1
|
Soma H, Yoshida R, Ishizuka S. Quantitative analysis of sterol balance in a mouse model of hepatic lipid accumulation induced by cholesterol and cholic acid supplementation. Biosci Biotechnol Biochem 2025; 89:438-445. [PMID: 39656874 DOI: 10.1093/bbb/zbae183] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2024] [Accepted: 11/27/2024] [Indexed: 12/17/2024]
Abstract
The cholesterol balance and bile acid metabolism in a mouse model of hepatic lipid accumulation induced by a diet supplemented with cholesterol and cholic acid (CA) were quantitatively evaluated. The mice were fed diets supplemented with different levels of cholesterol (0, 3, or 6 g/kg of diet) and CA (0.5 g/kg of diet) for 6 weeks. Cholesterol supplementation doubled the hepatic triglyceride concentration, regardless of the supplementation level, without inflammation or gallstone formation. Both cholesterol supplementations enhanced fecal excretion of muricholic acid. Additionally, the higher cholesterol supplementation led to an increase in fecal cholesterol excretion, accompanied by elevated expression of hepatic cholesterol exporters and a reduction in fecal bile acid excretion. In this mouse study, supplementation with 3 g cholesterol/kg diet and 0.5 g CA/kg diet was sufficient to induce hepatic lipid accumulation.
Collapse
Affiliation(s)
- Hinata Soma
- Research Faculty of Agriculture, Hokkaido University, Sapporo, Japan
| | - Ryo Yoshida
- Research Faculty of Agriculture, Hokkaido University, Sapporo, Japan
| | - Satoshi Ishizuka
- Research Faculty of Agriculture, Hokkaido University, Sapporo, Japan
| |
Collapse
|
2
|
Bouwhuis N, Polak Y, Schimmel AM, Bijleveld YA, Giera MA, Heijink M, Vaz FM, Bootsma AH, Berg-Lammers LAT, Swart NEL, Hollak CEM, Jacobs BAW, Kemper EM. Challenges in the identification and quantification of an unknown impurity in chenodeoxycholic acid drug substance. Eur J Pharm Sci 2025; 205:106979. [PMID: 39643128 DOI: 10.1016/j.ejps.2024.106979] [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: 10/02/2024] [Revised: 11/27/2024] [Accepted: 12/02/2024] [Indexed: 12/09/2024]
Abstract
In 2018 the Amsterdam University Medical Centre decided to prepare chenodeoxycholic acid (CDCA) capsules (also known as pharmacy compounding) for patients with the genetic metabolic disease cerebrotendinous xanthomatosis (CTX) when the product with a marketing authorization was commercially unavailable for patients. However, after reanalysis, unknown impurities were identified in the CDCA active pharmaceutical ingredient (API) using thin-layer chromatography from the European Pharmacopoeia (Ph.Eur.) monograph. Therefore, the API did not comply with the Ph.Eur. specifications for related substances and as a result, pharmacy compounding was halted and an investigation was initiated to identify and quantify the unknown impurities. Meanwhile, a second CDCA API was sourced from another manufacturer. However, this API also appeared to contain an unknown impurity. This impurity could be identified as a dimer of CDCA using reversed phase liquid chromatography mass spectrometry. Since the Ph.Eur. at the time did not describe a suitable analytical method for the quantification of this new impurity, a high pressure liquid chromatography with differential refractometer (HPLC-RI) method was developed to quantify the dimer. Subsequently, in 2019, a new draft version of the CDCA Ph.Eur. monograph was published, including the dimer as a new impurity together with a HPLC-RI method for its identification and quantification. The CDCA-dimer is classified as non-toxic and permitted in the CDCA API up to a maximum of 0.5 %. Because the API complied with the updated Ph.Eur. specifications, pharmacy compounding of CDCA capsules could be resumed.
Collapse
Affiliation(s)
- Natalja Bouwhuis
- Department of Pharmacy and Clinical Pharmacology, Amsterdam UMC, Meibergdreef 9 1105 AZ, Amsterdam, The Netherlands; Platform Medicine for Society, Amsterdam UMC, Meibergdreef 9 1105 AZ, Amsterdam, The Netherlands.
| | - Yasmin Polak
- Department of Pharmacy and Clinical Pharmacology, Amsterdam UMC, Meibergdreef 9 1105 AZ, Amsterdam, The Netherlands; Platform Medicine for Society, Amsterdam UMC, Meibergdreef 9 1105 AZ, Amsterdam, The Netherlands
| | - Anneliene M Schimmel
- Department of Pharmacy and Clinical Pharmacology, Amsterdam UMC, Meibergdreef 9 1105 AZ, Amsterdam, The Netherlands
| | - Yuma A Bijleveld
- Department of Pharmacy and Clinical Pharmacology, Amsterdam UMC, Meibergdreef 9 1105 AZ, Amsterdam, The Netherlands
| | - Martin A Giera
- Center for Proteomics and Metabolomics, Leiden University Medical Center, Albinusdreef 2 2333 ZA, Leiden, The Netherlands
| | - Marieke Heijink
- Center for Proteomics and Metabolomics, Leiden University Medical Center, Albinusdreef 2 2333 ZA, Leiden, The Netherlands
| | - Frédéric M Vaz
- Department of Clinical Chemistry and Pediatrics, Laboratory Genetic Metabolic Diseases, Emma Children's Hospital, Amsterdam UMC, Meibergdreef 9 1105 AZ, Amsterdam, The Netherlands; Amsterdam Gastroenterology Endocrinology Metabolism, Inborn errors of metabolism, Meibergdreef 9 1105 AZ, Amsterdam, The Netherlands; Core Facility Metabolomics, Amsterdam UMC, Meibergdreef 9 1105 AZ, Amsterdam, The Netherlands
| | - Albert H Bootsma
- Department of Clinical Chemistry and Pediatrics, Laboratory Genetic Metabolic Diseases, Emma Children's Hospital, Amsterdam UMC, Meibergdreef 9 1105 AZ, Amsterdam, The Netherlands
| | - Laureen A Ten Berg-Lammers
- Department of Pharmacy and Clinical Pharmacology, Amsterdam UMC, Meibergdreef 9 1105 AZ, Amsterdam, The Netherlands
| | - Noortje E L Swart
- Department of Pharmacy and Clinical Pharmacology, Amsterdam UMC, Meibergdreef 9 1105 AZ, Amsterdam, The Netherlands
| | - Carla E M Hollak
- Platform Medicine for Society, Amsterdam UMC, Meibergdreef 9 1105 AZ, Amsterdam, The Netherlands; Department of Endocrinology and Metabolism, Amsterdam UMC, Meibergdreef 9 1105 AZ, Amsterdam, The Netherlands
| | - Bart A W Jacobs
- Department of Pharmacy and Clinical Pharmacology, Amsterdam UMC, Meibergdreef 9 1105 AZ, Amsterdam, The Netherlands; Platform Medicine for Society, Amsterdam UMC, Meibergdreef 9 1105 AZ, Amsterdam, The Netherlands
| | - E Marleen Kemper
- Department of Pharmacy and Clinical Pharmacology, Amsterdam UMC, Meibergdreef 9 1105 AZ, Amsterdam, The Netherlands; Platform Medicine for Society, Amsterdam UMC, Meibergdreef 9 1105 AZ, Amsterdam, The Netherlands
| |
Collapse
|
3
|
Xu Y, Qian Y, Yu Y, Zhan X, Jin P, Hong J, Dong M. SLC10A5 deficiency causes hypercholanemia. Hepatology 2025; 81:408-422. [PMID: 38986003 PMCID: PMC11737122 DOI: 10.1097/hep.0000000000000994] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/13/2024] [Accepted: 05/19/2024] [Indexed: 07/12/2024]
Abstract
BACKGROUND AND AIMS Solute Carrier Family 10 Member 5 (SLC10A5) is a member of SLC10, comprising transporters of bile acids, steroidal hormones, and other substrates, but its function remains unclear. The aim of the current investigation was to clarify its function in the metabolism of bile acid and hypercholanemia. APPROACH AND RESULTS Whole-exome sequencing and Sanger sequencing were used to identify and confirm the variant in the subjects of hypercholanemia. CRISPR/Cas9-mediated genome engineering was used to establish the knockout and point mutation mice. Primary mouse hepatocytes were isolated, and cell lines were cultured. SLC10A5 was silenced by siRNA and overexpressed by wild-type and mutant plasmids. The fluorescent bile acid derivative was used for the bile acid uptake assay. Bile acids were assessed with ultra-performance liquid chromatography tandem mass spectrometry. A heterozygous variant SLC10A5 : c.994_995del (p.D332X) was identified in subjects with elevated total bile acid or altered bile acid profiles. Bile acids were increased in the serum and liver of knockout and point mutation mice. The expressions of FXR and SHP, regulators involved in the negative feedback of bile acid synthesis, were downregulated, while the bile acid synthesis genes CYP7A1 and CYP8B1 were upregulated in both gene-edited mice. Both the wild and mutant SLC10A5 proteins were localized on the plasma membrane. Knockdown, knockout, or targeted mutation of SLC10A5 led to the inhibition of bile acid uptake by cell lines and primary mouse hepatocytes. CONCLUSION SLC10A5 is involved in the uptake of bile acid, and its deficiency causes hypercholanemia.
Collapse
Affiliation(s)
- Yuqing Xu
- Women's Hospital, School of Medicine, Zhejiang University, Hangzhou, China
- Key Laboratory of Reproductive Genetics (Zhejiang University), Ministry of Education, Hangzhou, China
| | - Yeqing Qian
- Women's Hospital, School of Medicine, Zhejiang University, Hangzhou, China
- Key Laboratory of Reproductive Genetics (Zhejiang University), Ministry of Education, Hangzhou, China
| | - Ying Yu
- Women's Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Xin Zhan
- Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Pengzhen Jin
- Women's Hospital, School of Medicine, Zhejiang University, Hangzhou, China
- Key Laboratory of Reproductive Genetics (Zhejiang University), Ministry of Education, Hangzhou, China
| | - Jiawei Hong
- Women's Hospital, School of Medicine, Zhejiang University, Hangzhou, China
- Key Laboratory of Reproductive Genetics (Zhejiang University), Ministry of Education, Hangzhou, China
| | - Minyue Dong
- Women's Hospital, School of Medicine, Zhejiang University, Hangzhou, China
- Key Laboratory of Reproductive Genetics (Zhejiang University), Ministry of Education, Hangzhou, China
| |
Collapse
|
4
|
Lucas LN, Mallikarjun J, Cattaneo LE, Gangwar B, Zhang Q, Kerby RL, Stevenson D, Rey FE, Amador-Noguez D. Investigation of Bile Salt Hydrolase Activity in Human Gut Bacteria Reveals Production of Conjugated Secondary Bile Acids. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.16.633392. [PMID: 39868271 PMCID: PMC11760432 DOI: 10.1101/2025.01.16.633392] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 01/28/2025]
Abstract
Through biochemical transformation of host-derived bile acids (BAs), gut bacteria mediate host-microbe crosstalk and sit at the interface of nutrition, the microbiome, and disease. BAs play a crucial role in human health by facilitating the absorption of dietary lipophilic nutrients, interacting with hormone receptors to regulate host physiology, and shaping gut microbiota composition through antimicrobial activity. Bile acid deconjugation by bacterial bile salt hydrolase (BSH) has long been recognized as the first necessary BA modification required before further transformations can occur. Here, we show that BSH activity is common among human gut bacterial isolates spanning seven major phyla. We observed variation in both the extent and the specificity of deconjugation of BAs among the tested taxa. Unexpectedly, we discovered that certain strains were capable of directly dehydrogenating conjugated BAs via hydroxysteroid dehydrogenases (HSD) to produce conjugated secondary BAs. These results challenge the prevailing notion that deconjugation is a prerequisite for further BA modifications and lay a foundation for new hypotheses regarding how bacteria act individually or in concert to diversify the BA pool and influence host physiology.
Collapse
Affiliation(s)
- Lauren N. Lucas
- Department of Bacteriology, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Jillella Mallikarjun
- Department of Bacteriology, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Lea E. Cattaneo
- Doctoral Training Program, University of Wisconsin-Madison, Wisconsin, USA
| | - Bhavana Gangwar
- Department of Bacteriology, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Qijun Zhang
- Department of Bacteriology, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Robert L. Kerby
- Department of Bacteriology, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - David Stevenson
- Department of Bacteriology, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Federico E. Rey
- Department of Bacteriology, University of Wisconsin-Madison, Madison, Wisconsin, USA
- Department of Medical Microbiology and Immunology, University of Wisconsin-Madison, Madison, WI, USA
| | - Daniel Amador-Noguez
- Department of Bacteriology, University of Wisconsin-Madison, Madison, Wisconsin, USA
| |
Collapse
|
5
|
Aydin Ö, Wahlström A, de Jonge PA, Meijnikman AS, Sjöland W, Olsson L, Henricsson M, de Goffau MC, Oonk S, Bruin SC, Acherman YIZ, Marschall HU, Gerdes VEA, Nieuwdorp M, Bäckhed F, Groen AK. An integrated analysis of bile acid metabolism in humans with severe obesity. Hepatology 2025; 81:19-31. [PMID: 39010331 DOI: 10.1097/hep.0000000000000938] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/12/2024] [Accepted: 03/26/2024] [Indexed: 07/17/2024]
Abstract
BACKGROUND AND AIMS Bile acids (BA) are vital regulators of metabolism. BAs are AQ6 secreted in the small intestine, reabsorbed, and transported back to the liver, where they can modulate metabolic functions. There is a paucity of data regarding the portal BA composition in humans. This study aimed to address this knowledge gap by investigating portal BA composition and the relation with peripheral and fecal BA dynamics in conjunction with the gut microbiome. APPROACH AND RESULTS Thirty-three individuals from the BARIA cohort were included. Portal plasma, peripheral plasma, and feces were collected. BA and C4 levels were measured employing mass spectrometry. FGF19 was measured using ELISA. Gut microbiota composition was determined through metagenomics analysis on stool samples. Considerable diversity in the portal BA composition was observed. The majority (n = 26) of individuals had a 9-fold higher portal than peripheral BA concentration. In contrast, 8 individuals showed lower portal BA concentration compared with peripheral and had higher levels of unconjugated and secondary BA in this compartment, suggesting more distal origin. The altered portal BA profile was associated with altered gut microbiota composition. In particular, taxa within Bacteroides were reduced in abundance in the feces of these individuals. CONCLUSIONS Characterization of the portal BA composition in relation to peripheral and fecal BA increased insight into the dynamics of BA metabolism in individuals with obesity. Peripheral BA composition was much more diverse due to microbial metabolism. About 24% of the portal samples was surprisingly low in total BA; the underlying mechanism requires further exploration.
