technical University Dortmund, Ardeystr. 67, 44139, Dortmund, Germany
| | - Thomas Longerich
- Translational Gastrointestinal Pathology, Institute of Pathology, University Hospital Heidelberg, Heidelberg, Germany
| | - Mihael Vucur
- Department of Gastroenterology, Hepatology and Infectious Diseases, University Hospital Duesseldorf, Medical Faculty at Heinrich-Heine-University, Dusseldorf, Germany
| | - Tom Luedde
- Department of Gastroenterology, Hepatology and Infectious Diseases, University Hospital Duesseldorf, Medical Faculty at Heinrich-Heine-University, Dusseldorf, Germany
| | - Stephan Urban
- Department of Infectious Diseases, Molecular Virology, University Hospital Heidelberg, Heidelberg, Germany,German Center for Infection Research, Heidelberg University, Heidelberg, Germany
| | - Ali Canbay
- Department of Medicine, Ruhr University Bochum, University Hospital Knappschaftskrankenhaus Bochum, 44892 Bochum, Germany
| | - Thomas Schreiter
- Department of Medicine, Ruhr University Bochum, University Hospital Knappschaftskrankenhaus Bochum, 44892 Bochum, Germany
| | - Michael Trauner
- Hans Popper Laboratory of Molecular Hepatology, Division of Gastroenterology and Hepatology, Department of Internal Medicine III, Medical University of Vienna, Vienna, Austria
| | - Jephte Y. Akakpo
- Department of Pharmacology, Toxicology, and Therapeutics, University of Kansas Medical Center, Kansas City, KS, USA
| | - Mojtaba Olyaee
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, University of Kansas Medical Center, Kansas City, KS, USA
| | - Steven C. Curry
- Division of Clinical Data Analytics and Decision Support, Division of Medical Toxicology and Precision Medicine, Department of Medicine, University of Arizona College of Medicine-Phoenix, Phoenix, AZ, USA
| | - Jan-Peter Sowa
- Department of Medicine, Ruhr University Bochum, University Hospital Knappschaftskrankenhaus Bochum, 44892 Bochum, Germany
| | - Hartmut Jaeschke
- Department of Pharmacology, Toxicology, and Therapeutics, University of Kansas Medical Center, Kansas City, KS, USA
| | - Stefan Hoehme
- Institute of Computer Science & Saxonian Incubator for Clinical Research (SIKT), University of Leipzig, Haertelstraße 16-18, 04107, Leipzig, Germany
| | - Jan G. Hengstler
- Leibniz Research Centre for Working Environment and Human Factors, Technical University Dortmund, Ardeystr. 67, 44139, Dortmund, Germany,Leibniz Research Centre for Working Environment and Human Factors, Technical University Dortmund, Ardeystr. 67, 44139, Dortmund, Germany; telephone: +49 (0)231-1084- 348; Fax: +49 (0)231-1084- 403;
| |
Collapse
|
21
|
Brousseau V, Caron P, Trottier J, Di Paolo T, Milkiewicz P, Barbier O. Liquid chromatography coupled to tandem mass spectrometry methods for the selective and sensitive determination of 24S-hydroxycholesterol, its sulfate, and/or glucuronide conjugates in plasma. JOURNAL OF MASS SPECTROMETRY : JMS 2022; 57:e4827. [PMID: 35460139 DOI: 10.1002/jms.4827] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/22/2021] [Revised: 03/22/2022] [Accepted: 04/03/2022] [Indexed: 06/14/2023]
Abstract
24S-hydroxycholesterol (i.e., cerebrosterol, 24S-OH-Chol) is the main form of cholesterol elimination from the brain. Liquid chromatography-tandem mass spectrometry methods were developed for the quantification of the total and unesterified/unbound fractions of 24S-OH-Chol, its monosulfate, monoglucuronide, and diconjugate derivatives (24S-OH-Chol-3sulfate [3S], 24S-OH-Chol-24glucuronide [24G] and 24S-OH-Chol-3S, 24G, respectively) in human plasma. Linearity, precision, accuracy, and extraction recovery were validated within the typical physiological and pathological ranges of concentrations for each compound. The lower limit of quantifications was 2.00, 0.33, 0.26, and 0.74 ng/ml for 24S-OH-Chol, 24S-OH-Chol-24G, 24S-OH-Chol-3S, and 24-OH-Chol-3S, 24G, respectively. Extraction recovery values in total and unbound plasma fractions were also analyzed in murine and monkey plasma and varied from 73% in mouse to 113% in cynomolgus monkey. The methods could rapidly (less than 7 min) quantify individual compounds with high sensitivity, accuracy (bias ≤15%), and reproducibility (coefficient of variation [CV] ≤ 17%). Their clinical applications were validated by measuring levels of the 4 compounds in samples from 20 noncholestatic donors, 5 cholestatic patients suffering from primary biliary cirrhosis, and 10 patients suffering from biliary stenosis. Results highlight the abundance of 24S-OH-Chol in the total fraction and the abundance of 24S-OH-Chol-3S and 24G in the unbound ones. While the latter strongly accumulate in plasma fractions of cholestatic patients, levels of 24S-OH-Chol remained similar to those of healthy donors. Our results indicate that this approach is suitable for monitoring cerebrosterol and its conjugates in large-scale clinical studies.
Collapse
Affiliation(s)
- Valérie Brousseau
- Laboratory of Molecular Pharmacology, Endocrinology and Nephrology Axis, CHU de Québec Research Centre and the Faculty of Pharmacy, Laval University, Québec City, Quebec, Canada
| | - Patrick Caron
- Laboratory of Molecular Pharmacology, Endocrinology and Nephrology Axis, CHU de Québec Research Centre and the Faculty of Pharmacy, Laval University, Québec City, Quebec, Canada
| | - Jocelyn Trottier
- Laboratory of Molecular Pharmacology, Endocrinology and Nephrology Axis, CHU de Québec Research Centre and the Faculty of Pharmacy, Laval University, Québec City, Quebec, Canada
| | - Thérèse Di Paolo
- Neuroscience Research Unit, CHU de Québec Research Centre and the Faculty of Pharmacy, Laval University, Québec City, Quebec, Canada
| | - Piotr Milkiewicz
- Liver and Internal Medicine Unit, Department of Transplant and Liver Surgery, Medical University of Warsaw, Warszawa, Poland
| | - Olivier Barbier
- Laboratory of Molecular Pharmacology, Endocrinology and Nephrology Axis, CHU de Québec Research Centre and the Faculty of Pharmacy, Laval University, Québec City, Quebec, Canada
| |
Collapse
|
22
|
Režen T, Rozman D, Kovács T, Kovács P, Sipos A, Bai P, Mikó E. The role of bile acids in carcinogenesis. Cell Mol Life Sci 2022; 79:243. [PMID: 35429253 PMCID: PMC9013344 DOI: 10.1007/s00018-022-04278-2] [Citation(s) in RCA: 110] [Impact Index Per Article: 55.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2021] [Revised: 03/03/2022] [Accepted: 03/28/2022] [Indexed: 12/17/2022]
Abstract
AbstractBile acids are soluble derivatives of cholesterol produced in the liver that subsequently undergo bacterial transformation yielding a diverse array of metabolites. The bulk of bile acid synthesis takes place in the liver yielding primary bile acids; however, other tissues have also the capacity to generate bile acids (e.g. ovaries). Hepatic bile acids are then transported to bile and are subsequently released into the intestines. In the large intestine, a fraction of primary bile acids is converted to secondary bile acids by gut bacteria. The majority of the intestinal bile acids undergo reuptake and return to the liver. A small fraction of secondary and primary bile acids remains in the circulation and exert receptor-mediated and pure chemical effects (e.g. acidic bile in oesophageal cancer) on cancer cells. In this review, we assess how changes to bile acid biosynthesis, bile acid flux and local bile acid concentration modulate the behavior of different cancers. Here, we present in-depth the involvement of bile acids in oesophageal, gastric, hepatocellular, pancreatic, colorectal, breast, prostate, ovarian cancer. Previous studies often used bile acids in supraphysiological concentration, sometimes in concentrations 1000 times higher than the highest reported tissue or serum concentrations likely eliciting unspecific effects, a practice that we advocate against in this review. Furthermore, we show that, although bile acids were classically considered as pro-carcinogenic agents (e.g. oesophageal cancer), the dogma that switch, as lower concentrations of bile acids that correspond to their serum or tissue reference concentration possess anticancer activity in a subset of cancers. Differences in the response of cancers to bile acids lie in the differential expression of bile acid receptors between cancers (e.g. FXR vs. TGR5). UDCA, a bile acid that is sold as a generic medication against cholestasis or biliary surge, and its conjugates were identified with almost purely anticancer features suggesting a possibility for drug repurposing. Taken together, bile acids were considered as tumor inducers or tumor promoter molecules; nevertheless, in certain cancers, like breast cancer, bile acids in their reference concentrations may act as tumor suppressors suggesting a Janus-faced nature of bile acids in carcinogenesis.
Collapse
Affiliation(s)
- Tadeja Režen
- Centre for Functional Genomics and Bio-Chips, Institute of Biochemistry and Molecular Genetics, Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| | - Damjana Rozman
- Centre for Functional Genomics and Bio-Chips, Institute of Biochemistry and Molecular Genetics, Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| | - Tünde Kovács
- Department of Medical Chemistry, University of Debrecen, Egyetem tér 1., Debrecen, 4032, Hungary
- MTA-DE Lendület Laboratory of Cellular Metabolism, Debrecen, 4032, Hungary
| | - Patrik Kovács
- Department of Medical Chemistry, University of Debrecen, Egyetem tér 1., Debrecen, 4032, Hungary
| | - Adrienn Sipos
- Department of Medical Chemistry, University of Debrecen, Egyetem tér 1., Debrecen, 4032, Hungary
| | - Péter Bai
- Department of Medical Chemistry, University of Debrecen, Egyetem tér 1., Debrecen, 4032, Hungary
- MTA-DE Lendület Laboratory of Cellular Metabolism, Debrecen, 4032, Hungary
- Research Center for Molecular Medicine, Faculty of Medicine, University of Debrecen, Debrecen, 4032, Hungary
| | - Edit Mikó
- Department of Medical Chemistry, University of Debrecen, Egyetem tér 1., Debrecen, 4032, Hungary.