Collapse
Affiliation(s)
- Ömrüm Aydin
- Department of Internal and (Experimental) Vascular Medicine, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, the Netherlands
- Department of Vascular Medicine, ACS Institute, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, the Netherlands
| | - Annika Wahlström
- Wallenberg Laboratory and Department of Molecular and Clinical Medicine, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Patrick A de Jonge
- Department of Internal and (Experimental) Vascular Medicine, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, the Netherlands
- Department of Vascular Medicine, ACS Institute, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, the Netherlands
| | - Abraham S Meijnikman
- Department of Internal and (Experimental) Vascular Medicine, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, the Netherlands
- Department of Vascular Medicine, ACS Institute, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, the Netherlands
| | - Wilhelm Sjöland
- Wallenberg Laboratory and Department of Molecular and Clinical Medicine, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Lisa Olsson
- Wallenberg Laboratory and Department of Molecular and Clinical Medicine, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Marcus Henricsson
- Wallenberg Laboratory and Department of Molecular and Clinical Medicine, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Marcus C de Goffau
- Department of Internal and (Experimental) Vascular Medicine, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, the Netherlands
- Department of Vascular Medicine, ACS Institute, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, the Netherlands
| | - Stijn Oonk
- Department of Scientific Research, Data Science, Spaarne Gasthuis Hospital, Hoofddorp, the Netherlands
| | - Sjoerd C Bruin
- Department of Bariatric Surgery, Spaarne Gasthuis Hospital, Hoofddorp, the Netherlands
| | - Yair I Z Acherman
- Department of Bariatric Surgery, Spaarne Gasthuis Hospital, Hoofddorp, the Netherlands
| | - Hanns-Ulrich Marschall
- Wallenberg Laboratory and Department of Molecular and Clinical Medicine, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Victor E A Gerdes
- Department of Internal and (Experimental) Vascular Medicine, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, the Netherlands
- Department of Vascular Medicine, ACS Institute, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, the Netherlands
- Department of Bariatric Surgery, Spaarne Gasthuis Hospital, Hoofddorp, the Netherlands
- Department of Internal Medicine, Spaarne Gasthuis Hospital, Hoofddorp, the Netherlands
| | - Max Nieuwdorp
- Department of Internal and (Experimental) Vascular Medicine, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, the Netherlands
- Department of Vascular Medicine, ACS Institute, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, the Netherlands
| | - Fredrik Bäckhed
- Wallenberg Laboratory and Department of Molecular and Clinical Medicine, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Department of Clinical Physiology, Region Västra Götaland, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Albert K Groen
- Department of Internal and (Experimental) Vascular Medicine, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, the Netherlands
- Department of Vascular Medicine, ACS Institute, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, the Netherlands
| |
Collapse
|
6
|
Fuchs CD, Simbrunner B, Baumgartner M, Campbell C, Reiberger T, Trauner M. Bile acid metabolism and signalling in liver disease. J Hepatol 2025; 82:134-153. [PMID: 39349254 DOI: 10.1016/j.jhep.2024.09.032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/07/2024] [Revised: 09/12/2024] [Accepted: 09/17/2024] [Indexed: 10/02/2024]
Abstract
Bile acids (BAs) serve as signalling molecules, efficiently regulating their own metabolism and transport, as well as key aspects of lipid and glucose homeostasis. BAs shape the gut microbial flora and conversely are metabolised by microbiota. Disruption of BA transport, metabolism and physiological signalling functions contribute to the pathogenesis and progression of a wide range of liver diseases including cholestatic disorders and MASLD (metabolic dysfunction-associated steatotic liver disease), as well as hepatocellular and cholangiocellular carcinoma. Additionally, impaired BA signalling may also affect the intestine and kidney, thereby contributing to failure of gut integrity and driving the progression and complications of portal hypertension, cholemic nephropathy and the development of extrahepatic malignancies such as colorectal cancer. In this review, we will summarise recent advances in the understanding of BA signalling, metabolism and transport, focusing on transcriptional regulation and novel BA-focused therapeutic strategies for cholestatic and metabolic liver diseases.
Collapse
Affiliation(s)
- Claudia D Fuchs
- Division of Gastroenterology and Hepatology, Department of Internal Medicine III, Medical University of Vienna, Vienna, Austria
| | - Benedikt Simbrunner
- Division of Gastroenterology and Hepatology, Department of Internal Medicine III, Medical University of Vienna, Vienna, Austria
| | - Maximillian Baumgartner
- Division of Gastroenterology and Hepatology, Department of Internal Medicine III, Medical University of Vienna, Vienna, Austria; CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
| | - Clarissa Campbell
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
| | - Thomas Reiberger
- Division of Gastroenterology and Hepatology, Department of Internal Medicine III, Medical University of Vienna, Vienna, Austria
| | - Michael Trauner
- Division of Gastroenterology and Hepatology, Department of Internal Medicine III, Medical University of Vienna, Vienna, Austria.
| |
Collapse
|
7
|
Salam R, Bakker M, Krutáková M, Štefela A, Pávek P, Duintjer Tebbens J, Zitko J. The discovery of a new nonbile acid modulator of Takeda G protein-coupled receptor 5: An integrated computational approach. Arch Pharm (Weinheim) 2025; 358:e2400423. [PMID: 39801251 PMCID: PMC11726147 DOI: 10.1002/ardp.202400423] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2024] [Revised: 12/11/2024] [Accepted: 12/19/2024] [Indexed: 01/16/2025]
Abstract
The Takeda G protein-coupled receptor 5 (TGR5), also known as GPBAR1 (G protein-coupled bile acid receptor), is a membrane-type bile acid receptor that regulates blood glucose levels and energy expenditure. These essential functions make TGR5 a promising target for the treatment of type 2 diabetes and metabolic disorders. Currently, most research on developing TGR5 agonists focuses on modifying the structure of bile acids, which are the endogenous ligands of TGR5. However, TGR5 agonists with nonsteroidal structures have not been widely explored. This study aimed at discovering new TGR5 agonists using bile acid derivatives as a basis for a computational approach. We applied a combination of pharmacophore-based, molecular docking, and molecular dynamic (MD) simulation to identify potential compounds as new TGR5 agonists. Through pharmacophore screening and molecular docking, we identified 41 candidate compounds. From these, five candidates were selected based on criteria including pharmacophore features, a docking score of less than 9.2 kcal/mol, and similarity in essential interaction patterns with a reference ligand. Biological assays of the five hits confirmed that Hit-3 activates TGR5 similarly to the bile acid control. This was supported by MD simulation results, which indicated that a hydrogen bond interaction with Tyr240 is involved in TGR5 activation. Hit-3 (CSC089939231) represents a new nonsteroidal lead that can be further optimized to design potent TGR5 agonists.
Collapse
Affiliation(s)
- Rudy Salam
- Department of Biophysics and Physical Chemistry, Faculty of PharmacyCharles UniversityHradec KrálovéCzech Republic
- Department of Pharmacy, Faculty of MedicineUniversitas BrawijayaMalangIndonesia
| | - Michael Bakker
- Department of Biophysics and Physical Chemistry, Faculty of PharmacyCharles UniversityHradec KrálovéCzech Republic
| | - Mária Krutáková
- Department of Pharmacology and Toxicology, Faculty of PharmacyCharles UniversityHradec KrálovéCzech Republic
| | - Alžbeta Štefela
- Department of Pharmacology and Toxicology, Faculty of PharmacyCharles UniversityHradec KrálovéCzech Republic
| | - Petr Pávek
- Department of Pharmacology and Toxicology, Faculty of PharmacyCharles UniversityHradec KrálovéCzech Republic
| | - Jurjen Duintjer Tebbens
- Department of Biophysics and Physical Chemistry, Faculty of PharmacyCharles UniversityHradec KrálovéCzech Republic
| | - Jan Zitko
- Department of Pharmaceutical Chemistry and Pharmaceutical Analysis, Faculty of PharmacyCharles UniversityHradec KrálovéCzech Republic
| |
Collapse
|
8
|
Mustan F, Ivanova A, Tcholakova S. Taurodeoxycholate Aggregation Explored by Molecular Dynamics: Primary-To-Secondary Micelle Transition and Formation of Mixed Micelles with Fatty Acids. Molecules 2024; 29:5897. [PMID: 39769986 PMCID: PMC11677267 DOI: 10.3390/molecules29245897] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2024] [Revised: 12/10/2024] [Accepted: 12/12/2024] [Indexed: 01/11/2025] Open
Abstract
Micelles formed by bile salts in aqueous solution are important for the solubilization of hydrophobic molecules in the gastrointestinal tract. The molecular level information about the mechanism and driving forces for primary-to-secondary micelle transition is still missing. In the current study, the micelle formation of 50 mM solutions of taurodeoxycholate (TDC) is studied by atomistic molecular dynamics simulations. It is shown that primary micelles with an aggregation number of 8-10 emerge and persist within the first 50 ns. Then, they coalesce to form secondary micelles with an aggregation number of 19 molecules. This transition is governed by hydrophobic interactions, which significantly decrease the solvent-accessible surface area per molecule in the secondary micelles. The addition of monomers of the sodium salt of fatty acids (FAs), as agents aiding hydrophobic drug delivery, to secondary TDC micelles results in the co-existence of mixed FA-TDC and pure FA micelles. The studied saturated FAs, with chain lengths of C14:0 and C18:0, are incorporated into the micelle core, whereas TDC molecules position themselves around the FAs, forming a shell on the micelle surface. In contrast, the tails of the C18:1 unsaturated fatty acid mix homogeneously with TDC molecules throughout the entire micelle volume. The latter creates a very suitable medium for hosting hydrophobic molecules in the micelles containing unsaturated fatty acids.
Collapse
Affiliation(s)
- Fatmegyul Mustan
- Department of Chemical and Pharmaceutical Engineering, Faculty of Chemistry and Pharmacy, University of Sofia, 1164 Sofia, Bulgaria
| | - Anela Ivanova
- Department of Physical Chemistry, Faculty of Chemistry and Pharmacy, University of Sofia, 1 James Bourchier Ave., 1164 Sofia, Bulgaria;
| | - Slavka Tcholakova
- Department of Chemical and Pharmaceutical Engineering, Faculty of Chemistry and Pharmacy, University of Sofia, 1164 Sofia, Bulgaria
| |
Collapse
|
9
|
Li Y, Zhao H, Shen Z, Zheng Y, Jiang Y, Song Y, Cai Y. Enhancing DOX efficacy against NSCLC through UDCA-mediated modulation of the TGF-β/MAPK autophagy pathways. Sci Rep 2024; 14:27169. [PMID: 39511265 PMCID: PMC11544154 DOI: 10.1038/s41598-024-73736-7] [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: 05/29/2024] [Accepted: 09/20/2024] [Indexed: 11/15/2024] Open
Abstract
Lung carcinoma, predominantly manifested as non-small cell lung cancer (NSCLC), significantly contributes to oncological mortality, underscoring an imperative for novel therapeutic paradigms. Amidst this context, the present investigation delineates the synergistic potentiation of doxorubicin (DOX)-a canonical chemotherapeutic-by Ursodeoxycholic acid (UDCA), a compound with a historical pedigree in hepatobiliary medicine, now repositioned within oncological pharmacotherapy due to its dichotomous cellular modulation-affording cytoprotection to non-malignant epithelia whilst eliciting apoptotic cascades in neoplastic counterparts. This study, through a rigorous methodological framework, elucidates UDCA's capacity to inhibit NSCLC cellular proliferation and induce apoptosis, thereby significantly amplifying DOX's chemotherapeutic efficacy. Notably, the co-administration of UDCA and DOX was observed to attenuate DOX-induced autophagy via the modulation of the TGF-β/MAPK signaling axis, a pathway pivotal in mediating cellular survival and autophagic mechanisms. Such findings not only underscore the therapeutic potential of UDCA as a chemosensitizer but also illuminate the molecular underpinnings of its modulatory effects, thereby contributing to the corpus of knowledge necessary to surmount chemoresistance in NSCLC. The implications of this research are twofold: firstly, it offers a compelling evidence base for the clinical reevaluation of UDCA in combinatory chemotherapeutic regimens; secondly, it posits a novel mechanistic insight into the modulation of chemotherapeutic efficacy and resistance. Collectively, these insights advocate for the expedited clinical translation of UDCA-DOX synergy, potentially heralding a paradigm shift in the management of NSCLC, thereby addressing a critical lacuna in contemporary oncological therapy.
Collapse
Affiliation(s)
- Ying Li
- Department of Nursing, The First Affiliated Hospital of Hunan Normal University (Hunan Provincial People's Hospital), Changsha, 410005, China
- Department of Respiratory Medicine, The First Affiliated Hospital of Hunan Normal University (Hunan Provincial People's Hospital), Changsha, 410005, China
| | - Helian Zhao
- Department of Nursing, School of Medicine, Hunan Normal University, Changsha, 410013, China
| | - Zhoumin Shen
- Department of Nursing, The First Affiliated Hospital of Hunan Normal University (Hunan Provincial People's Hospital), Changsha, 410005, China
| | - Yao Zheng
- Department of Respiratory Medicine, The First Affiliated Hospital of Hunan Normal University (Hunan Provincial People's Hospital), Changsha, 410005, China
| | - Yuanyuan Jiang
- Department of Nursing, School of Medicine, Hunan Normal University, Changsha, 410013, China
| | - Ying Song
- Department of Respiratory Medicine, The First Affiliated Hospital of Hunan Normal University (Hunan Provincial People's Hospital), Changsha, 410005, China.
| | - Yimin Cai
- Department of Nursing, The First Affiliated Hospital of Hunan Normal University (Hunan Provincial People's Hospital), Changsha, 410005, China.
| |
Collapse
|
10
|
Lee MH, Nuccio SP, Mohanty I, Hagey LR, Dorrestein PC, Chu H, Raffatellu M. How bile acids and the microbiota interact to shape host immunity. Nat Rev Immunol 2024; 24:798-809. [PMID: 39009868 DOI: 10.1038/s41577-024-01057-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/11/2024] [Indexed: 07/17/2024]
Abstract
Bile acids are increasingly appearing in the spotlight owing to their novel impacts on various host processes. Similarly, there is growing attention on members of the microbiota that are responsible for bile acid modifications. With recent advances in technology enabling the discovery and continued identification of microbially conjugated bile acids, the chemical complexity of the bile acid landscape in the body is increasing at a rapid pace. In this Review, we summarize our current understanding of how bile acids and the gut microbiota interact to modulate immune responses during homeostasis and disease, with a particular focus on the gut.
Collapse
Affiliation(s)
- Michael H Lee
- Division of Host-Microbe Systems and Therapeutics, Department of Paediatrics, University of California San Diego, La Jolla, CA, USA
| | - Sean-Paul Nuccio
- Division of Host-Microbe Systems and Therapeutics, Department of Paediatrics, University of California San Diego, La Jolla, CA, USA
| | - Ipsita Mohanty
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, La Jolla, CA, USA
| | - Lee R Hagey
- Department of Medicine, University of California San Diego, La Jolla, CA, USA
| | - Pieter C Dorrestein
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, La Jolla, CA, USA
- Collaborative Mass Spectrometry Innovation Center, University of California San Diego, La Jolla, CA, USA
- Center for Microbiome Innovation, University of California San Diego, La Jolla, CA, USA
| | - Hiutung Chu
- Center for Microbiome Innovation, University of California San Diego, La Jolla, CA, USA
- Department of Pathology, University of California San Diego, La Jolla, CA, USA
- Chiba University-UC San Diego Center for Mucosal Immunology, Allergy and Vaccines (CU-UCSD cMAV), La Jolla, CA, USA
| | - Manuela Raffatellu
- Division of Host-Microbe Systems and Therapeutics, Department of Paediatrics, University of California San Diego, La Jolla, CA, USA.
- Center for Microbiome Innovation, University of California San Diego, La Jolla, CA, USA.
- Chiba University-UC San Diego Center for Mucosal Immunology, Allergy and Vaccines (CU-UCSD cMAV), La Jolla, CA, USA.
| |
Collapse
|
11
|
Pérez-Cova M, Bedia C, Checa A, Meister I, Tauler R, Wheelock CE, Jaumot J. Metabolomic and sphingolipidomic profiling of human hepatoma cells exposed to widely used pharmaceuticals. J Pharm Biomed Anal 2024; 249:116378. [PMID: 39074424 DOI: 10.1016/j.jpba.2024.116378] [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: 04/11/2024] [Revised: 06/08/2024] [Accepted: 07/21/2024] [Indexed: 07/31/2024]
Abstract
Pharmaceutical compounds have become one of the main contaminants of emerging concern (CECs) due to their high usage and increased release into the environment. This study aims to assess the effects caused by three widely consumed hepatotoxic pharmaceutical compounds: an antibiotic (amoxicillin), an antiepileptic (carbamazepine), and an antidepressant (trazodone), on human health when indirectly exposed to toxicologically relevant concentrations (30, 15, and 7.5 μM for amoxicillin and carbamazepine, and 4, 2, and 1 μM for trazodone). A combination of semi-targeted metabolomic and targeted sphingolipid analyses was chosen to unravel the metabolic alterations in human hepatic cells exposed to these CECs at three concentrations for 24 h. HepG2 hepatoma cells were encapsulated in sodium alginate spheroids to improve the physiological relevance of this in vitro approach. Statistical analysis was used to identify the most affected metabolites and sphingolipids for each drug exposure. The results revealed small but significant changes in response to carbamazepine and trazodone exposures, affecting sphingolipid, glycerophospholipid precursors, and amino acid metabolism. Under both drug treatments, a decrease in various ceramide species (related to cell signaling) was observed, along with reduced taurine levels (related to the biosynthesis of bile acid conjugates) and carnitine levels (suggesting an impact on energy production). These and other drug-specific changes indicate that cellular functions in liver cells might be altered under low doses of these CECs, potentially affecting the health of other organs.
Collapse
Affiliation(s)
- Miriam Pérez-Cova
- Department of Environmental Chemistry, IDAEA-CSIC, Jordi Girona 18-26, Barcelona E08034, Spain; Department of Chemical Engineering and Analytical Chemistry, University of Barcelona, Diagonal 647, Barcelona, Barcelona E08028, Spain
| | - Carmen Bedia
- Department of Environmental Chemistry, IDAEA-CSIC, Jordi Girona 18-26, Barcelona E08034, Spain
| | - Antonio Checa
- Unit of Integrative Metabolomics, Institute of Environmental Medicine, Karolinska Institute, Stockholm, Sweden
| | - Isabel Meister
- Unit of Integrative Metabolomics, Institute of Environmental Medicine, Karolinska Institute, Stockholm, Sweden; Gunma University Initiative for Advanced Research (GIAR), Gunma University, 3-39-22 Showa-machi, Maebashi, Gunma 371-8511, Japan
| | - Romà Tauler
- Department of Environmental Chemistry, IDAEA-CSIC, Jordi Girona 18-26, Barcelona E08034, Spain
| | - Craig E Wheelock
- Unit of Integrative Metabolomics, Institute of Environmental Medicine, Karolinska Institute, Stockholm, Sweden; Gunma University Initiative for Advanced Research (GIAR), Gunma University, 3-39-22 Showa-machi, Maebashi, Gunma 371-8511, Japan; Department of Respiratory Medicine and Allergy, Karolinska University Hospital, Stockholm 141-86, Sweden
| | - Joaquim Jaumot
- Department of Environmental Chemistry, IDAEA-CSIC, Jordi Girona 18-26, Barcelona E08034, Spain.
| |
Collapse
|
12
|
Chen Y, Zhang M, Liu Z, Zhang N, Wang Q. Ursodeoxycholic Acid Platinum(IV) Conjugates as Antiproliferative and Antimetastatic Agents: Remodel the Tumor Microenvironment through Suppressing JAK2/STAT3 Signaling. J Med Chem 2024; 67:17551-17567. [PMID: 39292635 DOI: 10.1021/acs.jmedchem.4c01549] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/20/2024]
Abstract
Tumor microenvironment (TME) is a pivotal factor driving the tumor metastasis and leading to the failure of tumor therapy. Here, a series of ursodeoxycholic acid platinum(IV) conjugates with potency in remodeling the TME through suppressing JAK2/STAT3 signaling was developed. A candidate was screened out, which displayed potent antiproliferative and antimetastatic performance both in vitro and in vivo. It displayed superior pharmacokinetic properties compared to cisplatin. Serious DNA injury was induced, and then mitochondria-mediated apoptosis was initiated through the Bcl-2/Bax/Caspase3 pathway. The JAK2/STAT3 and TGF-β1 signaling pathways were remarkably inhibited, and pro-death autophagy was subsequently promoted. The inflammatory and hypoxic TME was suppressed by downregulating COX-2, MMP9, and HIF-1α, which resulted in inhibited angiogenesis in tumors by inhibiting the HIF-1α/VEGFA axis. Additionally, the immunosuppressive TME was reversed by blocking the immune checkpoint PD-L1, further improving the density of CD3+ and CD8+ tumor-infiltrating lymphocytes, and promoting macrophage polarization from M2- to M1-type.
Collapse
Affiliation(s)
- Yan Chen
- Institute of Biopharmaceutical Research, Liaocheng University, Liaocheng 252059, P.R. China
| | - Ming Zhang
- Institute of Biopharmaceutical Research, Liaocheng University, Liaocheng 252059, P.R. China
| | - Zhifang Liu
- Institute of Biopharmaceutical Research, Liaocheng University, Liaocheng 252059, P.R. China
| | - Ning Zhang
- Institute of Biopharmaceutical Research, Liaocheng University, Liaocheng 252059, P.R. China
| | - Qingpeng Wang
- Institute of Biopharmaceutical Research, Liaocheng University, Liaocheng 252059, P.R. China
| |
Collapse
|
13
|
Herrmann M, Rodriguez-Blanco G, Balasso M, Sobolewska K, Semeraro MD, Alonso N, Herrmann W. The role of bile acid metabolism in bone and muscle: from analytics to mechanisms. Crit Rev Clin Lab Sci 2024; 61:510-528. [PMID: 38488591 DOI: 10.1080/10408363.2024.2323132] [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: 12/06/2023] [Revised: 02/09/2024] [Accepted: 02/21/2024] [Indexed: 08/25/2024]
Abstract
Osteoporosis and sarcopenia are both common age-related disorders that are associated with increased morbidity and mortality. Bone and muscle are metabolically very active tissues that require large amounts of energy. Bile acids (BAs), a group of liver-derived steroid compounds, are primarily known as emulsifiers that facilitate the resorption of dietary fat and lipids. In addition, they have pleiotropic metabolic functions in lipoprotein and glucose metabolism, inflammation, and intestinal bacterial growth. Through these effects, they are related to metabolic diseases, such as diabetes, hypertriglyceridemia, atherosclerosis, and nonalcoholic steatohepatitis. BAs mediate their metabolic effects through receptor dependent and receptor-independent mechanisms. Emerging evidence suggests that BAs are also involved in bone and muscle metabolism. Under normal circumstances, BAs support bone health by shifting the delicate equilibrium of bone turnover toward bone formation. In contrast, low or excessive amounts of BAs promote bone resorption. In cholestatic liver disease, BAs accumulate in the liver, reach toxic concentrations in the circulation, and thus may contribute to bone loss and muscle wasting. In addition, the measurement of BAs is in rapid evolution with modern mass spectrometry techniques that allow for the detection of a continuously growing number of BAs. This review provides a comprehensive overview of the biochemistry, physiology and measurement of bile acids. Furthermore, it summarizes the existing literature regarding their role in bone and muscle.
Collapse
Affiliation(s)
- Markus Herrmann
- Clinical Institute of Medical and Chemical Laboratory Diagnostics, Medical University of Graz, Graz, Austria
| | - Giovanny Rodriguez-Blanco
- Clinical Institute of Medical and Chemical Laboratory Diagnostics, Medical University of Graz, Graz, Austria
| | - Marco Balasso
- Clinical Institute of Medical and Chemical Laboratory Diagnostics, Medical University of Graz, Graz, Austria
| | - Katarzyna Sobolewska
- Clinical Institute of Medical and Chemical Laboratory Diagnostics, Medical University of Graz, Graz, Austria
| | - Maria Donatella Semeraro
- Clinical Institute of Medical and Chemical Laboratory Diagnostics, Medical University of Graz, Graz, Austria
| | - Nerea Alonso
- Clinical Institute of Medical and Chemical Laboratory Diagnostics, Medical University of Graz, Graz, Austria
| | - Wolfgang Herrmann
- Clinical Institute of Medical and Chemical Laboratory Diagnostics, Medical University of Graz, Graz, Austria
| |
Collapse
|
14
|
Kwon SJ, Kim YS, Tak J, Lee SG, Lee EB, Kim SG. Hepatic Gα13 ablation shifts region-specific colonic inflammatory status by modulating the bile acid synthetic pathway in mice. Sci Rep 2024; 14:19580. [PMID: 39179591 PMCID: PMC11344048 DOI: 10.1038/s41598-024-70254-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2024] [Accepted: 08/14/2024] [Indexed: 08/26/2024] Open
Abstract
Inflammatory bowel disease is defined by inflammation and immune dysregulation. This study investigated the effects of Gα13 liver-specific knockout (LKO) on proximal and distal colons of dextran sodium sulfate (DSS)-induced mice in conjunction with a high-fat diet (HFD). HFD improved body weight gain and disease activity index scores. Gα13LKO exerted no improvement. In the proximal colon, HFD augmented the DSS effect on Il6, which was not observed in Gα13LKO mice. In the distal colon, HFD plus DSS oppositely fortified an increase in Tnfa and Cxcl10 mRNA in Gα13LKO but not WT. Il6 levels remained unchanged. Bioinformatic approaches using Gα13LKO livers displayed bile acid and cholesterol metabolism-related gene sets. Cholic acid and chenodeoxycholic acid levels were increased in the liver of mice treated with DSS, which was reversed by Gα13LKO. Notably, mice treated with DSS showed a reduction in hepatic ABCB11, CYP7B1, CYP7A1, and CYP8B1, which was reversed by Gα13LKO. Overall, feeding HFD augments the effect of DSS on Il6 in the proximal colon of WT, but not Gα13LKO mice, and enhances DSS effect on Tnfa and Cxcl10 in the distal colon of Gα13LKO mice, suggesting site-specific changes in the inflammatory cytokines, potentially resulting from changes in BA synthesis and excretion.