- MTA-DE Lendület Laboratory of Cellular Metabolism, Debrecen, 4032, Hungary.
| |
Collapse
|
23
|
Urinary BA Indices as Prognostic Biomarkers for Complications Associated with Liver Diseases. Int J Hepatol 2022; 2022:5473752. [PMID: 35402050 PMCID: PMC8986411 DOI: 10.1155/2022/5473752] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Accepted: 02/25/2022] [Indexed: 11/17/2022] Open
Abstract
Hepatobiliary diseases and their complications cause the accumulation of toxic bile acids (BA) in the liver, blood, and other tissues, which may exacerbate the underlying condition and lead to unfavorable prognosis. To develop and validate prognostic biomarkers for the prediction of complications of cholestatic liver disease based on urinary BA indices, liquid chromatography-tandem mass spectrometry was used to analyze urine samples from 257 patients with cholestatic liver diseases during a 7-year follow-up period. The urinary BA profile and non-BA parameters were monitored, and logistic regression models were used to predict the prognosis of hepatobiliary disease-related complications. Urinary BA indices were applied to quantify the composition, metabolism, hydrophilicity, and toxicity of the BA profile. We have developed and validated the bile-acid liver disease complication (BALDC) model based on BA indices using logistic regression model, to predict the prognosis of cholestatic liver disease complications including ascites. The mixed BA and non-BA model was the most accurate and provided higher area under the receiver operating characteristic (ROC) and smaller akaike information criterion (AIC) values compared to both non-BA and MELD (models for end stage liver disease) models. Therefore, the mixed BA and non-BA model could be used to predict the development of ascites in patients diagnosed with liver disease at early stages of intervention. This will help physicians to make a better decision when treating hepatobiliary disease-related ascites.
Collapse
|
24
|
Sestrin2 protects against cholestatic liver injury by inhibiting endoplasmic reticulum stress and NLRP3 inflammasome-mediated pyroptosis. Exp Mol Med 2022; 54:239-251. [PMID: 35260799 PMCID: PMC8980001 DOI: 10.1038/s12276-022-00737-9] [Citation(s) in RCA: 39] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2021] [Revised: 11/18/2021] [Accepted: 12/01/2021] [Indexed: 12/13/2022] Open
Abstract
Chronic exposure to bile acid in the liver due to impaired bile flow induces cholestatic liver disease, resulting in hepatotoxicity and liver fibrosis. Sestrin2, a highly conserved, stress-inducible protein, has been implicated in cellular responses to multiple stress conditions and the maintenance of cellular homeostasis. However, its role in cholestatic liver injury is not fully understood. In this study, we investigated the role of hepatic Sestrin2 in cholestatic liver injury and its underlying mechanisms using in vivo and in vitro approaches. Hepatic Sestrin2 expression was upregulated by activating transcription factor 4 (ATF4) and CCAAT/enhancer-binding protein-β (C/EBP-β) after treatment with bile acids and correlated with endoplasmic reticulum (ER) stress responses. Bile-duct ligation (BDL)-induced hepatocellular apoptosis and liver fibrosis were exacerbated in Sestrin2-knockout (Sesn2−/−) mice. Moreover, Sestrin2 deficiency enhanced cholestasis-induced hepatic ER stress, whereas Sestrin2 overexpression ameliorated bile acid-induced ER stress. Notably, the mammalian target of rapamycin (mTOR) inhibitor rapamycin and the AMP-activated protein kinase (AMPK) activator AICAR reversed bile acid-induced ER stress in Sestrin2-deficient cells. Furthermore, Sestrin2 deficiency promoted cholestasis-induced hepatic pyroptosis by activating NLRP3 inflammasomes. Thus, our study provides evidence for the biological significance of Sestrin2 and its relationship with cholestatic liver injury, suggesting the potential role of Sestrin2 in regulating ER stress and inflammasome activation during cholestatic liver injury. A protein that manages the response to cellular stress can help prevent disruptions in bile flow from progressing to liver fibrosis or failure. Disrupted flow leads to the accumulation of bile acids, which triggers a state known as endoplasmic reticulum (ER) stress, fueling inflammation and eventual cell death. Researchers led by Hwan-Woo Park and Jongdae Shin at Konyang University, Daejon, South Korea, have demonstrated that the Sestrin2 protein plays a prominent role in managing this ER stress response to cytotoxic bile acids in cultured liver cells. They subsequently used a Sestrin2-deficient mouse model to demonstrate that the absence of this protein contributes to heightened ER stress and greatly increased liver damage following impaired bile flow. These results suggest that Sestrin2 modulators could offer effective treatments for liver disorders associated with bile flow obstruction.
Collapse
|
25
|
Azer SA, Hasanato R. Use of bile acids as potential markers of liver dysfunction in humans: A systematic review. Medicine (Baltimore) 2021; 100:e27464. [PMID: 34731122 PMCID: PMC8519223 DOI: 10.1097/md.0000000000027464] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/15/2020] [Accepted: 09/20/2021] [Indexed: 01/05/2023] Open
Abstract
OBJECTIVE This study aimed to determine the effectiveness of using total, individual serum, or urinary bile acids (BA) as potential markers of liver dysfunction. METHODS We searched the PubMed and Web of Science databases using the following keywords- "serum bile acids," "liver dysfunction," "liver injury," "liver disease," "traditional liver function tests," "Chronic liver disease," "acute liver injury". The search was complemented by manual screening of the list of references for relevant articles. We selected only English-language manuscripts for adult patients based on predetermined inclusion and exclusion criteria. Animal studies and studies on neonates and children were not included. OUTCOME MEASURES Changes in BA concentrations or ratios at or prior to changes in liver function tests. RESULTS A total of 547 studies were identified, of which 28 were included after reading the entire manuscript. These studies included 1630 patients and 836 controls published between 1990 and 2017. The methods used in BA assays varied significantly, and the studies did not agree. on specific individual BA or BA ratios as biomarkers of specific liver injury or dysfunction. Except for the prognostic value of BA in intrahepatic cholestasis of pregnancy (ICP), studies have failed to provide evidence for BA as a liver biomarker. CONCLUSIONS Despite the research conducted on BA for over 27 years, there are inconsistencies in the reported results and a lack of solid evidence to support the use of individual BA or BA ratios as biomarkers of liver injury. Adequately conducted studies needed to resolve this limitation in the literature.
Collapse
Affiliation(s)
- Samy A. Azer
- Gastroenterologist and Chair of Curriculum Development and Research Unit, Department of Medical Education, College of Medicine, King Saud University, Riyadh, Saudi Arabia
| | - Rana Hasanato
- Clinical Biochemistry Consultant and Chair of Biochemistry Unit, Director of the Laboratories at King Saud University Medical City, College of Medicine, King Saud University, Riyadh, Saudi Arabia
| |
Collapse
|
26
|
Bile Acids Activate NLRP3 Inflammasome, Promoting Murine Liver Inflammation or Fibrosis in a Cell Type-Specific Manner. Cells 2021; 10:cells10102618. [PMID: 34685598 PMCID: PMC8534222 DOI: 10.3390/cells10102618] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2021] [Revised: 09/10/2021] [Accepted: 09/22/2021] [Indexed: 12/12/2022] Open
Abstract
Bile acids (BA) as important signaling molecules are considered crucial in development of cholestatic liver injury, but there is limited understanding on the involved cell types and signaling pathways. The aim of this study was to evaluate the inflammatory and fibrotic potential of key BA and the role of distinct liver cell subsets focusing on the NLRP3 inflammasome. C57BL/6 wild-type (WT) and Nlrp3−/− mice were fed with a diet supplemented with cholic (CA), deoxycholic (DCA) or lithocholic acid (LCA) for 7 days. Additionally, primary hepatocytes, Kupffer cells (KC) and hepatic stellate cells (HSC) from WT and Nlrp3−/− mice were stimulated with aforementioned BA ex vivo. LCA feeding led to strong liver damage and activation of NLRP3 inflammasome. Ex vivo KC were the most affected cells by LCA, resulting in a pro-inflammatory phenotype. Liver damage and primary KC activation was both ameliorated in Nlrp3-deficient mice or cells. DCA feeding induced fibrotic alterations. Primary HSC upregulated the NLRP3 inflammasome and early fibrotic markers when stimulated with DCA, but not LCA. Pro-fibrogenic signals in liver and primary HSC were attenuated in Nlrp3−/− mice or cells. The data shows that distinct BA induce NLRP3 inflammasome activation in HSC or KC, promoting fibrosis or inflammation.
Collapse
|
27
|
Chang HJ, Zúñiga A, Conejero I, Voyvodic PL, Gracy J, Fajardo-Ruiz E, Cohen-Gonsaud M, Cambray G, Pageaux GP, Meszaros M, Meunier L, Bonnet J. Programmable receptors enable bacterial biosensors to detect pathological biomarkers in clinical samples. Nat Commun 2021; 12:5216. [PMID: 34471137 PMCID: PMC8410942 DOI: 10.1038/s41467-021-25538-y] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2021] [Accepted: 08/12/2021] [Indexed: 12/17/2022] Open
Abstract
Bacterial biosensors, or bactosensors, are promising agents for medical and environmental diagnostics. However, the lack of scalable frameworks to systematically program ligand detection limits their applications. Here we show how novel, clinically relevant sensing modalities can be introduced into bactosensors in a modular fashion. To do so, we have leveraged a synthetic receptor platform, termed EMeRALD (Engineered Modularized Receptors Activated via Ligand-induced Dimerization) which supports the modular assembly of sensing modules onto a high-performance, generic signaling scaffold controlling gene expression in E. coli. We apply EMeRALD to detect bile salts, a biomarker of liver dysfunction, by repurposing sensing modules from enteropathogenic Vibrio species. We improve the sensitivity and lower the limit-of-detection of the sensing module by directed evolution. We then engineer a colorimetric bactosensor detecting pathological bile salt levels in serum from patients having undergone liver transplant, providing an output detectable by the naked-eye. The EMeRALD technology enables functional exploration of natural sensing modules and rapid engineering of synthetic receptors for diagnostics, environmental monitoring, and control of therapeutic microbes.
Collapse
Affiliation(s)
- Hung-Ju Chang
- Centre de Biologie Structurale (CBS), INSERM U1054, CNRS UMR5048, University of Montpellier, Montpellier, France
| | - Ana Zúñiga
- Centre de Biologie Structurale (CBS), INSERM U1054, CNRS UMR5048, University of Montpellier, Montpellier, France
| | - Ismael Conejero
- Centre de Biologie Structurale (CBS), INSERM U1054, CNRS UMR5048, University of Montpellier, Montpellier, France
- Neuropsychiatry: Epidemiological and Clinical Research, Inserm Unit 1061, Montpellier, France
- Department of Psychiatry, CHU Nimes, University of Montpellier, Montpellier, France
| | - Peter L Voyvodic
- Centre de Biologie Structurale (CBS), INSERM U1054, CNRS UMR5048, University of Montpellier, Montpellier, France
| | - Jerome Gracy
- Centre de Biologie Structurale (CBS), INSERM U1054, CNRS UMR5048, University of Montpellier, Montpellier, France
| | - Elena Fajardo-Ruiz
- Centre de Biologie Structurale (CBS), INSERM U1054, CNRS UMR5048, University of Montpellier, Montpellier, France
| | - Martin Cohen-Gonsaud
- Centre de Biologie Structurale (CBS), INSERM U1054, CNRS UMR5048, University of Montpellier, Montpellier, France
| | - Guillaume Cambray
- Centre de Biologie Structurale (CBS), INSERM U1054, CNRS UMR5048, University of Montpellier, Montpellier, France
| | - Georges-Philippe Pageaux
- Department of Hepatogastroenterology, Hepatology and Liver Transplantation Unit, Saint Eloi Hospital, University of Montpellier, Montpellier, France
| | - Magdalena Meszaros
- Department of Hepatogastroenterology, Hepatology and Liver Transplantation Unit, Saint Eloi Hospital, University of Montpellier, Montpellier, France
| | - Lucy Meunier
- Department of Hepatogastroenterology, Hepatology and Liver Transplantation Unit, Saint Eloi Hospital, University of Montpellier, Montpellier, France
| | - Jerome Bonnet
- Centre de Biologie Structurale (CBS), INSERM U1054, CNRS UMR5048, University of Montpellier, Montpellier, France.
| |
Collapse
|
28
|
Sangaraju D, Shi Y, Van Parys M, Ray A, Walker A, Caminiti R, Milanowski D, Jaochico A, Dean B, Liang X. Robust and Comprehensive Targeted Metabolomics Method for Quantification of 50 Different Primary, Secondary, and Sulfated Bile Acids in Multiple Biological Species (Human, Monkey, Rabbit, Dog, and Rat) and Matrices (Plasma and Urine) Using Liquid Chromatography High Resolution Mass Spectrometry (LC-HRMS) Analysis. JOURNAL OF THE AMERICAN SOCIETY FOR MASS SPECTROMETRY 2021; 32:2033-2049. [PMID: 33826317 DOI: 10.1021/jasms.0c00435] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
Bile acids (BAs) are biomolecules synthesized in the liver from cholesterol and are constituents of bile. The in-vivo BA pool includes more than 50 known diverse BAs which are unconjugated, amino acid conjugated, sulfated, and glucuronidated metabolites. Hemostasis of bile acids is known to be highly regulated and an interplay between liver metabolism, gut microbiome function, intestinal absorption, and enterohepatic recirculation. Interruption of BA homeostasis has been attributed to several metabolic diseases and drug induced liver injury (DILI), and their use as potential biomarkers is increasingly becoming important. Speciated quantitative and comprehensive profiling of BAs in various biomatrices from humans and preclinical animal species are important to understand their significance and biological function. Consequently, a versatile one single bioanalytical method for BAs is required to accommodate quantitation in a broad range of biomatrices from human and preclinical animal species. Here we report a versatile, comprehensive, and high throughput liquid chromatography-high resolution mass spectrometry (LC-HRMS) targeted metabolomics method for quantitative analysis of 50 different BAs in multiple matrices including human serum, plasma, and urine and plasma and urine of preclinical animal species (rat, rabbit, dog, and monkey). The method has been sufficiently qualified for accuracy, precision, robustness, and ruggedness and addresses the issue of nonspecific binding of bile acids to plastic for urine samples. Application of this method includes comparison for BA analysis between matched plasma and serum samples, human and animal species differences in BA pools, data analysis, and visualization of complex BA data using BA indices or ratios to understand BA biology, metabolism, and transport.
Collapse
Affiliation(s)
- Dewakar Sangaraju
- Drug Metabolism and Pharmacokinetics, Genentech Inc., 1 DNA Way, South San Francisco, California 94080, United States
| | - Yao Shi
- Bioanalytical Department, Covance Laboratories, Inc., 3301 Kinsman Blvd, Madison, Wisconsin 53704, United States
| | - Michael Van Parys
- Bioanalytical Department, Covance Laboratories, Inc., 3301 Kinsman Blvd, Madison, Wisconsin 53704, United States
| | - Adam Ray
- Bioanalytical Department, Covance Laboratories, Inc., 3301 Kinsman Blvd, Madison, Wisconsin 53704, United States
| | - Abigail Walker
- Bioanalytical Department, Covance Laboratories, Inc., 3301 Kinsman Blvd, Madison, Wisconsin 53704, United States
| | - Rachel Caminiti
- Bioanalytical Department, Covance Laboratories, Inc., 3301 Kinsman Blvd, Madison, Wisconsin 53704, United States
| | - Dennis Milanowski
- Bioanalytical Department, Covance Laboratories, Inc., 3301 Kinsman Blvd, Madison, Wisconsin 53704, United States
| | - Allan Jaochico
- Drug Metabolism and Pharmacokinetics, Genentech Inc., 1 DNA Way, South San Francisco, California 94080, United States
| | - Brian Dean
- Drug Metabolism and Pharmacokinetics, Genentech Inc., 1 DNA Way, South San Francisco, California 94080, United States
| | - Xiaorong Liang
- Drug Metabolism and Pharmacokinetics, Genentech Inc., 1 DNA Way, South San Francisco, California 94080, United States
| |
Collapse
|
29
|
Alamoudi JA, Li W, Gautam N, Olivera M, Meza J, Mukherjee S, Alnouti Y. Bile acid indices as biomarkers for liver diseases I: Diagnostic markers. World J Hepatol 2021; 13:433-455. [PMID: 33959226 PMCID: PMC8080550 DOI: 10.4254/wjh.v13.i4.433] [Citation(s) in RCA: 4] [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] [Received: 01/05/2021] [Revised: 02/11/2021] [Accepted: 03/22/2021] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Hepatobiliary diseases result in the accumulation of toxic bile acids (BA) in the liver, blood, and other tissues which may contribute to an unfavorable prognosis.
AIM To discover and validate diagnostic biomarkers of cholestatic liver diseases based on the urinary BA profile.
METHODS We analyzed urine samples by liquid chromatography-tandem mass spectrometry and compared the urinary BA profile between 300 patients with hepatobiliary diseases vs 103 healthy controls by statistical analysis. The BA profile was characterized using BA indices, which quantifies the composition, metabolism, hydrophilicity, and toxicity of the BA profile. BA indices have much lower inter- and intra-individual variability compared to absolute concentrations of BA. In addition, BA indices demonstrate high area under the receiver operating characteristic curves, and changes of BA indices are associated with the risk of having a liver disease, which demonstrates their use as diagnostic biomarkers for cholestatic liver diseases.
RESULTS Total and individual BA concentrations were higher in all patients. The percentage of secondary BA (lithocholic acid and deoxycholic acid) was significantly lower, while the percentage of primary BA (chenodeoxycholic acid, cholic acid, and hyocholic acid) was markedly higher in patients compared to controls. In addition, the percentage of taurine-amidation was higher in patients than controls. The increase in the non-12α-OH BA was more profound than 12α-OH BA (cholic acid and deoxycholic acid) causing a decrease in the 12α-OH/ non-12α-OH ratio in patients. This trend was stronger in patients with more advanced liver diseases as reflected by the model for end-stage liver disease score and the presence of hepatic decompensation. The percentage of sulfation was also higher in patients with more severe forms of liver diseases.
CONCLUSION BA indices have much lower inter- and intra-individual variability compared to absolute BA concentrations and changes of BA indices are associated with the risk of developing liver diseases.