Collapse
Affiliation(s)
- Soon Jae Kwon
- College of Pharmacy and Integrated Research Institute for Drug Development, Dongguk University-Seoul, Goyang-si, Gyeonggi-do, 10326, Republic of Korea
| | - Yun Seok Kim
- Department of Clinical Pharmacology and Therapeutics, Seoul National University College of Medicine, Seoul, 03080, Republic of Korea
| | - Jihoon Tak
- College of Pharmacy and Integrated Research Institute for Drug Development, Dongguk University-Seoul, Goyang-si, Gyeonggi-do, 10326, Republic of Korea
| | - Sang Gil Lee
- Research and Development Institute, A Pharma Inc., Goyang-si, Gyeonggi-do, Republic of Korea
| | - Eun Byul Lee
- Research and Development Institute, A Pharma Inc., Goyang-si, Gyeonggi-do, Republic of Korea
| | - Sang Geon Kim
- College of Pharmacy and Integrated Research Institute for Drug Development, Dongguk University-Seoul, Goyang-si, Gyeonggi-do, 10326, Republic of Korea.
| |
Collapse
|
15
|
Sudo K, Delmas-Eliason A, Soucy S, Barrack KE, Liu J, Balasubramanian A, Shu CJ, James MJ, Hegner CL, Dionne HD, Rodriguez-Palacios A, Krause HM, O'Toole GA, Karpen SJ, Dawson PA, Schultz D, Sundrud MS. Quantifying Forms and Functions of Enterohepatic Bile Acid Pools in Mice. Cell Mol Gastroenterol Hepatol 2024; 18:101392. [PMID: 39179177 PMCID: PMC11490680 DOI: 10.1016/j.jcmgh.2024.101392] [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: 05/01/2024] [Revised: 08/06/2024] [Accepted: 08/08/2024] [Indexed: 08/26/2024]
Abstract
BACKGROUNDS & AIMS Bile acids (BAs) are core gastrointestinal metabolites with dual functions in lipid absorption and cell signaling. BAs circulate between the liver and distal small intestine (i.e., ileum), yet the dynamics through which complex BA pools are absorbed in the ileum and interact with host intestinal cells in vivo remain poorly understood. Because ileal absorption is rate-limiting in determining which BAs in the intestinal lumen gain access to host intestinal cells and receptors, and at what concentrations, we hypothesized that defining the rates and routes of ileal BA absorption in vivo would yield novel insights into the physiological forms and functions of mouse enterohepatic BA pools. METHODS Using ex vivo mass spectrometry, we quantified 88 BA species and metabolites in the intestinal lumen and superior mesenteric vein of individual wild-type mice, and cage-mates lacking the ileal BA transporter, Asbt/Slc10a2. RESULTS Using these data, we calculated that the pool of BAs circulating through ileal tissue (i.e., the ileal BA pool) in fasting C57BL/6J female mice is ∼0.3 μmol/g. Asbt-mediated transport accounted for ∼80% of this pool and amplified size. Passive permeability explained the remaining ∼20% and generated diversity. Compared with wild-type mice, the ileal BA pool in Asbt-deficient mice was ∼5-fold smaller, enriched in secondary BA species and metabolites normally found in the colon, and elicited unique transcriptional responses on addition to exvivo-cultured ileal explants. CONCLUSIONS This study defines quantitative traits of the mouse enterohepatic BA pool and reveals how aberrant BA metabolism can impinge directly on host intestinal physiology.
Collapse
Affiliation(s)
- Koichi Sudo
- Center for Digestive Health, Dartmouth Health, Lebanon, New Hampshire
| | - Amber Delmas-Eliason
- Department of Immunology and Microbiology, Herbert Wertheim UF Scripps Institute for Biomedical Innovation and Technology, Jupiter, Florida
| | - Shannon Soucy
- Department of Biomedical Data Science, Geisel School of Medicine, Hanover, New Hampshire
| | - Kaitlyn E Barrack
- Department of Microbiology and Immunology, Geisel School of Medicine, Hanover, New Hampshire
| | - Jiabao Liu
- Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Toronto, Ontario, Canada
| | - Akshaya Balasubramanian
- Department of Microbiology and Immunology, Geisel School of Medicine, Hanover, New Hampshire
| | | | | | - Courtney L Hegner
- The Skaggs Graduate School of Chemical and Biological Sciences, The Scripps Research Institute, Jupiter, Florida
| | - Henry D Dionne
- Center for Digestive Health, Dartmouth Health, Lebanon, New Hampshire
| | - Alex Rodriguez-Palacios
- Division of Gastroenterology and Liver Disease, Case Western Reserve University School of Medicine, Cleveland, Ohio; Digestive Health Research Institute, Case Western Reserve University School of Medicine, Cleveland, Ohio; Department of Molecular Biology and Microbiology, Case Western Reserve University School of Medicine, Cleveland, Ohio; University Hospitals Research and Education Institute, University Hospitals Cleveland Medical Center, Cleveland, Ohio
| | - Henry M Krause
- Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Toronto, Ontario, Canada; Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada
| | - George A O'Toole
- Department of Microbiology and Immunology, Geisel School of Medicine, Hanover, New Hampshire
| | - 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; Stravitz-Sanyal Liver Institute for Liver Disease and Metabolic Health, Virginia Commonwealth University, Richmond, Virginia
| | - Paul A Dawson
- Division of Pediatric Gastroenterology, Department of Pediatrics, Hepatology, and Nutrition, Children's Healthcare of Atlanta and Emory University School of Medicine, Atlanta, Georgia
| | - Daniel Schultz
- Department of Microbiology and Immunology, Geisel School of Medicine, Hanover, New Hampshire
| | - Mark S Sundrud
- Center for Digestive Health, Dartmouth Health, Lebanon, New Hampshire; Department of Immunology and Microbiology, Herbert Wertheim UF Scripps Institute for Biomedical Innovation and Technology, Jupiter, Florida; Department of Microbiology and Immunology, Geisel School of Medicine, Hanover, New Hampshire; The Skaggs Graduate School of Chemical and Biological Sciences, The Scripps Research Institute, Jupiter, Florida; Department of Medicine, Geisel School of Medicine, Dartmouth College, Hanover, New Hampshire; Dartmouth Cancer Center, Lebanon, New Hampshire.
| |
Collapse
|
16
|
Li X, Lu C, Mao X, Fan J, Yao J, Jiang J, Wu L, Ren J, Shen J. Bibliometric analysis of research on gut microbiota and bile acids: publication trends and research frontiers. Front Microbiol 2024; 15:1433910. [PMID: 39234549 PMCID: PMC11371755 DOI: 10.3389/fmicb.2024.1433910] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2024] [Accepted: 08/07/2024] [Indexed: 09/06/2024] Open
Abstract
The gut microbiota is widely regarded as a "metabolic organ" that could generate myriad metabolites to regulate human metabolism. As the microbiota metabolites, bile acids (BAs) have recently been identified as the critical endocrine molecules that mediate the cross-talk between the host and intestinal microbiota. This study provided a comprehensive insight into the gut microbiota and BA research through bibliometric analysis from 2003 to 2022. The publications on this subject showed a dramatic upward trend. Although the USA and China have produced the most publications, the USA plays a dominant role in this expanding field. Specifically, the University of Copenhagen was the most productive institution. Key research hotspots are the gut-liver axis, short-chain fatty acids (SCFAs), cardiovascular disease (CVD), colorectal cancer (CRC), and the farnesoid x receptor (FXR). The molecular mechanisms and potential applications of the gut microbiota and BAs in cardiometabolic disorders and gastrointestinal cancers have significant potential for further research.
Collapse
Affiliation(s)
- Xin Li
- Department of General Medicine and Geriatrics, Linping Campus, The Second Affiliated Hospital, Zhejiang University, Hangzhou, Zhejiang, China
- Department of General Practice, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Can Lu
- Department of Colorectal Surgery and Oncology, Key Laboratory of Cancer Prevention and Intervention, Ministry of Education, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Department of Medical Oncology, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Cancer Institute, Key Laboratory of Cancer Prevention and Intervention, Ministry of Education, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Cancer Center, Zhejiang University, Hangzhou, Zhejiang, China
| | - Xue Mao
- Department of General Medicine and Geriatrics, Linping Campus, The Second Affiliated Hospital, Zhejiang University, Hangzhou, Zhejiang, China
| | - Jiahong Fan
- Department of General Medicine and Geriatrics, Linping Campus, The Second Affiliated Hospital, Zhejiang University, Hangzhou, Zhejiang, China
| | - Jianting Yao
- Department of Ultrasound in Medicine, The Second Affiliated Hospital of Zhejiang University School of Medicine, Zhejiang University, Hangzhou, China
| | - Jingjie Jiang
- Department of General Medicine and Geriatrics, Linping Campus, The Second Affiliated Hospital, Zhejiang University, Hangzhou, Zhejiang, China
| | - Lele Wu
- Department of General Medicine and Geriatrics, Linping Campus, The Second Affiliated Hospital, Zhejiang University, Hangzhou, Zhejiang, China
| | - Jingjing Ren
- Department of General Practice, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Jun Shen
- Department of General Medicine and Geriatrics, Linping Campus, The Second Affiliated Hospital, Zhejiang University, Hangzhou, Zhejiang, China
| |
Collapse
|
17
|
Rodrigues DSL, Abreu CMR, Sauvage F, Coelho JFJ, Serra AC, Ivanou D, Mendes A. Improved Interfacial Electron Dynamics with Block Poly(4-vinylpyridine)-Poly(styrene) Polymers for Efficient and Long-Lasting Dye-Sensitized Solar Cells. ACS APPLIED POLYMER MATERIALS 2024; 6:8939-8949. [PMID: 39144281 PMCID: PMC11320385 DOI: 10.1021/acsapm.4c01238] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Revised: 07/03/2024] [Accepted: 07/07/2024] [Indexed: 08/16/2024]
Abstract
Dye-sensitized solar cells (DSSCs) have recently entered the market for indoor photovoltaics. Fast electron injection from dye to titania, the lifetime of the excited dye, and the suppression of back electron recombination at the photoanode/electrolyte interface are crucial for a high photocurrent conversion efficiency (PCE). This study presents block copolymers of poly(4-vinylpyridine) and poly(styrene)-P4VP67-b-PSt x (x=23;61) as efficient accelerators of electron injection from dye to titania with extended lifetime excited states and long-lasting back electron recombination suppression. P4VP67-b-PSt23 and P4VP67-b-PSt61 rendered devices with PCEs of 10.0 and 9.8%, respectively, under AM 1.5G light; PCEs of 19.4 and 16.4% under 1000 lx LED light were attained. Copolymers provided a stable PCE with the two most popular I3 -/3I- electrolytes based on ACN and 3-methoxypropionitrile solvents; PCE history was tracked in the dark and under 1000 h of continuous light soaking with passive load according to ISOS-D1 and ISOS-L2 aging protocols, respectively. The impact of the polymer molecular structure on electron recombination, charge injection, dye anchoring, light absorption, photocurrent generation, and PCE and the long-term history of photovoltaic metrics are discussed.