Collapse
Affiliation(s)
- Jawaher Abdullah Alamoudi
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Nebraska Medical Center, Omaha, NE 68198, United States
- Department of Pharmaceutical Sciences, College of Pharmacy, Princess Nourah Bint Abdulrahman University, Riyadh 11564, Saudi Arabia
| | - Wenkuan Li
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Nebraska Medical Center, Omaha, NE 68198, United States
| | - Nagsen Gautam
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Nebraska Medical Center, Omaha, NE 68198, United States
| | - Marco Olivera
- Department of Internal Medicine, College of Medicine, University of Nebraska Medical Center, Omaha, NE 68198, United States
| | - Jane Meza
- Department of Biostatistics, College of Public Health, University of Nebraska Medical Center, Omaha, NE 68198, United States
| | - Sandeep Mukherjee
- Department of Internal Medicine, College of Medicine, Creighton University Medical Center, Omaha, NE 68124, United States
| | - Yazen Alnouti
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Nebraska Medical Center, Omaha, NE 68198, United States
| |
Collapse
|
30
|
Circulating Bile Acids in Liver Failure Activate TGR5 and Induce Monocyte Dysfunction. Cell Mol Gastroenterol Hepatol 2021; 12:25-40. [PMID: 33545429 PMCID: PMC8082115 DOI: 10.1016/j.jcmgh.2021.01.011] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/19/2020] [Revised: 01/15/2021] [Accepted: 01/15/2021] [Indexed: 12/12/2022]
Abstract
BACKGROUND & AIMS Retention of bile acids in the blood is a hallmark of liver failure. Recent studies have shown that increased serum bile acid levels correlate with bacterial infection and increased mortality. However, the mechanisms by which circulating bile acids influence patient outcomes still are elusive. METHODS Serum bile acid profiles in 33 critically ill patients with liver failure and their effects on Takeda G-protein-coupled receptor 5 (TGR5), an immunomodulatory receptor that is highly expressed in monocytes, were analyzed using tandem mass spectrometry, novel highly sensitive TGR5 bioluminescence resonance energy transfer using nanoluciferase (NanoBRET, Promega Corp, Madison, WI) technology, and in vitro assays with human monocytes. RESULTS Twenty-two patients (67%) had serum bile acids that led to distinct TGR5 activation. These TGR5-activating serum bile acids severely compromised monocyte function. The release of proinflammatory cytokines (eg, tumor necrosis factor α or interleukin 6) in response to bacterial challenge was reduced significantly if monocytes were incubated with TGR5-activating serum bile acids from patients with liver failure. By contrast, serum bile acids from healthy volunteers did not influence cytokine release. Monocytes that did not express TGR5 were protected from the bile acid effects. TGR5-activating serum bile acids were a risk factor for a fatal outcome in patients with liver failure, independent of disease severity. CONCLUSIONS Depending on their composition and quantity, serum bile acids in liver failure activate TGR5. TGR5 activation leads to monocyte dysfunction and correlates with mortality, independent of disease activity. This indicates an active role of TGR5 in liver failure. Therefore, TGR5 and bile acid metabolism might be promising targets for the treatment of immune dysfunction in liver failure.
Collapse
|
31
|
Nicolas NJ, Duffy MA, Hansen A, Aizenberg J. Inverse Opal Films for Medical Sensing: Application in Diagnosis of Neonatal Jaundice. Adv Healthc Mater 2021; 10:e2001326. [PMID: 33191607 DOI: 10.1002/adhm.202001326] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2020] [Revised: 09/24/2020] [Indexed: 12/26/2022]
Abstract
A non-invasive, at-home test for neonatal jaundice can facilitate early jaundice detection in infants, improving clinical outcomes for neonates with severe jaundice and helping to prevent the development of kernicterus, a type of brain damage whose symptoms include hearing loss, impairment of cognitive capacity, and death. Here a photonic sensor that utilizes color changes induced by analyte infiltration into a chemically functionalized inverse opal structure is developed. The sensor is calibrated to detect differences in urinary surface tension due to increased bile salt concentration in urine, which is symptomatic of abnormal liver function and linked to jaundice. The correlation between neonatal urinary surface tension and excess serum bilirubin, the physiologic cause of neonatal jaundice, is explored. It is shown that these non-invasive sensors can improve the preliminary diagnosis of neonatal jaundice, reducing the number of invasive blood tests and hospital visits necessary for healthy infants while ensuring that jaundiced infants are treated in a timely manner. The use of inverse opal sensors to measure bulk property changes in bodily fluids can be extended to the detection of several other conditions, making this technology a versatile platform for convenient point-of-care diagnosis.
Collapse
Affiliation(s)
| | | | - Anne Hansen
- Harvard Medical School 25 Shattuck St Boston MA 02115 USA
- Boston Children's Hospital 300 Longwood Ave Boston MA 02115 USA
| | | |
Collapse
|
32
|
Harnisch LO, Moerer O. The Specific Bile Acid Profile of Shock: A Hypothesis Generating Appraisal of the Literature. J Clin Med 2020; 9:E3844. [PMID: 33256244 PMCID: PMC7761042 DOI: 10.3390/jcm9123844] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2020] [Revised: 11/21/2020] [Accepted: 11/25/2020] [Indexed: 12/29/2022] Open
Abstract
BACKGROUND Bile acid synthesis and regulation of metabolism are tightly regulated. In critical illness, these regulations are impaired. Consequently, the physiologic bile acid pattern in serum becomes disturbed and a disease-specific bile acid profile seems to become evident. METHODS A literature review was performed and trials reporting the broken-down bile acid pattern were condensed with regard to percent differences in bile acid profiles of defined diseases compared to a human control. RESULTS Ten articles were identified. Most of the studied bile acid profiles differ statistically significant between disease states, furthermore, neither of the reported disease entities show the same broken-down pattern of individual bile acids. Deoxycholic acid (DCA) was found to be decreased in almost all diseases, except for the two shock-states investigated (cardiogenic shock, septic shock) where it was elevated by about 100% compared to the control. Moreover, the pattern of both examined shock-states are very similar, rendering a specific shock-pattern possible, that we argue could eventually maintain or even worsen the pathological state. CONCLUSION The specific broken-down bile acid profile of defined diseases might aid in gaining insight into the body's adaptive reaction and the differential diagnosis, as well as in the therapy of disease states in the early course of the disease.
Collapse
Affiliation(s)
- Lars-Olav Harnisch
- Department of Anesthesiology, University of Göttingen, 37075 Göttingen, Germany;
| | | |
Collapse
|
33
|
Žížalová K, Vecka M, Vítek L, Leníček M. Enzymatic methods may underestimate the total serum bile acid concentration. PLoS One 2020; 15:e0236372. [PMID: 32706797 PMCID: PMC7380613 DOI: 10.1371/journal.pone.0236372] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2020] [Accepted: 07/02/2020] [Indexed: 12/15/2022] Open
Abstract
Enzymatic assays based on bacterial 3α-hydroxysteroid dehydrogenase are the method of choice for quantification of total bile acids (BAs) in serum. Although non-specific, it is generally considered precise and robust. The aim of this study was to investigate how changes in the BA spectrum might affect the reliability of the method. We measured standard solutions of twenty-three human and murine BAs using a commercial enzymatic assay and compared the measured vs. expected concentrations. Additionally, total BA concentrations in rat and human cholestatic samples with an abnormal BA spectrum were measured using an enzymatic assay, and a more specific LC-MS/MS method. We observed a great variability in the response of individual BAs in the enzymatic assay. Relative signal intensities ranged from 100% in glycocholic acid (reference) to only 20% in α-muricholic acid. The enzymatic assay markedly underestimated the BA concentrations in both human and rat cholestatic sera when compared to the LC-MS/MS assay. Our study indicated that the performance of an enzymatic assay largely depends on the BA spectrum, and the total concentration of BAs can be markedly underestimated. Samples with an atypical BA spectrum (viz. in rodents) should preferably be measured by other methods.
Collapse
Affiliation(s)
- Kateřina Žížalová
- Institute of Medical Biochemistry and Laboratory Diagnostics, 1 Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Marek Vecka
- Institute of Medical Biochemistry and Laboratory Diagnostics, 1 Faculty of Medicine, Charles University, Prague, Czech Republic
- 4 Department of Internal Medicine, 1 Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Libor Vítek
- Institute of Medical Biochemistry and Laboratory Diagnostics, 1 Faculty of Medicine, Charles University, Prague, Czech Republic
- 4 Department of Internal Medicine, 1 Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Martin Leníček
- Institute of Medical Biochemistry and Laboratory Diagnostics, 1 Faculty of Medicine, Charles University, Prague, Czech Republic
- * E-mail:
| |
Collapse
|
34
|
Abstract
Cholestasis results in blockage of bile flow whether the point of obstruction occurs extrahepatically or intrahepatically. Bile acids are a primary constituent of bile, and thus one of the primary outcomes is acute retention of bile acids in hepatocytes. Bile acids are normally secreted into the biliary tracts and then released into the small bowel before recirculating back to the liver. Retention of bile acids has long been hypothesized to be a primary cause of the associated liver injury that occurs during acute or chronic cholestasis. Despite this, a surge of papers in the last decade have reported a primary role for inflammation in the pathophysiology of cholestatic liver injury. Furthermore, it has increasingly been recognized that both the constituency of individual bile acids that make up the greater pool, as well as their conjugation status, is intimately involved in their toxicity, and this varies between species. Finally, the role of bile acids in drug-induced cholestatic liver injury remains an area of increasing interest. The purpose of this review is to critically evaluate current proposed mechanisms of cholestatic liver injury, with a focus on the evolving role of bile acids in cell death and inflammation.
Collapse
Affiliation(s)
| | - Hartmut Jaeschke
- †Department of Pharmacology, Toxicology and Therapeutics, University of Kansas Medical Center, Kansas City, KS, USA
| |
Collapse
|
35
|
Liu L, Panzitt K, Racedo S, Wagner M, Platzer W, Zaufel A, Theiler‐Schwetz V, Obermayer‐Pietsch B, Müller H, Höfler G, Heinemann A, Zollner G, Fickert P. Bile acids increase steroidogenesis in cholemic mice and induce cortisol secretion in adrenocortical H295R cells via S1PR2, ERK and SF-1. Liver Int 2019; 39:2112-2123. [PMID: 30664326 PMCID: PMC6899711 DOI: 10.1111/liv.14052] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/18/2018] [Revised: 01/10/2019] [Accepted: 01/13/2019] [Indexed: 02/06/2023]
Abstract
BACKGROUND AND AIMS Bile acids are now accepted as central signalling molecules for the regulation of glucose, amino acid and lipid metabolism. Adrenal gland cortex cells express the bile acid receptors farnesoid X receptor (FXR), the G protein-coupled bile acid receptor (TGR5) and the sphingosine-1-phosphate receptor 2 (S1PR2). We aimed to determine the effects of cholestasis and more specifically of bile acids on cortisol production. METHODS FXR and TGR5 knockout mice and controls were subjected to common bile duct ligation (CBDL) or chenodeoxycholic acid (CDCA) feeding to model cholestasis. Human adrenocortical H295R cells were challenged with bile acids for mechanistic studies. RESULTS We found that CBDL and CDCA feeding increased the levels of corticosterone, the rodent equivalent to human cortisol and mRNA and protein levels of steroidogenesis-related enzymes in adrenals independent of FXR and TGR5. Taurine-conjugated CDCA (TCDCA) significantly stimulated cortisol secretion, phosphorylation of extracellular signal-regulated kinase (ERK) and expression of steroidogenesis-related genes in human adrenocortical H295R cells. FXR and TGR5 agonists failed to induce cortisol secretion in H295R cells. S1PR2 inhibition significantly abolished TCDCA-induced cortisol secretion, lowered phosphorylation of ERK and abrogated enhanced transcription of steroidogenesis-related genes in H295R cells. Likewise, siRNA S1PR2 treatment reduced the phosphorylation of ERK and cortisol secretion. Steroidogenic factor-1 (SF-1) transactivation activity was increased upon TCDCA treatment suggesting that bile acid signalling is linked to SF-1. Treatment with SF-1 inverse agonist AC45594 also reduced TCDCA-induced steroidogenesis. CONCLUSIONS Our findings indicate that supraphysiological bile acid levels as observed in cholestasis stimulate steroidogenesis via an S1PR2-ERK-SF-1 signalling pathway.
Collapse
Affiliation(s)
- Lei Liu
- Research Unit for Experimental and Molecular HepatologyDivision of Gastroenterology and HepatologyDepartment of Internal MedicineMedical University of GrazGrazAustria
| | - Katrin Panzitt
- Research Unit for Translational Nuclear Receptor Research in Liver MetabolismDivision of Gastroenterology and HepatologyDepartment of Internal MedicineMedical University of GrazGrazAustria
| | - Silvia Racedo
- Research Unit for Experimental and Molecular HepatologyDivision of Gastroenterology and HepatologyDepartment of Internal MedicineMedical University of GrazGrazAustria
| | - Martin Wagner
- Research Unit for Translational Nuclear Receptor Research in Liver MetabolismDivision of Gastroenterology and HepatologyDepartment of Internal MedicineMedical University of GrazGrazAustria
| | - Wolfgang Platzer
- Institute of Experimental and Clinical PharmacologyMedical University of GrazGrazAustria
| | - Alex Zaufel
- Research Unit for Experimental and Molecular HepatologyDivision of Gastroenterology and HepatologyDepartment of Internal MedicineMedical University of GrazGrazAustria
| | | | | | - Helmut Müller
- Division of Transplant SurgeryMedical University of GrazGrazAustria
| | - Gerald Höfler
- Institute of PathologyMedical University of GrazGrazAustria
| | - Akos Heinemann
- Institute of Experimental and Clinical PharmacologyMedical University of GrazGrazAustria
| | - Gernot Zollner
- Research Unit for Experimental and Molecular HepatologyDivision of Gastroenterology and HepatologyDepartment of Internal MedicineMedical University of GrazGrazAustria
| | - Peter Fickert
- Research Unit for Experimental and Molecular HepatologyDivision of Gastroenterology and HepatologyDepartment of Internal MedicineMedical University of GrazGrazAustria
| |
Collapse
|
36
|
Kenna JG, Taskar KS, Battista C, Bourdet DL, Brouwer KLR, Brouwer KR, Dai D, Funk C, Hafey MJ, Lai Y, Maher J, Pak YA, Pedersen JM, Polli JW, Rodrigues AD, Watkins PB, Yang K, Yucha RW. Can Bile Salt Export Pump Inhibition Testing in Drug Discovery and Development Reduce Liver Injury Risk? An International Transporter Consortium Perspective. Clin Pharmacol Ther 2019; 104:916-932. [PMID: 30137645 PMCID: PMC6220754 DOI: 10.1002/cpt.1222] [Citation(s) in RCA: 67] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2018] [Accepted: 08/06/2018] [Indexed: 12/15/2022]
Abstract
Bile salt export pump (BSEP) inhibition has emerged as an important mechanism that may contribute to the initiation of human drug‐induced liver injury (DILI). Proactive evaluation and understanding of BSEP inhibition is recommended in drug discovery and development to aid internal decision making on DILI risk. BSEP inhibition can be quantified using in vitro assays. When interpreting assay data, it is important to consider in vivo drug exposure. Currently, this can be undertaken most effectively by consideration of total plasma steady state drug concentrations (Css,plasma). However, because total drug concentrations are not predictive of pharmacological effect, the relationship between total exposure and BSEP inhibition is not causal. Various follow‐up studies can aid interpretation of in vitro BSEP inhibition data and may be undertaken on a case‐by‐case basis. BSEP inhibition is one of several mechanisms by which drugs may cause DILI, therefore, it should be considered alongside other mechanisms when evaluating possible DILI risk.
Collapse
Affiliation(s)
| | - Kunal S Taskar
- Mechanistic Safety and Disposition, IVIVT, GlaxoSmithKline, Ware, Hertfordshire, UK
| | - Christina Battista
- DILIsym Services Inc., a Simulations Plus Company, Research Triangle Park, North Carolina, USA
| | - David L Bourdet
- Drug Metabolism and Pharmacokinetics, Theravance Biopharma, South San Francisco, California, USA
| | - 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
| | | | - David Dai
- Clinical Pharmacology, Research and Development Sciences, Agios Pharmaceuticals, Cambridge, Massachusetts, USA
| | - Christoph Funk
- Roche Pharmaceutical Research and Early Development, Pharmaceutical Sciences, Roche Innovation Center Basel, Basel, Switzerland
| | - Michael J Hafey
- Department of Pharmacokinetics, Pharmacodynamics, and Drug Metabolism, Merck & Co., Inc, Kenilworth, New Jersey, USA
| | - Yurong Lai
- Drug Metabolism, Gilead Sciences Inc., Foster City, California, USA
| | - Jonathan Maher
- Safety Assessment, Genentech, South San Francisco, California, USA
| | - Y Anne Pak
- Lilly Research Laboratory, Lilly Corporate Center, Eli Lilly and Company, Indianapolis, Indiana, USA
| | - Jenny M Pedersen
- Department of Neurobiology, Care Sciences and Society, Karolinska Institute, Novum, Huddinge, Sweden
| | - Joseph W Polli
- Mechanistic Safety and Drug Disposition, GlaxoSmithKline, King of Prussia, Pennsylvania, USA
| | | | - Paul B Watkins
- Institute for Drug Safety Sciences, UNC Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, North Carolina, USA
| | - Kyunghee Yang
- DILIsym Services Inc., a Simulations Plus Company, Research Triangle Park, North Carolina, USA
| | - Robert W Yucha
- Takeda Pharmaceuticals, Global Drug Metabolism and Pharmacokinetics, Cambridge, Massachusetts, USA
| | | |
Collapse
|
37
|
Suga T, Yamaguchi H, Ogura J, Shoji S, Maekawa M, Mano N. Altered bile acid composition and disposition in a mouse model of non-alcoholic steatohepatitis. Toxicol Appl Pharmacol 2019; 379:114664. [PMID: 31306673 DOI: 10.1016/j.taap.2019.114664] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2019] [Revised: 07/10/2019] [Accepted: 07/11/2019] [Indexed: 01/07/2023]
Abstract
Non-alcoholic steatohepatitis (NASH) is a progressive inflammatory and fibrotic disease. However, the progression mechanism of NASH is not well understood. Bile acids are endogenous molecules that regulate cholesterol homeostasis, lipid solubilization in the intestinal lumen, and metabolic signaling via several receptors. In this study, we investigated the relationship between bile acid composition and NASH-associated fibrosis using a mouse model fed choline-deficient, L-amino-acid-defined, high-fat diet with 0.1% methionine (CDAHFD). C57BL/6 J mice fed CDAHFD developed NASH and fibrosis within few weeks. With the progress of NASH-associated liver fibrosis, altered bile acid composition was observed in the liver, bile, and peripheral plasma. Decreased mRNA levels of bile acid metabolizing enzymes such as Cyp7a1 and Baat were observed in contrast to increased Sult2a1 level in the liver. Increased mRNA levels of Ostβ and Abcc4 and decreased in mRNA levels of Bsep, Abcc2, Ntcp, and Oatp1b2, suggesting that bile acids efflux from hepatocytes into the peripheral plasma rather than into bile. In conclusion, the changes in bile acid metabolizing enzymes and transporters expression, resulting in increasing the total bile acid concentration in the plasma, signify a protection mechanism by the hepatocyte to reduce hepatotoxicity during disease progression to NASH but may promote liver fibrosis.