Collapse
Affiliation(s)
- Daniela
F. S. L. Rodrigues
- CEMMPRE,
ARISE, Department of Chemical Engineering, University of Coimbra, Rua Sílvio Lima—Polo II, 3030-790 Coimbra, Portugal
- LEPABE,
Departamento de Engenharia Química, Faculdade de Engenharia, Universidade do Porto, Rua Dr. Roberto Frias, 4200-465 Porto, Portugal
| | - Carlos M. R. Abreu
- CEMMPRE,
ARISE, Department of Chemical Engineering, University of Coimbra, Rua Sílvio Lima—Polo II, 3030-790 Coimbra, Portugal
| | - Frédéric Sauvage
- Laboratoire
de Réactivité et Chimie des Solides, Université
de Picardie Jules Verne (UPJV), CNRS UMR 7314, Hub de l’énergie, 15 rue Baudelocque, 80039 Amiens, France
| | - Jorge F. J. Coelho
- CEMMPRE,
ARISE, Department of Chemical Engineering, University of Coimbra, Rua Sílvio Lima—Polo II, 3030-790 Coimbra, Portugal
- IPN,
Instituto Pedro Nunes, Associação para a Inovação
e Desenvolvimento em Ciência e Tecnologia, Rua Pedro Nunes, 3030-199 Coimbra, Portugal
| | - Arménio C. Serra
- CEMMPRE,
ARISE, Department of Chemical Engineering, University of Coimbra, Rua Sílvio Lima—Polo II, 3030-790 Coimbra, Portugal
| | - Dzmitry Ivanou
- LEPABE,
Departamento de Engenharia Química, Faculdade de Engenharia, Universidade do Porto, Rua Dr. Roberto Frias, 4200-465 Porto, Portugal
- ALiCE—Associate
Laboratory in Chemical Engineering, Faculty of Engineering, University of Porto, Rua Dr. Roberto Frias, Porto 4200-465, Portugal
| | - Adélio Mendes
- LEPABE,
Departamento de Engenharia Química, Faculdade de Engenharia, Universidade do Porto, Rua Dr. Roberto Frias, 4200-465 Porto, Portugal
- ALiCE—Associate
Laboratory in Chemical Engineering, Faculty of Engineering, University of Porto, Rua Dr. Roberto Frias, Porto 4200-465, Portugal
| |
Collapse
|
18
|
Vico-Oton E, Volet C, Jacquemin N, Dong Y, Hapfelmeier S, Meibom KL, Bernier-Latmani R. Strain-dependent induction of primary bile acid 7-dehydroxylation by cholic acid. BMC Microbiol 2024; 24:286. [PMID: 39090543 PMCID: PMC11293179 DOI: 10.1186/s12866-024-03433-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2024] [Accepted: 07/22/2024] [Indexed: 08/04/2024] Open
Abstract
BACKGROUND Bile acids (BAs) are steroid-derived molecules with important roles in digestion, the maintenance of host metabolism, and immunomodulation. Primary BAs are synthesized by the host, while secondary BAs are produced by the gut microbiome through transformation of the former. The regulation of microbial production of secondary BAs is not well understood, particularly the production of 7-dehydroxylated BAs, which are the most potent agonists for host BA receptors. The 7-dehydroxylation of cholic acid (CA) is well established and is linked to the expression of a bile acid-inducible (bai) operon responsible for this process. However, little to no 7-dehydroxylation has been reported for other host-derived BAs (e.g., chenodeoxycholic acid, CDCA or ursodeoxycholic acid, UDCA). RESULTS Here, we demonstrate that the 7-dehydroxylation of CDCA and UDCA by the human isolate Clostridium scindens is induced when CA is present, suggesting that CA-dependent transcriptional regulation is required for substantial 7-dehydroxylation of these primary BAs. This is supported by the finding that UDCA alone does not promote expression of bai genes. CDCA upregulates expression of the bai genes but the expression is greater when CA is present. In contrast, the murine isolate Extibacter muris exhibits a distinct response; CA did not induce significant 7-dehydroxylation of primary BAs, whereas BA 7-dehydroxylation was promoted upon addition of germ-free mouse cecal content in vitro. However, E. muris was found to 7-dehydroxylate in vivo. CONCLUSIONS The distinct expression responses amongst strains indicate that bai genes are regulated differently. CA promoted bai operon gene expression and the 7-dehydroxylating activity in C. scindens strains. Conversely, the in vitro activity of E. muris was promoted only after the addition of cecal content and the isolate did not alter bai gene expression in response to CA. The accessory gene baiJ was only upregulated in the C. scindens ATCC 35704 strain, implying mechanistic differences amongst isolates. Interestingly, the human-derived C. scindens strains were also capable of 7-dehydroxylating murine bile acids (muricholic acids) to a limited extent. This study shows novel 7-dehydroxylation activity in vitro resulting from the presence of CA and suggests distinct bai gene expression across bacterial species.
Collapse
Affiliation(s)
- Eduard Vico-Oton
- Environmental Microbiology Laboratory, School of Architecture, Civil and Environmental Engineering, École Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
| | - Colin Volet
- Environmental Microbiology Laboratory, School of Architecture, Civil and Environmental Engineering, École Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
| | - Nicolas Jacquemin
- Environmental Microbiology Laboratory, School of Architecture, Civil and Environmental Engineering, École Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
| | - Yuan Dong
- Institute for Infectious Diseases, University of Bern, Bern, Switzerland
| | | | - Karin Lederballe Meibom
- Environmental Microbiology Laboratory, School of Architecture, Civil and Environmental Engineering, École Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
| | - Rizlan Bernier-Latmani
- Environmental Microbiology Laboratory, School of Architecture, Civil and Environmental Engineering, École Polytechnique Fédérale de Lausanne, Lausanne, Switzerland.
- EPFL ENAC IIE EML CH A1 375 (Bâtiment CH), Station 6, CH-1015, Lausanne, Switzerland.
| |
Collapse
|
19
|
Mohanty I, Allaband C, Mannochio-Russo H, El Abiead Y, Hagey LR, Knight R, Dorrestein PC. The changing metabolic landscape of bile acids - keys to metabolism and immune regulation. Nat Rev Gastroenterol Hepatol 2024; 21:493-516. [PMID: 38575682 DOI: 10.1038/s41575-024-00914-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 02/14/2024] [Indexed: 04/06/2024]
Abstract
Bile acids regulate nutrient absorption and mitochondrial function, they establish and maintain gut microbial community composition and mediate inflammation, and they serve as signalling molecules that regulate appetite and energy homeostasis. The observation that there are hundreds of bile acids, especially many amidated bile acids, necessitates a revision of many of the classical descriptions of bile acids and bile acid enzyme functions. For example, bile salt hydrolases also have transferase activity. There are now hundreds of known modifications to bile acids and thousands of bile acid-associated genes, especially when including the microbiome, distributed throughout the human body (for example, there are >2,400 bile salt hydrolases alone). The fact that so much of our genetic and small-molecule repertoire, in both amount and diversity, is dedicated to bile acid function highlights the centrality of bile acids as key regulators of metabolism and immune homeostasis, which is, in large part, communicated via the gut microbiome.
Collapse
Affiliation(s)
- Ipsita Mohanty
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, La Jolla, CA, USA
| | - Celeste Allaband
- Department of Pediatrics, University of California San Diego School of Medicine, La Jolla, CA, USA
| | - Helena Mannochio-Russo
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, La Jolla, CA, USA
| | - Yasin El Abiead
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, La Jolla, CA, USA
| | - Lee R Hagey
- Department of Medicine, University of California San Diego, San Diego, CA, USA
| | - Rob Knight
- Department of Pediatrics, University of California San Diego School of Medicine, La Jolla, CA, USA
- Center for Microbiome Innovation, University of California San Diego, La Jolla, CA, USA
- Department of Computer Science and Engineering, University of California San Diego, La Jolla, CA, USA
- Department of Bioengineering, University of California San Diego, La Jolla, CA, USA
| | - Pieter C Dorrestein
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, La Jolla, CA, USA.
- Center for Microbiome Innovation, University of California San Diego, La Jolla, CA, USA.
- Department of Pharmacology, University of California San Diego, La Jolla, CA, USA.
- Collaborative Mass Spectrometry Innovation Center, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, La Jolla, CA, USA.
| |
Collapse
|
20
|
Liu S, Yang S, Blazekovic B, Li L, Zhang J, Wang Y. Bioactivities, Mechanisms, Production, and Potential Application of Bile Acids in Preventing and Treating Infectious Diseases. ENGINEERING 2024; 38:13-26. [DOI: 10.1016/j.eng.2023.11.017] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
21
|
Komaniecka N, Maroszek S, Drozdzik M, Oswald S, Drozdzik M. Transporter Proteins as Therapeutic Drug Targets-With a Focus on SGLT2 Inhibitors. Int J Mol Sci 2024; 25:6926. [PMID: 39000033 PMCID: PMC11241231 DOI: 10.3390/ijms25136926] [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: 04/30/2024] [Revised: 06/03/2024] [Accepted: 06/21/2024] [Indexed: 07/14/2024] Open
Abstract
Membrane transporters interact not only with endogenous substrates but are also engaged in the transport of xenobiotics, including drugs. While the coordinated function of uptake (solute carrier family-SLC and SLCO) and efflux (ATP-binding cassette family-ABC, multidrug and toxic compound extrusion family-MATE) transporter system allows vectorial drug transport, efflux carriers alone achieve barrier functions. The modulation of transport functions was proved to be effective in the treatment strategies of various pathological states. Sodium-glucose cotransporter-2 (SGLT2) inhibitors are the drugs most widely applied in clinical practice, especially in the treatment of diabetes mellitus and heart failure. Sodium taurocholate co-transporting polypeptide (NTCP) serves as virus particles (HBV/HDV) carrier, and inhibition of its function is applied in the treatment of hepatitis B and hepatitis D by myrcludex B. Inherited cholestatic diseases, such as Alagille syndrome (ALGS) and progressive familial intrahepatic cholestasis (PFIC) can be treated by odevixibat and maralixibat, which inhibit activity of apical sodium-dependent bile salt transporter (ASBT). Probenecid can be considered to increase uric acid excretion in the urine mainly via the inhibition of urate transporter 1 (URAT1), and due to pharmacokinetic interactions involving organic anion transporters 1 and 3 (OAT1 and OAT3), it modifies renal excretion of penicillins or ciprofloxacin as well as nephrotoxicity of cidofovir. This review discusses clinically approved drugs that affect membrane/drug transporter function.
Collapse
Affiliation(s)
- Nina Komaniecka
- Department of Experimental and Clinical Pharmacology, Pomeranian Medical University, 70-111 Szczecin, Poland; (N.K.); (S.M.); (M.D.)
| | - Sonia Maroszek
- Department of Experimental and Clinical Pharmacology, Pomeranian Medical University, 70-111 Szczecin, Poland; (N.K.); (S.M.); (M.D.)
| | - Maria Drozdzik
- Department of Experimental and Clinical Pharmacology, Pomeranian Medical University, 70-111 Szczecin, Poland; (N.K.); (S.M.); (M.D.)
| | - Stefan Oswald
- Institute of Pharmacology and Toxicology, Rostock University Medical Center, 18057 Rostock, Germany;
| | - Marek Drozdzik
- Department of Experimental and Clinical Pharmacology, Pomeranian Medical University, 70-111 Szczecin, Poland; (N.K.); (S.M.); (M.D.)
| |
Collapse
|
22
|
Nguyen Y, Rudd Zhong Manis J, Ronczkowski NM, Bui T, Oxenrider A, Jadeja RN, Thounaojam MC. Unveiling the gut-eye axis: how microbial metabolites influence ocular health and disease. Front Med (Lausanne) 2024; 11:1377186. [PMID: 38799150 PMCID: PMC11122920 DOI: 10.3389/fmed.2024.1377186] [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: 01/26/2024] [Accepted: 04/19/2024] [Indexed: 05/29/2024] Open
Abstract
The intricate interplay between the gut microbiota and ocular health has surpassed conventional medical beliefs, fundamentally reshaping our understanding of organ interconnectivity. This review investigates into the intricate relationship between gut microbiota-derived metabolites and their consequential impact on ocular health and disease pathogenesis. By examining the role of specific metabolites, such as short-chain fatty acids (SCFAs) like butyrate and bile acids (BAs), herein we elucidate their significant contributions to ocular pathologies, thought-provoking the traditional belief of organ sterility, particularly in the field of ophthalmology. Highlighting the dynamic nature of the gut microbiota and its profound influence on ocular health, this review underlines the necessity of comprehending the complex workings of the gut-eye axis, an emerging field of science ready for further exploration and scrutiny. While acknowledging the therapeutic promise in manipulating the gut microbiome and its metabolites, the available literature advocates for a targeted, precise approach. Instead of broad interventions, it emphasizes the potential of exploiting specific microbiome-related metabolites as a focused strategy. This targeted approach compared to a precision tool rather than a broad-spectrum solution, aims to explore the therapeutic applications of microbiome-related metabolites in the context of various retinal diseases. By proposing a nuanced strategy targeted at specific microbial metabolites, this review suggests that addressing specific deficiencies or imbalances through microbiome-related metabolites might yield expedited and pronounced outcomes in systemic health, extending to the eye. This focused strategy holds the potential in bypassing the irregularity associated with manipulating microbes themselves, paving a more efficient pathway toward desired outcomes in optimizing gut health and its implications for retinal diseases.
Collapse
Affiliation(s)
- Yvonne Nguyen
- Mercer University School of Medicine, Macon, GA, United States
| | | | | | - Tommy Bui
- Departments of Cellular Biology and Anatomy, Augusta University, Augusta, GA, United States
| | - Allston Oxenrider
- Departments of Cellular Biology and Anatomy, Augusta University, Augusta, GA, United States
| | - Ravirajsinh N. Jadeja
- Biochemistry and Molecular Biology, Medical College of Georgia at Augusta University, Augusta, GA, United States
| | - Menaka C. Thounaojam
- Departments of Cellular Biology and Anatomy, Augusta University, Augusta, GA, United States
| |
Collapse
|
23
|
Ridlon JM, Gaskins HR. Another renaissance for bile acid gastrointestinal microbiology. Nat Rev Gastroenterol Hepatol 2024; 21:348-364. [PMID: 38383804 PMCID: PMC11558780 DOI: 10.1038/s41575-024-00896-2] [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] [Accepted: 01/16/2024] [Indexed: 02/23/2024]
Abstract
The field of bile acid microbiology in the gastrointestinal tract is going through a current rebirth after a peak of activity in the late 1970s and early 1980s. This renewed activity is a result of many factors, including the discovery near the turn of the century that bile acids are potent signalling molecules and technological advances in next-generation sequencing, computation, culturomics, gnotobiology, and metabolomics. We describe the current state of the field with particular emphasis on questions that have remained unanswered for many decades in both bile acid synthesis by the host and metabolism by the gut microbiota. Current knowledge of established enzymatic pathways, including bile salt hydrolase, hydroxysteroid dehydrogenases involved in the oxidation and epimerization of bile acid hydroxy groups, the Hylemon-Bjӧrkhem pathway of bile acid C7-dehydroxylation, and the formation of secondary allo-bile acids, is described. We cover aspects of bile acid conjugation and esterification as well as evidence for bile acid C3-dehydroxylation and C12-dehydroxylation that are less well understood but potentially critical for our understanding of bile acid metabolism in the human gut. The physiological consequences of bile acid metabolism for human health, important caveats and cautionary notes on experimental design and interpretation of data reflecting bile acid metabolism are also explored.
Collapse
Affiliation(s)
- Jason M Ridlon
- Department of Animal Sciences, University of Illinois Urbana-Champaign, Urbana, IL, USA.
- Division of Nutritional Sciences, University of Illinois Urbana-Champaign, Urbana, IL, USA.
- Carl R. Woese Institute for Genomic Biology, University of Illinois Urbana-Champaign, Urbana, IL, USA.