Collapse
Affiliation(s)
- Takahiro Suga
- Graduate School of Pharmaceutical Sciences, Tohoku University, 1-1 Seiryo-machi, Aoba-ku, Sendai 980-8574, Japan
| | - Hiroaki Yamaguchi
- Graduate School of Pharmaceutical Sciences, Tohoku University, 1-1 Seiryo-machi, Aoba-ku, Sendai 980-8574, Japan; Department of Pharmaceutical Sciences, Tohoku University Hospital, 1-1 Seiryo-machi, Aoba-ku, Sendai 980-8574, Japan; Faculty of Pharmaceutical Sciences, Tohoku University, 1-1 Seiryo-machi, Aoba-ku, Sendai 980-8574, Japan.
| | - Jiro Ogura
- Department of Pharmaceutical Sciences, Tohoku University Hospital, 1-1 Seiryo-machi, Aoba-ku, Sendai 980-8574, Japan
| | - Saori Shoji
- Faculty of Pharmaceutical Sciences, Tohoku University, 1-1 Seiryo-machi, Aoba-ku, Sendai 980-8574, Japan
| | - Masamitsu Maekawa
- Department of Pharmaceutical Sciences, Tohoku University Hospital, 1-1 Seiryo-machi, Aoba-ku, Sendai 980-8574, Japan
| | - Nariyasu Mano
- Graduate School of Pharmaceutical Sciences, Tohoku University, 1-1 Seiryo-machi, Aoba-ku, Sendai 980-8574, Japan; Department of Pharmaceutical Sciences, Tohoku University Hospital, 1-1 Seiryo-machi, Aoba-ku, Sendai 980-8574, Japan; Faculty of Pharmaceutical Sciences, Tohoku University, 1-1 Seiryo-machi, Aoba-ku, Sendai 980-8574, Japan.
| |
Collapse
|
38
|
Mezza T, Moffa S, Ferraro PM, Quero G, Capece U, Carfì A, Cefalo CMA, Cinti F, Sorice GP, Impronta F, Mari A, Pontecorvi A, Alfieri S, Holst JJ, Giaccari A. Bile Modulates Secretion of Incretins and Insulin: A Study of Human Extrahepatic Cholestasis. J Clin Endocrinol Metab 2019; 104:2685-2694. [PMID: 30874733 DOI: 10.1210/jc.2018-02804] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/29/2018] [Accepted: 03/11/2019] [Indexed: 02/09/2023]
Abstract
OBJECTIVE Changes in bile flow after bariatric surgery may beneficially modulate secretion of insulin and incretins, leading to diabetes remission. However, the exact mechanism(s) involved is still unclear. Here, we propose an alternative method to investigate the relationship between alterations in physiological bile flow and insulin and incretin secretion by studying changes in gut-pancreatic function in extrahepatic cholestasis in nondiabetic humans. METHODS To pursue this aim, 58 nondiabetic patients with recent diagnosis of periampullary tumors underwent an oral glucose tolerance test (OGTT), and a subgroup of 16 patients also underwent 4-hour mixed meal tests and hyperinsulinemic-euglycemic clamps. RESULTS The analysis of the entire cohort revealed a strong inverse correlation between total bilirubin levels and insulinogenic index. When subjects were divided on the basis of bilirubin levels, used as a marker of altered bile flow, subjects with high bilirubin levels displayed inferior glucose control and decreased insulin secretion during the OGTT. Altered bile flow elicited a markedly greater increase in glucagon and glucagon-like peptide 1 (GLP-1) secretion at fasting state, and following the meal, both glucagon and GLP-1 levels remained increased over time. Conversely, Glucose-dependent insulinotropic polypeptide (GIP) levels were comparable at the fasting state, whereas the increase following meal ingestion was significantly blunted with high bilirubin levels. We reveal strong correlations between total bilirubin and glucagon and GLP-1 levels. CONCLUSIONS Our findings suggest that acute extrahepatic cholestasis determines major impairment in enteroendocrine gut-pancreatic secretory function. The altered bile flow may determine a direct deleterious effect on β-cell function, perhaps mediated by the impairment of incretin hormone function.
Collapse
Affiliation(s)
- Teresa Mezza
- U.O.C., Endocrinologia e Diabetologia, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
- Istituto Patologia Speciale Medica, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Simona Moffa
- U.O.C., Endocrinologia e Diabetologia, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
- Istituto Patologia Speciale Medica, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Pietro Manuel Ferraro
- U.O.C., Nefrologia, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
- Istituto di Medicina Interna e Geriatria, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Giuseppe Quero
- U.O.C., Chirurgia Digestiva, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
- U.O.C., Istituto di Semeiotica Chirurgica, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Umberto Capece
- U.O.C., Endocrinologia e Diabetologia, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
- Istituto Patologia Speciale Medica, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Andrea Carfì
- U.O.C., Endocrinologia e Diabetologia, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
- Istituto Patologia Speciale Medica, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Chiara M A Cefalo
- U.O.C., Endocrinologia e Diabetologia, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
- Istituto Patologia Speciale Medica, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Francesca Cinti
- U.O.C., Endocrinologia e Diabetologia, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
- Istituto Patologia Speciale Medica, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Gian Pio Sorice
- U.O.C., Endocrinologia e Diabetologia, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
- Istituto Patologia Speciale Medica, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Flavia Impronta
- U.O.C., Endocrinologia e Diabetologia, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
- Istituto Patologia Speciale Medica, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Andrea Mari
- U.O.C., Institute of Neuroscience, National Research Council, Padua, Italy
| | - Alfredo Pontecorvi
- U.O.C., Endocrinologia e Diabetologia, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
- Istituto Patologia Speciale Medica, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Sergio Alfieri
- U.O.C., Chirurgia Digestiva, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
- U.O.C., Istituto di Semeiotica Chirurgica, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Jens J Holst
- NNF Center for Basic Metabolic Research and Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Andrea Giaccari
- U.O.C., Endocrinologia e Diabetologia, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
- Istituto Patologia Speciale Medica, Università Cattolica del Sacro Cuore, Rome, Italy
| |
Collapse
|
39
|
Alsaleh M, Barbera TA, Reeves HL, Cramp ME, Ryder S, Gabra H, Nash K, Shen YL, Holmes E, Williams R, Taylor-Robinson SD. Characterization of the urinary metabolic profile of cholangiocarcinoma in a United Kingdom population. Hepat Med 2019; 11:47-67. [PMID: 31118840 PMCID: PMC6507078 DOI: 10.2147/hmer.s193996] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/09/2018] [Accepted: 03/06/2019] [Indexed: 01/09/2023] Open
Abstract
Background: Outside South-East Asia, most cases of cholangiocarcinoma (CCA) have an obscure etiology. There is often diagnostic uncertainty. Metabolomics using ultraperformance liquid chromatography mass spectrometry (UPLC-MS) offers the portent to distinguish disease-specific metabolic signatures. We aimed to define such a urinary metabolic signature in a patient cohort with sporadic CCA and investigate whether there were characteristic differences from those in patients with hepatocellular carcinoma (HCC), metastatic secondary liver cancer, pancreatic cancer and ovarian cancer (OCA). Methods: Spot urine specimens were obtained from 211 subjects in seven participating centers across the UK. Samples were collected from healthy controls and from patients with benign hepatic disease (gallstone, biliary strictures, sphincter of Oddi dysfunction and viral hepatitis) and patients with malignant conditions (HCC, pancreatic cancer, OCA and metastatic cancer in the liver). The spectral metabolite profiles were generated using a UPLC-MS detector and data were analyzed using multivariate and univariate statistical analyses. Results: The greatest class differences were seen between the metabolic profiles of disease-free controls compared to individuals with CCA with altered acylcarnitine, bile acid and purine levels. Individuals with benign strictures showed comparable urine profiles to patients with malignant bile duct lesions. The metabolic signatures of patients with bile duct tumors were distinguishable from patients with hepatocellular and ovarian tumors, but no difference was observed between CCA cases and patients with pancreatic cancer or hepatic secondary metastases. Conclusion: CCA causes subtle but detectable changes in the urine metabolic profiles. The findings point toward potential applications of metabonomics in early tumor detection. However, it is key to utilize both global and targeted metabonomics in a larger cohort for in-depth characterization of the urine metabolome in hepato-pancreato-biliary disease.