- Cancer Center at Illinois, University of Illinois Urbana-Champaign, Urbana, IL, USA.
- Center for Advanced Study, University of Illinois Urbana-Champaign, Urbana, IL, USA.
- Department of Microbiology & Immunology, Virginia Commonwealth University, Richmond, VA, USA.
| | - H Rex Gaskins
- Department of Animal Sciences, University of Illinois Urbana-Champaign, Urbana, IL, USA.
- Division of Nutritional Sciences, University of Illinois Urbana-Champaign, Urbana, IL, USA.
- Carl R. Woese Institute for Genomic Biology, University of Illinois Urbana-Champaign, Urbana, IL, USA.
- Cancer Center at Illinois, University of Illinois Urbana-Champaign, Urbana, IL, USA.
- Department of Biomedical and Translational Sciences, University of Illinois Urbana-Champaign, Urbana, IL, USA.
- Department of Pathobiology, University of Illinois Urbana-Champaign, Urbana, IL, USA.
| |
Collapse
|
24
|
Zhang L, Liu X, Jin T, Dong J, Li X, Zhang Y, Liu D. Isomers-oriented separation of forty-five plasma bile acids with liquid chromatography-tandem mass spectrometry. J Chromatogr A 2024; 1721:464827. [PMID: 38520985 DOI: 10.1016/j.chroma.2024.464827] [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: 12/20/2023] [Revised: 03/15/2024] [Accepted: 03/19/2024] [Indexed: 03/25/2024]
Abstract
Some bile acids (BAs) were considered as biomarkers or have therapeutical effect on metabolic diseases. However, due to the existence of isomers and limitations in sensitivity, simultaneous quantification of multiple BAs remains a challenge. The aim of this study is to establish an accurate and sensitive method for the determination of multiple BAs with similar polarity. A LC-MS/MS analytical method capable of quantifying forty-five BAs simultaneously using nine stable isotope internal standards was developed and fully validated based on key isomers-oriented separation strategy. The method was further applied to analyze plasma samples to describe the dynamic profile of BAs after high glucose intake. The chromatography and mass spectrum conditions were optimized to enable the accurate quantification of forty-five BAs, while ensuring the lower limit of quantification between 0.05-10 ng/mL. The results of system suitability, linearity, dilution integrity, accuracy and precision demonstrated the good quantitative capacity and robustness of the method. A total of thirty-five BAs were quantified in plasma samples from twelve healthy Chinese individuals. The established method featured superior sensitivity and better separation efficiency compared to previous studies. Meanwhile, BAs exhibited correlations with glucose and insulin, suggesting their potential as biomarkers for metabolic disorders.
Collapse
Affiliation(s)
- Lei Zhang
- Center of Clinical Medical Research, Institute of Medical Innovation and Research, Peking University Third Hospital, Beijing 100191, China; Drug Clinical Trial Center, Peking University Third Hospital, Beijing 100191, China
| | - Xu Liu
- Drug Clinical Trial Center, Peking University Third Hospital, Beijing 100191, China
| | - Tenghui Jin
- Beijing Institute of Petrochemical Technology, Beijing 102617, China
| | - Jing Dong
- Shimadzu China Innovation Center, Beijing 100020, China
| | - Xiaodong Li
- Shimadzu China Innovation Center, Beijing 100020, China
| | - Youyi Zhang
- Department of Cardiology, Institute of Vascular Medicine, Peking University Third Hospital, Beijing 100191, China.
| | - Dongyang Liu
- Center of Clinical Medical Research, Institute of Medical Innovation and Research, Peking University Third Hospital, Beijing 100191, China; Drug Clinical Trial Center, Peking University Third Hospital, Beijing 100191, China.
| |
Collapse
|
25
|
Han D, Wang W, Gong J, Ma Y, Li Y. Microbiota metabolites in bone: Shaping health and Confronting disease. Heliyon 2024; 10:e28435. [PMID: 38560225 PMCID: PMC10979239 DOI: 10.1016/j.heliyon.2024.e28435] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2023] [Revised: 02/16/2024] [Accepted: 03/19/2024] [Indexed: 04/04/2024] Open
Abstract
The intricate interplay between the gut microbiota and bone health has become increasingly recognized as a fundamental determinant of skeletal well-being. Microbiota-derived metabolites play a crucial role in dynamic interaction, specifically in bone homeostasis. In this sense, short-chain fatty acids (SCFAs), including acetate, propionate, and butyrate, indirectly promote bone formation by regulating insulin-like growth factor-1 (IGF-1). Trimethylamine N-oxide (TMAO) has been found to increase the expression of osteoblast genes, such as Runt-related transcription factor 2 (RUNX2) and bone morphogenetic protein-2 (BMP2), thus enhancing osteogenic differentiation and bone quality through BMP/SMADs and Wnt signaling pathways. Remarkably, in the context of bone infections, the role of microbiota metabolites in immune modulation and host defense mechanisms potentially affects susceptibility to infections such as osteomyelitis. Furthermore, ongoing research elucidates the precise mechanisms through which microbiota-derived metabolites influence bone cells, such as osteoblasts and osteoclasts. Understanding the multifaceted influence of microbiota metabolites on bone, from regulating homeostasis to modulating susceptibility to infections, has the potential to revolutionize our approach to bone health and disease management. This review offers a comprehensive exploration of this evolving field, providing a holistic perspective on the impact of microbiota metabolites on bone health and diseases.
Collapse
Affiliation(s)
- Dong Han
- Department of Trauma Orthopedics, Yantaishan Hospital, Yantai 264000, China
| | - Weijiao Wang
- Department of Otolaryngology, Yantaishan Hospital, Yantai 264000, China
| | - Jinpeng Gong
- Department of Trauma Orthopedics, Yantaishan Hospital, Yantai 264000, China
| | - Yupeng Ma
- Department of Trauma Orthopedics, Yantaishan Hospital, Yantai 264000, China
| | - Yu Li
- Department of Trauma Orthopedics, Yantaishan Hospital, Yantai 264000, China
| |
Collapse
|
26
|
Ay Ü, Leníček M, Haider RS, Classen A, van Eijk H, Koelfat KV, van der Kroft G, Neumann UP, Hoffmann C, Bolm C, Olde Damink SW, Schaap FG. Microbially conjugated bile salts found in human bile activate the bile salt receptors TGR5 and FXR. Hepatol Commun 2024; 8:e0383. [PMID: 38517202 PMCID: PMC10962891 DOI: 10.1097/hc9.0000000000000383] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/18/2023] [Accepted: 12/06/2023] [Indexed: 03/23/2024] Open
Abstract
BACKGROUND Bile salts of hepatic and microbial origin mediate interorgan cross talk in the gut-liver axis. Here, we assessed whether the newly discovered class of microbial bile salt conjugates (MBSCs) activate the main host bile salt receptors (Takeda G protein-coupled receptor 5 [TGR5] and farnesoid X receptor [FXR]) and enter the human systemic and enterohepatic circulation. METHODS N-amidates of (chenodeoxy) cholic acid and leucine, tyrosine, and phenylalanine were synthesized. Receptor activation was studied in cell-free and cell-based assays. MBSCs were quantified in mesenteric and portal blood and bile of patients undergoing pancreatic surgery. RESULTS MBSCs were activating ligands of TGR5 as evidenced by recruitment of Gsα protein, activation of a cAMP-driven reporter, and diminution of lipopolysaccharide-induced cytokine release from macrophages. Intestine-enriched and liver-enriched FXR isoforms were both activated by MBSCs, provided that a bile salt importer was present. The affinity of MBSCs for TGR5 and FXR was not superior to host-derived bile salt conjugates. Individual MBSCs were generally not detected (ie, < 2.5 nmol/L) in human mesenteric or portal blood, but Leu-variant and Phe-variant were readily measurable in bile, where MBSCs comprised up to 213 ppm of biliary bile salts. CONCLUSIONS MBSCs activate the cell surface receptor TGR5 and the transcription factor FXR and are substrates for intestinal (apical sodium-dependent bile acid transporter) and hepatic (Na+ taurocholate co-transporting protein) transporters. Their entry into the human circulation is, however, nonsubstantial. Given low systemic levels and a surplus of other equipotent bile salt species, the studied MBSCs are unlikely to have an impact on enterohepatic TGR5/FXR signaling in humans. The origin and function of biliary MBSCs remain to be determined.
Collapse
Affiliation(s)
- Ümran Ay
- Department of General, Visceral and Transplant Surgery, University Hospital Aachen, Aachen, Germany
| | - Martin Leníček
- Institute of Medical Biochemistry and Laboratory Diagnostics, Faculty General Hospital and First Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Raphael S. Haider
- Institute of Molecular Cell Biology, Center for Molecular Biomedicine, Jena University Hospital, Jena, Germany
- Division of Physiology, Pharmacology and Neuroscience, School of Life Sciences, Queen’s Medical Center, University of Nottingham, Nottingham, United Kingdom
- Center of Membrane Protein and Receptors, Universities of Birmingham and Nottingham, Midlands, United Kingdom
| | - Arno Classen
- Institute of Organic Chemistry, RWTH Aachen University, Aachen, Germany
| | - Hans van Eijk
- Department of Surgery, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University, Maastricht, The Netherlands
| | - Kiran V.K. Koelfat
- Department of General, Visceral and Transplant Surgery, University Hospital Aachen, Aachen, Germany
| | - Gregory van der Kroft
- Department of General, Visceral and Transplant Surgery, University Hospital Aachen, Aachen, Germany
| | - Ulf. P. Neumann
- Department of General, Visceral and Transplant Surgery, University Hospital Aachen, Aachen, Germany
- Department of Surgery, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University, Maastricht, The Netherlands
| | - Carsten Hoffmann
- Institute of Molecular Cell Biology, Center for Molecular Biomedicine, Jena University Hospital, Jena, Germany
| | - Carsten Bolm
- Institute of Organic Chemistry, RWTH Aachen University, Aachen, Germany
| | - Steven W.M. Olde Damink
- Department of General, Visceral and Transplant Surgery, University Hospital Aachen, Aachen, Germany
- Department of Surgery, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University, Maastricht, The Netherlands
| | - Frank G. Schaap
- Department of General, Visceral and Transplant Surgery, University Hospital Aachen, Aachen, Germany
- Department of Surgery, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University, Maastricht, The Netherlands
| |
Collapse
|
27
|
Perazza F, Leoni L, Colosimo S, Musio A, Bocedi G, D’Avino M, Agnelli G, Nicastri A, Rossetti C, Sacilotto F, Marchesini G, Petroni ML, Ravaioli F. Metformin and the Liver: Unlocking the Full Therapeutic Potential. Metabolites 2024; 14:186. [PMID: 38668314 PMCID: PMC11052067 DOI: 10.3390/metabo14040186] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2024] [Revised: 03/20/2024] [Accepted: 03/22/2024] [Indexed: 04/28/2024] Open
Abstract
Metformin is a highly effective medication for managing type 2 diabetes mellitus. Recent studies have shown that it has significant therapeutic benefits in various organ systems, particularly the liver. Although the effects of metformin on metabolic dysfunction-associated steatotic liver disease and metabolic dysfunction-associated steatohepatitis are still being debated, it has positive effects on cirrhosis and anti-tumoral properties, which can help prevent the development of hepatocellular carcinoma. Furthermore, it has been proven to improve insulin resistance and dyslipidaemia, commonly associated with liver diseases. While more studies are needed to fully determine the safety and effectiveness of metformin use in liver diseases, the results are highly promising. Indeed, metformin has a terrific potential for extending its full therapeutic properties beyond its traditional use in managing diabetes.
Collapse
Affiliation(s)
- Federica Perazza
- Department of Medical and Surgical Sciences, IRCCS Azienda Ospedaliero, Universitaria di Bologna, 40138 Bologna, Italy; (F.P.); (L.L.); (G.A.); (A.N.); (C.R.); (F.S.); (G.M.); (M.L.P.)
| | - Laura Leoni
- Department of Medical and Surgical Sciences, IRCCS Azienda Ospedaliero, Universitaria di Bologna, 40138 Bologna, Italy; (F.P.); (L.L.); (G.A.); (A.N.); (C.R.); (F.S.); (G.M.); (M.L.P.)
| | - Santo Colosimo
- Doctorate School of Nutrition Science, University of Milan, 20122 Milan, Italy;
| | | | - Giulia Bocedi
- U.O. Diabetologia, Ospedale C. Magati, Scandiano, 42019 Reggio Emilia, Italy;
| | - Michela D’Avino
- S.C. Endocrinologia Arcispedale Santa Maria Nuova, 42123 Reggio Emilia, Italy;
| | - Giulio Agnelli
- Department of Medical and Surgical Sciences, IRCCS Azienda Ospedaliero, Universitaria di Bologna, 40138 Bologna, Italy; (F.P.); (L.L.); (G.A.); (A.N.); (C.R.); (F.S.); (G.M.); (M.L.P.)
| | - Alba Nicastri
- Department of Medical and Surgical Sciences, IRCCS Azienda Ospedaliero, Universitaria di Bologna, 40138 Bologna, Italy; (F.P.); (L.L.); (G.A.); (A.N.); (C.R.); (F.S.); (G.M.); (M.L.P.)
| | - Chiara Rossetti
- Department of Medical and Surgical Sciences, IRCCS Azienda Ospedaliero, Universitaria di Bologna, 40138 Bologna, Italy; (F.P.); (L.L.); (G.A.); (A.N.); (C.R.); (F.S.); (G.M.); (M.L.P.)
| | - Federica Sacilotto
- Department of Medical and Surgical Sciences, IRCCS Azienda Ospedaliero, Universitaria di Bologna, 40138 Bologna, Italy; (F.P.); (L.L.); (G.A.); (A.N.); (C.R.); (F.S.); (G.M.); (M.L.P.)
| | - Giulio Marchesini
- Department of Medical and Surgical Sciences, IRCCS Azienda Ospedaliero, Universitaria di Bologna, 40138 Bologna, Italy; (F.P.); (L.L.); (G.A.); (A.N.); (C.R.); (F.S.); (G.M.); (M.L.P.)
| | - Maria Letizia Petroni
- Department of Medical and Surgical Sciences, IRCCS Azienda Ospedaliero, Universitaria di Bologna, 40138 Bologna, Italy; (F.P.); (L.L.); (G.A.); (A.N.); (C.R.); (F.S.); (G.M.); (M.L.P.)
| | - Federico Ravaioli
- Department of Medical and Surgical Sciences, IRCCS Azienda Ospedaliero, Universitaria di Bologna, 40138 Bologna, Italy; (F.P.); (L.L.); (G.A.); (A.N.); (C.R.); (F.S.); (G.M.); (M.L.P.)
- Division of Hepatobiliary and Immunoallergic Diseases, Department of Internal Medicine, IRCCS Azienda Ospedaliero, Universitaria di Bologna, 40138 Bologna, Italy
| |
Collapse
|
28
|
Zhang Y, Tang N, Zhou H, Zhu Y. The role of microbial metabolites in endocrine tumorigenesis: From the mechanistic insights to potential therapeutic biomarkers. Biomed Pharmacother 2024; 172:116218. [PMID: 38308969 DOI: 10.1016/j.biopha.2024.116218] [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: 09/26/2023] [Revised: 12/28/2023] [Accepted: 01/22/2024] [Indexed: 02/05/2024] Open
Abstract
Microbial metabolites have been indicated to communicate with the host's endocrine system, regulating hormone production, immune-endocrine communications, and interactions along the gut-brain axis, eventually affecting the occurrence of endocrine cancer. Furthermore, microbiota metabolites such as short-chain fatty acids (SCFAs) have been found to affect the tumor microenvironment and boost immunity against tumors. SCFAs, including butyrate and acetate, have been demonstrated to exert anti-proliferative and anti-protective activity on pancreatic cancer cells. The employing of microbial metabolic products in conjunction with radiation and chemotherapy has shown promising outcomes in terms of reducing treatment side effects and boosting effectiveness. Certain metabolites, such as valerate and butyrate, have been made known to improve the efficiency of CAR T-cell treatment, whilst others, such as indole-derived tryptophan metabolites, have been shown to inhibit tumor immunity. This review explores the intricate interplay between microbial metabolites and endocrine tumorigenesis, spanning mechanistic insights to the discovery of potential therapeutic biomarkers.
Collapse
Affiliation(s)
- Yiyi Zhang
- Department of Endocrinology, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan, 610072, China
| | - Nie Tang
- Department of Endocrinology, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan, 610072, China
| | - Hui Zhou
- Department of Endocrinology, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan, 610072, China.
| | - Ying Zhu
- Department of Endocrinology, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan, 610072, China.
| |
Collapse
|
29
|
Rowe JC, Winston JA. Collaborative Metabolism: Gut Microbes Play a Key Role in Canine and Feline Bile Acid Metabolism. Vet Sci 2024; 11:94. [PMID: 38393112 PMCID: PMC10892723 DOI: 10.3390/vetsci11020094] [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: 01/05/2024] [Revised: 01/31/2024] [Accepted: 02/02/2024] [Indexed: 02/25/2024] Open
Abstract
Bile acids, produced by the liver and secreted into the gastrointestinal tract, are dynamic molecules capable of impacting the overall health of dogs and cats in many contexts. Importantly, the gut microbiota metabolizes host primary bile acids into chemically distinct secondary bile acids. This review explores the emergence of new literature connecting microbial-derived bile acid metabolism to canine and feline health and disease. Moreover, this review highlights multi-omic methodologies for translational research as an area for continued growth in veterinary medicine aimed at accelerating microbiome science and medicine as it pertains to bile acid metabolism in dogs and cats.
Collapse
Affiliation(s)
- John C. Rowe
- Department of Veterinary Clinical Sciences, The Ohio State University College of Veterinary Medicine, Columbus, OH 43210, USA;
- Comparative Hepatobiliary Intestinal Research Program (CHIRP), The Ohio State University College of Veterinary Medicine, Columbus, OH 43210, USA
| | - Jenessa A. Winston
- Department of Veterinary Clinical Sciences, The Ohio State University College of Veterinary Medicine, Columbus, OH 43210, USA;
- Comparative Hepatobiliary Intestinal Research Program (CHIRP), The Ohio State University College of Veterinary Medicine, Columbus, OH 43210, USA
| |
Collapse
|
30
|
Kim G, Chen Z, Li J, Luo J, Castro-Martinez F, Wisniewski J, Cui K, Wang Y, Sun J, Ren X, Crawford SE, Becerra SP, Zhu J, Liu T, Wang S, Zhao K, Wu C. Gut-liver axis calibrates intestinal stem cell fitness. Cell 2024; 187:914-930.e20. [PMID: 38280375 PMCID: PMC10923069 DOI: 10.1016/j.cell.2024.01.001] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Revised: 10/25/2023] [Accepted: 01/02/2024] [Indexed: 01/29/2024]
Abstract
The gut and liver are recognized to mutually communicate through the biliary tract, portal vein, and systemic circulation. However, it remains unclear how this gut-liver axis regulates intestinal physiology. Through hepatectomy and transcriptomic and proteomic profiling, we identified pigment epithelium-derived factor (PEDF), a liver-derived soluble Wnt inhibitor, which restrains intestinal stem cell (ISC) hyperproliferation to maintain gut homeostasis by suppressing the Wnt/β-catenin signaling pathway. Furthermore, we found that microbial danger signals resulting from intestinal inflammation can be sensed by the liver, leading to the repression of PEDF production through peroxisome proliferator-activated receptor-α (PPARα). This repression liberates ISC proliferation to accelerate tissue repair in the gut. Additionally, treating mice with fenofibrate, a clinical PPARα agonist used for hypolipidemia, enhances colitis susceptibility due to PEDF activity. Therefore, we have identified a distinct role for PEDF in calibrating ISC expansion for intestinal homeostasis through reciprocal interactions between the gut and liver.
Collapse
Affiliation(s)
- Girak Kim
- Experimental Immunology Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Zuojia Chen
- Experimental Immunology Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Jian Li
- Experimental Immunology Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Jialie Luo
- Experimental Immunology Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Felipe Castro-Martinez
- Experimental Immunology Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Jan Wisniewski
- Experimental Immunology Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Kairong Cui
- Laboratory of Epigenome Biology, Systems Biology Center, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Yan Wang
- Mass Spectrometry Facility, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD 20892, USA
| | - Jialei Sun
- Department of Gastroenterology and Hepatology, Shanghai Institute of Liver Diseases, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Xiaobai Ren
- Department of Ophthalmology, Mary M. and Sash A. Spencer Center for Vision Research, Byers Eye Institute, Stanford University, Stanford, CA 94304, USA
| | - Susan E Crawford
- Department of Surgery, North Shore University Research Institute, University of Chicago Pritzker School of Medicine, Chicago, IL 60637, USA
| | - S Patricia Becerra
- Section of Protein Structure and Function, Laboratory of Retinal Cell and Molecular Biology, National Eye Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Jimin Zhu
- Department of Gastroenterology and Hepatology, Shanghai Institute of Liver Diseases, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Taotao Liu
- Department of Gastroenterology and Hepatology, Shanghai Institute of Liver Diseases, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Sui Wang
- Department of Ophthalmology, Mary M. and Sash A. Spencer Center for Vision Research, Byers Eye Institute, Stanford University, Stanford, CA 94304, USA
| | - Keji Zhao
- Laboratory of Epigenome Biology, Systems Biology Center, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Chuan Wu
- Experimental Immunology Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA.
| |
Collapse
|
31
|
Zi Z, Rao Y. Discoveries of GPR39 as an evolutionarily conserved receptor for bile acids and of its involvement in biliary acute pancreatitis. SCIENCE ADVANCES 2024; 10:eadj0146. [PMID: 38306436 PMCID: PMC10836733 DOI: 10.1126/sciadv.adj0146] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/02/2023] [Accepted: 01/04/2024] [Indexed: 02/04/2024]
Abstract
Acute pancreatitis (AP) is one of the most common gastrointestinal diseases. Bile acids (BAs) were proposed to be a cause of AP nearly 170 years ago, though the underlying mechanisms remain unclear. Here, we report that two G protein-coupled receptors, GPR39 and GHSR, mediated cellular responses to BAs. Our results revealed GPR39 as an evolutionarily conserved receptor for BAs, particularly 3-O-sulfated lithocholic acids. In cultured cell lines, GPR39 is sufficient for BA-induced Ca2+ elevation. In pancreatic acinar cells, GPR39 mediated BA-induced Ca2+ elevation and necrosis. Furthermore, AP induced by BAs was significantly reduced in GPR39 knockout mice. Our findings provide in vitro and in vivo evidence demonstrating that GPR39 is necessary and sufficient to mediate BA signaling, highlighting its involvement in biliary AP pathogenesis, and suggesting it as a promising therapeutic target for biliary AP.
Collapse
Affiliation(s)
- Zhentao Zi
- Chinese Institutes for Medical Research, Beijing (CIMR, Beijing) and the State Key Laboratory of Digestive Health, Beijing Friendship Hospital, Capital Medical University, Beijing, China
- Peking-Tsinghua Center for Life Sciences, PKU-IDG/McGovern Institute for Brain Research, School of Life Sciences, School of Pharmaceutical Sciences, Department of Chemical Biology, College of Chemistry and Molecular Engineering, Peking University, Beijing, China
| | - Yi Rao
- Chinese Institutes for Medical Research, Beijing (CIMR, Beijing) and the State Key Laboratory of Digestive Health, Beijing Friendship Hospital, Capital Medical University, Beijing, China
- Peking-Tsinghua Center for Life Sciences, PKU-IDG/McGovern Institute for Brain Research, School of Life Sciences, School of Pharmaceutical Sciences, Department of Chemical Biology, College of Chemistry and Molecular Engineering, Peking University, Beijing, China
- Changping Laboratory, Chinese Institute of Brain Research Beijing and Research Unit of Medical Neurobiology, Chinese Academy of Medical Sciences, Beijing 102206, China
| |
Collapse
|
32
|
Gentry EC, Collins SL, Panitchpakdi M, Belda-Ferre P, Stewart AK, Carrillo Terrazas M, Lu HH, Zuffa S, Yan T, Avila-Pacheco J, Plichta DR, Aron AT, Wang M, Jarmusch AK, Hao F, Syrkin-Nikolau M, Vlamakis H, Ananthakrishnan AN, Boland BS, Hemperly A, Vande Casteele N, Gonzalez FJ, Clish CB, Xavier RJ, Chu H, Baker ES, Patterson AD, Knight R, Siegel D, Dorrestein PC. Reverse metabolomics for the discovery of chemical structures from humans. Nature 2024; 626:419-426. [PMID: 38052229 PMCID: PMC10849969 DOI: 10.1038/s41586-023-06906-8] [Citation(s) in RCA: 57] [Impact Index Per Article: 57.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2021] [Accepted: 11/28/2023] [Indexed: 12/07/2023]
Abstract
Determining the structure and phenotypic context of molecules detected in untargeted metabolomics experiments remains challenging. Here we present reverse metabolomics as a discovery strategy, whereby tandem mass spectrometry spectra acquired from newly synthesized compounds are searched for in public metabolomics datasets to uncover phenotypic associations. To demonstrate the concept, we broadly synthesized and explored multiple classes of metabolites in humans, including N-acyl amides, fatty acid esters of hydroxy fatty acids, bile acid esters and conjugated bile acids. Using repository-scale analysis1,2, we discovered that some conjugated bile acids are associated with inflammatory bowel disease (IBD). Validation using four distinct human IBD cohorts showed that cholic acids conjugated to Glu, Ile/Leu, Phe, Thr, Trp or Tyr are increased in Crohn's disease. Several of these compounds and related structures affected pathways associated with IBD, such as interferon-γ production in CD4+ T cells3 and agonism of the pregnane X receptor4. Culture of bacteria belonging to the Bifidobacterium, Clostridium and Enterococcus genera produced these bile amidates. Because searching repositories with tandem mass spectrometry spectra has only recently become possible, this reverse metabolomics approach can now be used as a general strategy to discover other molecules from human and animal ecosystems.