Collapse
Affiliation(s)
- Munirah Alsaleh
- Division of Surgery and Cancer, Imperial College London, London
| | | | - Helen L Reeves
- Northern Institute for Cancer Research, Medical School, University of Newcastle, Newcastle upon Tyne, UK
| | | | - Stephen Ryder
- Nottingham Digestive Diseases Centre, University of Nottingham, Nottingham, UK.,NIHR Biomedical Research Unit, Nottingham University Hospitals NHS Trust, Queen's Medical Centre, Nottingham, UK
| | - Hani Gabra
- Division of Surgery and Cancer, Imperial College London, London.,Early Clinical Development, IMED Biotech Unit, AstraZeneca, Cambridge, UK
| | - Kathryn Nash
- Liver Unit, Southampton General Hospital, Southampton, Hampshire, UK
| | - Yi-Liang Shen
- Division of Surgery and Cancer, Imperial College London, London.,Department of Radiation Oncology, Chang Gung Memorial Hospital and Chang Gung University, Taoyuan, Taiwan
| | - Elaine Holmes
- Division of Surgery and Cancer, Imperial College London, London
| | | | | |
Collapse
|
40
|
Wu M, Zhai S, Gao J, Wei D, Xue J, Zhou Y, Li N, Hu L. Diagnosis of hepatocellular carcinoma using a novel anti-glycocholic acid monoclonal antibody-based method. Oncol Lett 2019; 17:3103-3112. [PMID: 30867740 PMCID: PMC6396208 DOI: 10.3892/ol.2019.9943] [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: 03/28/2018] [Accepted: 12/05/2018] [Indexed: 02/05/2023] Open
Abstract
Glycocholic acid (GCA) is a novel identified biomarker for hepatocellular carcinoma (HCC). However, clinical pathological study of GCA has not been extensive due to the limited availability of anti-GCA monoclonal antibodies (mAbs) and restricted detection methods. In the present study, using human GCA conjugated with bovine serum albumin as the immunogen to immunize BALB/c mice, a novel anti-GCA mAb was generated and characterized. The isotypes of heavy chain and light chain of anti-GCA mAb were examined to be IgG2a and κ, respectively, with a high affinity constant (2.6×108 mol/l). The anti-GCA mAb binds GCA with high specificity and sensitivity, and the 50% inhibitory rate was 77.09 ng/ml. The present study also established a rapid, sensitive and efficient indirect competitive ELISA analysis using this anti-GCA mAb to detect the level of GCA produced by different HCC cell lines. Therefore, the present study may successfully develop a novel method for early HCC diagnosis, and also provide insights for further research and treatment of HCC.
Collapse
Affiliation(s)
- Miao Wu
- Department of Immunology, West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Songhui Zhai
- Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Julia Gao
- Department of Immunology, West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Dapeng Wei
- Department of Immunology, West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Jianxin Xue
- Department of Thoracic Oncology, Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Yuxi Zhou
- Department of Pharmacy, Mianyang People's Hospital, Mianyang, Sichuan 621000, P.R. China
| | - Nan Li
- Department of Immunology, West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Lijuan Hu
- Department of Immunology, West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| |
Collapse
|
41
|
Abstract
Cholestasis can be induced by obstruction of bile ducts or intrahepatic toxicity of drugs and chemicals. However, the mode of cell death during cholestasis, i.e., apoptosis or necrosis, has been controversial. There are fundamental reasons for the controversies, both of which are discussed here, namely the design of experiments and the use of parameters with limited specificity for a certain mode of cell death. Based on the assumption that cholestatic liver injury is caused by accumulation of bile acids, rodent (mainly rat) hepatocytes have been exposed to hydrophobic, glycine-conjugated bile acids, which resulted in apoptotic cell death. The problems with this experimental design are that in rodents bile acids are predominantly taurine conjugated and rodent hepatocytes are never exposed to these levels of glycine-conjugated bile acids. In contrast, taurine-conjugated bile acids trigger inflammatory gene activation in rodent hepatocytes and a necro-inflammatory injury in vivo. On the other hand, human hepatocytes are more resistant to glycine-conjugated bile acids and die by necrosis when exposed to high biliary levels of these bile acids. In this chapter, we describe multiple assays including the caspase activity assay, which is specific for apoptosis, and the general cell death assays alanine aminotransferase or lactate dehydrogenase activities in cell culture medium or plasma. An increase in these enzyme activities without caspase activity indicates necrotic cell death. Thus, both the experimental design and the selection of cell death parameters are critical for the relevance of the experiments for the human pathophysiology.
Collapse
Affiliation(s)
| | - Hartmut Jaeschke
- Department of Pharmacology, Toxicology and Therapeutics, University of Kansas Medical Center, Kansas City, KS, USA.
| |
Collapse
|
42
|
Yinchenhao Decoction Alleviates Liver Fibrosis by Regulating Bile Acid Metabolism and TGF-β/Smad/ERK Signalling Pathway. Sci Rep 2018; 8:15367. [PMID: 30337590 PMCID: PMC6194075 DOI: 10.1038/s41598-018-33669-4] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2018] [Accepted: 07/31/2018] [Indexed: 12/12/2022] Open
Abstract
Yinchenhao decoction (YCHD), comprising Yinchenhao (Artemisiae Scopariae Herba), Zhizi (Gardeniae Fructus) and Dahuang (Radix Rhei et Rhizoma), is widely used for treating various diseases. We aimed to investigate the bile acid metabolic mechanism of YCHD in dimethylnitrosamine (DMN)-induced liver fibrosis model. Rats received DMN (10 mg/kg, intraperitoneally) for four successive weeks for liver fibrosis induction and were treated with YCHD for the last 2 weeks. Histopathological analysis showed that YCHD prevented DMN-induced histopathological changes in liver tissues. Serum liver function in YCHD group improved. Ultraperformance liquid chromatography-mass spectrometry analysis showed that YCHD significantly restored both free and conjugated bile acid levels increased by DMN, to normal levels. RT-qPCR results showed that YCHD treatment upregulated the expression of genes related to bile acid synthesis, reabsorption, and excretion. Western blotting analysis showed that YCHD downregulated α-SMA, TGF-β1, p-Smad3, and p-ERK1/2 expression in chenodeoxycholic acid (CDCA)-activated hepatic stellate cells (HSCs). The viability of CDCA-activated HSCs significantly increased after treatment with YCHD and PD98059 (an ERK inhibitor) compared to YCHD treatment alone. Our findings suggest that YCHD alleviated DMN-induced liver fibrosis by regulating enzymes responsible for bile acid metabolism. Additionally, it inhibits CDCA-induced HSC proliferation and activation via TGF-β1/Smad/ERK signalling pathway.
Collapse
|
43
|
Ashby K, Navarro Almario EE, Tong W, Borlak J, Mehta R, Chen M. Review article: therapeutic bile acids and the risks for hepatotoxicity. Aliment Pharmacol Ther 2018; 47:1623-1638. [PMID: 29701277 DOI: 10.1111/apt.14678] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/15/2018] [Revised: 02/17/2018] [Accepted: 03/31/2018] [Indexed: 12/11/2022]
Abstract
BACKGROUND Bile acids play important roles in cholesterol metabolism and signal through farnesoid X receptor and G protein-coupled receptors. Given their importance in liver biology, bile acid therapy enables therapeutic applications beyond the treatment of cholestatic liver disease. However, predicting hepatotoxicity of bile acids in humans is obscured due to inconsistent extrapolations of animal data to humans. AIM To review the evidence that could explain discordant bile acids hepatotoxicity observed in humans and animals. METHOD Literature search was conducted in PubMed using keywords "bile acid," "transporter," "hepatotoxicity," "clinical study," "animal study," "species difference," "mechanism," "genetic disorder." Relevant articles were selected for review. RESULTS Clinically significant hepatotoxicity was reported in response to certain bile acids, namely chenodeoxycholic acid, which was given a boxed warning for potential hepatotoxicity. The chemical structure, specifically the number and orientation of hydroxyl groups, significantly affects their hydrophobicity, an important factor in bile acid toxicity. Experimental studies show that hydrophobic bile acids can lead to liver injury through various mechanisms, such as death receptor signalling, mitochondrial dysfunction and inflammation. Although animal studies play a central role in investigating bile acid safety, there are considerable differences in bile acid composition, metabolism and hepatobiliary disposition across species. This does not allow appropriate safety inference, especially for predicting hepatotoxicity in humans. Exploring evidences stemming from inborn errors, genetic models of disease and toxicology studies further improves an understanding of bile acid hepatotoxicity. CONCLUSION Species differences should be considered in the development of bile acid related therapeutics. Although the mechanism of bile acid hepatotoxicity is still not fully understood, continued mechanistic studies will deepen our understanding.
Collapse
Affiliation(s)
- K Ashby
- Division of Bioinformatics and Biostatistics, National Center for Toxicological Research, US Food and Drug Administration, Jefferson, AR, USA
| | - E E Navarro Almario
- Office of Computational Science, Center for Drug Evaluation and Research, US Food and Drug Administration, Silver Spring, MD, USA
| | - W Tong
- Division of Bioinformatics and Biostatistics, National Center for Toxicological Research, US Food and Drug Administration, Jefferson, AR, USA
| | - J Borlak
- Hannover Medical School, Center of Pharmacology and Toxicology, Hannover, Germany
| | - R Mehta
- Division of Gastroenterology and Inborn Error Products, Center for Drug Evaluation and Research, US Food and Drug Administration, Silver Spring, MD, USA
| | - M Chen
- Division of Bioinformatics and Biostatistics, National Center for Toxicological Research, US Food and Drug Administration, Jefferson, AR, USA
| |
Collapse
|
44
|
Urinary Elimination of Bile Acid Glucuronides under Severe Cholestatic Situations: Contribution of Hepatic and Renal Glucuronidation Reactions. Can J Gastroenterol Hepatol 2018; 2018:8096314. [PMID: 29850459 PMCID: PMC5925157 DOI: 10.1155/2018/8096314] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/06/2017] [Accepted: 02/15/2018] [Indexed: 12/23/2022] Open
Abstract
Biliary obstruction, a severe cholestatic complication, causes accumulation of toxic bile acids (BAs) in liver cells. Glucuronidation, catalyzed by UDP-glucuronosyltransferase (UGT) enzymes, detoxifies cholestatic BAs. Using liquid chromatography coupled to tandem mass spectrometry, 11 BA glucuronide (-G) species were quantified in prebiliary and postbiliary stenting serum and urine samples from 17 patients with biliary obstruction. Stenting caused glucuronide- and fluid-specific changes in BA-G levels and BA-G/BA metabolic ratios. In vitro glucuronidation assays with human liver and kidney microsomes revealed that even if renal enzymes generally displayed lower KM values, the two tissues shared similar glucuronidation capacities for BAs. By contrast, major differences between the two tissues were observed when four human BA-conjugating UGTs 1A3, 1A4, 2B4, and 2B7 were analyzed for mRNA and protein levels. Notably, the BA-24G producing UGT1A3 enzyme, abundant in the liver, was not detected in kidney microsomes. In conclusion, the circulating and urinary BA-G profiles are hugely impacted under severe cholestasis. The similar BA-glucuronidating abilities of hepatic and renal extracts suggest that both the liver and kidney may contribute to the urine BA-G pool.
Collapse
|
45
|
N-3 Polyunsaturated Fatty Acids Stimulate Bile Acid Detoxification in Human Cell Models. Can J Gastroenterol Hepatol 2018; 2018:6031074. [PMID: 29850457 PMCID: PMC5907406 DOI: 10.1155/2018/6031074] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/08/2017] [Accepted: 02/07/2018] [Indexed: 01/16/2023] Open
Abstract
Cholestasis is characterized by the accumulation of toxic bile acids (BAs) in liver cells. The present study aimed to evaluate the effects of n-3 polyunsaturated fatty acids (n-3 PUFAs), such as docosahexaenoic (DHA) and eicosapentaenoic (EPA) acids, on BA homeostasis and toxicity in human cell models. The effects of EPA and/or DHA on the expression of genes involved in the maintenance of BA homeostasis were analyzed in human hepatoma (HepG2) and colon carcinoma (Caco-2) cells, as well as in primary culture of human intestinal (InEpC) and renal (RPTEC) cells. Extracellular BA species were quantified in culture media using LC-MS/MS. BA-induced toxicity was evaluated using caspase-3 and flow cytometry assays. Gene expression analyses of HepG2 cells reveal that n-3 PUFAs reduce the expression of genes involved in BA synthesis (CYP7A1, CYP27A1) and uptake (NTCP), while activating genes encoding metabolic enzymes (SULT2A1) and excretion transporters (MRP2, MRP3). N-3 PUFAs also generate a less toxic BA pool and prevent the BA-dependent activation of apoptosis in HepG2 cells. Conclusion. The present study reveals that n-3 PUFAs stimulate BA detoxification.
Collapse
|
46
|
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: 113] [Impact Index Per Article: 18.8] [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
|
47
|
Reinicke M, Schröter J, Müller-Klieser D, Helmschrodt C, Ceglarek U. Free oxysterols and bile acids including conjugates - Simultaneous quantification in human plasma and cerebrospinal fluid by liquid chromatography-tandem mass spectrometry. Anal Chim Acta 2018; 1037:245-255. [PMID: 30292299 DOI: 10.1016/j.aca.2018.02.049] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2017] [Revised: 02/14/2018] [Accepted: 02/18/2018] [Indexed: 01/19/2023]
Abstract
A liquid chromatography-electrospray ionization-tandem mass spectrometry (LC-ESI(+)-MS/MS) assay was developed and qualified for analyzing 35 analytes of the cholesterol metabolism, including free cholesterol, 17 free, non-esterified oxysterols and 17 free and conjugated bile acids in plasma and cerebrospinal fluid. As internal standards, 25 commercially available stable deuterium-labeled analogs of the analytes were used. Pre-analytical investigations included stability tests of analyte concentrations affected by different anticoagulation additives: lithium heparin-, citrate-, EDTA-K3-stabilized plasma and serum, and the stability in EDTA whole blood at RT. This LC-ESI(+)-MS/MS method was successfully applied for the analysis of paired serum/cerebrospinal fluid samples of patients with and without blood-brain barrier disturbance, as well as of 100 plasma samples of a LIFE-Adult study sub-cohort. A fast and simple sample preparation including protein precipitation and on-line solid-phase extraction was developed. As little as 55 μL of human plasma/serum or cerebrospinal fluid were needed for the analysis. It was possible to separate isomeric oxysterols and bile acids within 23 min using a C18 core-shell column. The assay is capable of quantifying in a linear range of 0.8-250 ng mL-1 for free hydroxycholesterols, 0.2-10 ng mL-1 for dihydroxycholesterols, 0.2-500 ng mL-1 for bile acids and 16-2000 μg mL-1 for cholesterol with acceptable accuracy and precision. In cerebrospinal fluid one free oxysterols, five free and five conjugated bile acids could be quantified. No significant differences between patients with and without blood-brain barrier disturbance were obtained. In the LIFE-Adult sub-cohort two free oxysterols, four free and seven conjugated bile acids could be quantified in EDTA plasma. Men showed significantly higher concentrations of 26-OHC than women (p = 0.035). Furthermore, in women lower levels of cholic acid, glycocholic acid, glycodeoxycholic acid, chenodeoxycholic acid, glycochenodeoxycholic acid, glycoursodeoxycholic acid, glycolithocholic acid and higher levels of taurocholic acid, taurochenodeoxycholic acid, ursodeoxycholic acid/hyodeoxycholic acid were quantified.
Collapse
Affiliation(s)
- Madlen Reinicke
- Institute of Laboratory Medicine, Clinical Chemistry and Molecular Diagnostics, Leipzig University, Liebigstr. 27, 04103 Leipzig, Germany.
| | - Jenny Schröter
- Institute of Laboratory Medicine, Clinical Chemistry and Molecular Diagnostics, Leipzig University, Liebigstr. 27, 04103 Leipzig, Germany
| | - Daniel Müller-Klieser
- Institute of Laboratory Medicine, Clinical Chemistry and Molecular Diagnostics, Leipzig University, Liebigstr. 27, 04103 Leipzig, Germany
| | - Christin Helmschrodt
- Institute of Laboratory Medicine, Clinical Chemistry and Molecular Diagnostics, Leipzig University, Liebigstr. 27, 04103 Leipzig, Germany
| | - Uta Ceglarek
- Institute of Laboratory Medicine, Clinical Chemistry and Molecular Diagnostics, Leipzig University, Liebigstr. 27, 04103 Leipzig, Germany; LIFE - Leipzig Research Center for Civilization Diseases, Leipzig University, Philipp-Rosenthal-Str. 27, 04103 Leipzig, Germany
| |
Collapse
|
48
|
Reprogramming of pro-inflammatory human macrophages to an anti-inflammatory phenotype by bile acids. Sci Rep 2018; 8:255. [PMID: 29321478 PMCID: PMC5762890 DOI: 10.1038/s41598-017-18305-x] [Citation(s) in RCA: 55] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2017] [Accepted: 12/04/2017] [Indexed: 12/11/2022] Open
Abstract
Cholestasis is caused by autoimmune reactions, drug-induced hepatotoxicity, viral infections of the liver and the obstruction of bile ducts by tumours or gallstones. Cholestatic conditions are associated with impaired innate and adaptive immunity, including alterations of the cellular functions of monocytes, macrophages, NK cells and T-cells. Bile acids act as signalling molecules, affecting lipopolysaccharide (LPS)-induced cytokine expression in primary human macrophages. The present manuscript investigates the impact of bile acids, such as taurolithocholic acid (TLC), on the transcriptome of human macrophages in the presence or absence of LPS. While TLC itself has almost no effect on gene expression under control conditions, this compound modulates the expression of 202 out of 865 transcripts in the presence of LPS. Interestingly, pathway analysis revealed that TLC specifically supressed the expression of genes involved in mediating pro-inflammatory effects, phagocytosis, interactions with pathogens and autophagy as well as the recruitment of immune cells, such as NK cells, neutrophils and T cells. These data indicate a broad influence of bile acids on inflammatory responses and immune functions in macrophages. These findings may contribute to the clinical observation that patients with cholestasis present a lack of response to bacterial or viral infections.
Collapse
|
49
|
Fickert P, Wagner M. Biliary bile acids in hepatobiliary injury - What is the link? J Hepatol 2017; 67:619-631. [PMID: 28712691 DOI: 10.1016/j.jhep.2017.04.026] [Citation(s) in RCA: 131] [Impact Index Per Article: 18.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/04/2016] [Revised: 04/14/2017] [Accepted: 04/28/2017] [Indexed: 02/08/2023]
Abstract
The main trigger for liver injury in acquired cholestatic liver disease remains unclear. However, the accumulation of bile acids (BAs) undoubtedly plays a role. Recent progress in deciphering the pathomechanisms of inborn cholestatic liver diseases, decoding mechanisms of BA-induced cell death, and generating modern BA-derived drugs has improved the understanding of the regulation of BA synthesis and transport. Now is the appropriate time to reassess current knowledge about the specific role of BAs in hepatobiliary injury.
Collapse
Affiliation(s)
- Peter Fickert
- Department of Gastroenterology and Hepatology, Medical University Graz, Austria.
| | - Martin Wagner
- Department of Gastroenterology and Hepatology, Medical University Graz, Austria
| |
Collapse
|
50
|
Abstract
BACKGROUND The liver has a number of functions in innate immunity. These functions predispose the liver to innate immune-mediated liver injury when inflammation goes unchecked. Significant progress has been made in the last 25 years on sterile inflammatory liver injury in a number of models; however, a great deal of controversy and many questions about the nature of sterile inflammation still exist. AIM The goal of this article is to review sterile inflammatory liver injury using both a basic approach to what constitutes the inflammatory injury, and through examination of current models of liver injury and inflammation. This information will be tied to human patient conditions when appropriate. RELEVANCE FOR PATIENTS Inflammation is one of the most critical factors for managing in-patient liver disease in a number of scenarios. More information is needed for both scientists and clinicians to develop rational treatments.
Collapse
Affiliation(s)
- Benjamin L Woolbright
- Department of Pharmacology, Toxicology & Therapeutics, University of Kansas Medical Center, Kansas City, Kansas, USA
| | - Hartmut Jaeschke
- Department of Pharmacology, Toxicology & Therapeutics, University of Kansas Medical Center, Kansas City, Kansas, USA
| |
Collapse
|