Collapse
Affiliation(s)
- Emily C Gentry
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, La Jolla, CA, USA
- Collaborative Mass Spectrometry Innovation Center, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, La Jolla, CA, USA
- Department of Chemistry, Virginia Tech, Blacksburg, VA, USA
| | - Stephanie L Collins
- Department of Biochemistry and Molecular Biology, The Pennsylvania State University, University Park, PA, USA
| | - Morgan Panitchpakdi
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, La Jolla, CA, USA
- Collaborative Mass Spectrometry Innovation Center, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, La Jolla, CA, USA
| | - Pedro Belda-Ferre
- Department of Pediatrics, University of California, San Diego, La Jolla, CA, USA
- Department of Computer Science and Engineering, Jacobs School of Engineering, University of California, San Diego, San Diego, CA, USA
| | - Allison K Stewart
- Department of Chemistry, North Carolina State University, Raleigh, NC, USA
| | | | - Hsueh-Han Lu
- Department of Pathology, University of California, San Diego, La Jolla, CA, USA
| | - Simone Zuffa
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, La Jolla, CA, USA
- Collaborative Mass Spectrometry Innovation Center, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, La Jolla, CA, USA
| | - Tingting Yan
- Laboratory of Metabolism, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | | | | | - Allegra T Aron
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, La Jolla, CA, USA
- Collaborative Mass Spectrometry Innovation Center, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, La Jolla, CA, USA
| | - Mingxun Wang
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, La Jolla, CA, USA
- Collaborative Mass Spectrometry Innovation Center, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, La Jolla, CA, USA
| | - Alan K Jarmusch
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, La Jolla, CA, USA
- Collaborative Mass Spectrometry Innovation Center, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, La Jolla, CA, USA
- Immunity, Inflammation, and Disease Laboratory, Division of Intramural Research, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC, USA
| | - Fuhua Hao
- Center for Molecular Toxicology and Carcinogenesis, Department of Veterinary and Biomedical Sciences, The Pennsylvania State University, University Park, PA, USA
| | - Mashette Syrkin-Nikolau
- Division of Gastroenterology, Department of Pediatrics, Rady Children's Hospital University of California San Diego, La Jolla, CA, USA
| | - Hera Vlamakis
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Center for Microbiome Informatics and Therapeutics, Massachusetts Institute of Technology, Cambridge, MA, USA
| | | | - Brigid S Boland
- Division of Gastroenterology, University of California, San Diego, La Jolla, CA, USA
| | - Amy Hemperly
- Division of Gastroenterology, Department of Pediatrics, Rady Children's Hospital University of California San Diego, La Jolla, CA, USA
| | - Niels Vande Casteele
- Division of Gastroenterology, University of California, San Diego, La Jolla, CA, USA
| | - Frank J Gonzalez
- Laboratory of Metabolism, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Clary B Clish
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Ramnik J Xavier
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Center for Microbiome Informatics and Therapeutics, Massachusetts Institute of Technology, Cambridge, MA, USA
- Center for Computational and Integrative Biology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Department of Molecular Biology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Hiutung Chu
- Department of Pathology, University of California, San Diego, La Jolla, CA, USA
- CU-UCSD, Center for Mucosal Immunology, Allergy and Vaccine Development, University of California, San Diego, La Jolla, California, USA
| | - Erin S Baker
- Department of Chemistry, North Carolina State University, Raleigh, NC, USA
- Department of Chemistry, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Andrew D Patterson
- Center for Molecular Toxicology and Carcinogenesis, Department of Veterinary and Biomedical Sciences, The Pennsylvania State University, University Park, PA, USA
| | - Rob Knight
- Department of Pediatrics, University of California, San Diego, La Jolla, CA, USA
- Department of Computer Science and Engineering, Jacobs School of Engineering, University of California, San Diego, San Diego, CA, USA
- Center for Microbiome Innovation, Jacobs School of Engineering, University of California, San Diego, San Diego, CA, USA
- Department of Bioengineering, University of California, San Diego, San Diego, California, USA
| | - Dionicio Siegel
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, La Jolla, CA, USA
| | - Pieter C Dorrestein
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, La Jolla, CA, USA.
- Collaborative Mass Spectrometry Innovation Center, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, La Jolla, CA, USA.
| |
Collapse
|
33
|
Ichikawa M, Okada H, Nakamoto N, Taniki N, Chu PS, Kanai T. The gut-liver axis in hepatobiliary diseases. Inflamm Regen 2024; 44:2. [PMID: 38191517 PMCID: PMC10773109 DOI: 10.1186/s41232-023-00315-0] [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: 11/01/2023] [Accepted: 12/17/2023] [Indexed: 01/10/2024] Open
Abstract
Recent advances in the analysis of intestinal bacteria have led to reports of variations in intestinal bacterial levels among hepatobiliary diseases. The mechanisms behind the changes in intestinal bacteria in various hepatobiliary diseases include the abnormal composition of intestinal bacteria, weakening of the intestinal barrier, and bacterial translocation outside the intestinal tract, along with their metabolites, but many aspects remain unresolved. Further research employing clinical studies and animal models is expected to clarify the direct relationship between intestinal bacteria and hepatobiliary diseases and to validate the utility of intestinal bacteria as a diagnostic biomarker and potential therapeutic target. This review summarizes the involvement of the microbiota in the pathogenesis of hepatobiliary diseases via the gut-liver axis.
Collapse
Affiliation(s)
- Masataka Ichikawa
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Keio University School of Medicine, Shinanomachi, Tokyo, 1608582, Japan
| | - Haruka Okada
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Keio University School of Medicine, Shinanomachi, Tokyo, 1608582, Japan
| | - Nobuhiro Nakamoto
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Keio University School of Medicine, Shinanomachi, Tokyo, 1608582, Japan.
| | - Nobuhito Taniki
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Keio University School of Medicine, Shinanomachi, Tokyo, 1608582, Japan
| | - Po-Sung Chu
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Keio University School of Medicine, Shinanomachi, Tokyo, 1608582, Japan
| | - Takanori Kanai
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Keio University School of Medicine, Shinanomachi, Tokyo, 1608582, Japan.
| |
Collapse
|
34
|
Wang Y, Xu H, Zhang X, Ma J, Xue S, Shentu D, Mao T, Li S, Yue M, Cui J, Wang L. The Role of Bile Acids in Pancreatic Cancer. Curr Cancer Drug Targets 2024; 24:1005-1014. [PMID: 38284711 DOI: 10.2174/0115680096281168231215060301] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2023] [Revised: 11/02/2023] [Accepted: 11/06/2023] [Indexed: 01/30/2024]
Abstract
Bile acids are well known to promote the digestion and absorption of fat, and at the same time, they play an important role in lipid and glucose metabolism. More studies have found that bile acids such as ursodeoxycholic acid also have anti-inflammatory and immune-regulating effects. Bile acids have been extensively studied in biliary and intestinal tumors but less in pancreatic cancer. Patients with pancreatic cancer, especially pancreatic head cancer, are often accompanied by biliary obstruction and elevated bile acids caused by tumors. Elevated total bile acid levels in pancreatic cancer patients usually have a poor prognosis. There has been controversy over whether elevated bile acids are harmful or beneficial to pancreatic cancer. Still, there is no doubt that bile acids are important for the occurrence and development of pancreatic cancer. This article summarizes the research on bile acid as a biomarker and regulation of the occurrence, development and chemoresistance of pancreatic cancer, hoping to provide some inspiration for future research.
Collapse
Affiliation(s)
- Yanling Wang
- Oncology Department and State Key Laboratory of Systems Medicine for Cancer of Shanghai Cancer Institute, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, 200127, China
| | - Haiyan Xu
- Oncology Department and State Key Laboratory of Systems Medicine for Cancer of Shanghai Cancer Institute, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, 200127, China
| | - Xiaofei Zhang
- Oncology Department and State Key Laboratory of Systems Medicine for Cancer of Shanghai Cancer Institute, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, 200127, China
| | - Jingyu Ma
- Oncology Department and State Key Laboratory of Systems Medicine for Cancer of Shanghai Cancer Institute, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, 200127, China
| | - Shengbai Xue
- Oncology Department and State Key Laboratory of Systems Medicine for Cancer of Shanghai Cancer Institute, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, 200127, China
| | - Daiyuan Shentu
- Oncology Department and State Key Laboratory of Systems Medicine for Cancer of Shanghai Cancer Institute, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, 200127, China
| | - Tiebo Mao
- Oncology Department and State Key Laboratory of Systems Medicine for Cancer of Shanghai Cancer Institute, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, 200127, China
| | - Shumin Li
- Oncology Department and State Key Laboratory of Systems Medicine for Cancer of Shanghai Cancer Institute, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, 200127, China
| | - Ming Yue
- Oncology Department and State Key Laboratory of Systems Medicine for Cancer of Shanghai Cancer Institute, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, 200127, China
| | - Jiujie Cui
- Oncology Department and State Key Laboratory of Systems Medicine for Cancer of Shanghai Cancer Institute, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, 200127, China
| | - Liwei Wang
- Oncology Department and State Key Laboratory of Systems Medicine for Cancer of Shanghai Cancer Institute, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, 200127, China
| |
Collapse
|
35
|
Kubota N, Hori S, Ishizuka S. Differences in iron balance observed with dietary cholic acid supplementation and marginal iron deficiency in rats. Biosci Biotechnol Biochem 2023; 88:79-85. [PMID: 37813822 DOI: 10.1093/bbb/zbad140] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2023] [Accepted: 09/27/2023] [Indexed: 10/11/2023]
Abstract
We investigated whether a cholic acid (CA)-supplemented diet and marginal iron deficiency (MID) diet influence hepatic lipid accumulation and iron balance in rats for 2 weeks. The CA diet enhanced hepatic lipid accumulation and modulated iron metabolism such as enhancement of fecal iron excretion, reduction in iron absorption, and no alteration in plasma iron levels. The MID diet did not alter hepatic lipid concentrations with reduced iron concentration in the liver and plasma. In combination, influence of the CA supplementation on the hepatic iron concentration was opposite between iron-sufficient and MID conditions. In the liver, the CA diet enhanced lipocalin 2 expression, whereas the MID diet enhanced transferrin receptor 1 expression and reduced hepcidin expression. This study revealed an involvement of 12-hydroxylated bile acids in regulation of hepatic iron concentration under MID condition.
Collapse
Affiliation(s)
- Natsuki Kubota
- Research Faculty of Agriculture, Hokkaido University, Sapporo, Japan
| | - Shota Hori
- Research Faculty of Agriculture, Hokkaido University, Sapporo, Japan
| | - Satoshi Ishizuka
- Research Faculty of Agriculture, Hokkaido University, Sapporo, Japan
| |
Collapse
|
36
|
Niu Z, Liu Y, Wang Y, Liu Y, Chai L, Wang H. Impairment of bile acid metabolism and altered composition by lead and copper in Bufo gargarizans tadpoles. THE SCIENCE OF THE TOTAL ENVIRONMENT 2023; 900:165901. [PMID: 37524187 DOI: 10.1016/j.scitotenv.2023.165901] [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: 05/19/2023] [Revised: 07/10/2023] [Accepted: 07/28/2023] [Indexed: 08/02/2023]
Abstract
Lead (Pb) and copper (Cu) are two common heavy metal contaminants in environments, and liver is recognized as one of the main target organs for toxicity of Pb and Cu in animal organisms. Bile acids play a critical role in regulating hepatic metabolic homeostasis by activating farnesoid X receptor (Fxr). However, there were few studies on the interactions between bile acids and liver pathology caused by heavy metals. In this work, the histopathological changes, targeted metabolome and transcriptome responses in the liver of Bufo gargarizans tadpoles to Pb and/or Cu were examined. We found that exposure to Pb and/or Cu altered the hepatic bile acid profile, resulting in increased hydrophobicity and toxicity of the bile acid pool. And the expression of genes involved in bile acid metabolism and their downstream signaling pathways in the liver were significantly altered by Pb and/or Cu exposure. The alteration of bile acid profiles and the expression of genes related to bile acid metabolism might induce oxidative stress and inflammation, ultimately inducing hepatocyte injury observed in the histological sections. To our knowledge, this is the first study to provide histological, biochemical, and molecular evidence for establishing the link between Pb and Cu exposure, disturbances in hepatic bile acid metabolism, and liver injury.
Collapse
Affiliation(s)
- Ziyi Niu
- College of Life Science, Shaanxi Normal University, Xi'an 710119, China
| | - Yutian Liu
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing 210023, China
| | - Yaxi Wang
- College of Life Science, Shaanxi Normal University, Xi'an 710119, China
| | - Ying Liu
- College of Life Science, Shaanxi Normal University, Xi'an 710119, China
| | - Lihong Chai
- School of Water and Environment, Chang'an University, Xi'an 710054, China; Key Laboratory of Subsurface Hydrology and Ecological Effect in Arid Region of Ministry of Education, Chang'an University, Xi'an 710054, China
| | - Hongyuan Wang
- College of Life Science, Shaanxi Normal University, Xi'an 710119, China.
| |
Collapse
|
37
|
Ridlon JM, Daniel SL, Gaskins HR. The Hylemon-Björkhem pathway of bile acid 7-dehydroxylation: history, biochemistry, and microbiology. J Lipid Res 2023; 64:100392. [PMID: 37211250 PMCID: PMC10382948 DOI: 10.1016/j.jlr.2023.100392] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2023] [Revised: 05/07/2023] [Accepted: 05/11/2023] [Indexed: 05/23/2023] Open
Abstract
Bile acids are detergents derived from cholesterol that function to solubilize dietary lipids, remove cholesterol from the body, and act as nutrient signaling molecules in numerous tissues with functions in the liver and gut being the best understood. Studies in the early 20th century established the structures of bile acids, and by mid-century, the application of gnotobiology to bile acids allowed differentiation of host-derived "primary" bile acids from "secondary" bile acids generated by host-associated microbiota. In 1960, radiolabeling studies in rodent models led to determination of the stereochemistry of the bile acid 7-dehydration reaction. A two-step mechanism was proposed, which we have termed the Samuelsson-Bergström model, to explain the formation of deoxycholic acid. Subsequent studies with humans, rodents, and cell extracts of Clostridium scindens VPI 12708 led to the realization that bile acid 7-dehydroxylation is a result of a multi-step, bifurcating pathway that we have named the Hylemon-Björkhem pathway. Due to the importance of hydrophobic secondary bile acids and the increasing measurement of microbial bai genes encoding the enzymes that produce them in stool metagenome studies, it is important to understand their origin.
Collapse
Affiliation(s)
- Jason M Ridlon
- Department of Animal Sciences, University of Illinois Urbana-Champaign, Urbana, IL, USA; Division of Nutritional Sciences, University of Illinois Urbana-Champaign, Urbana, IL, USA; Carl R. Woese Institute for Genomic Biology, University of Illinois Urbana-Champaign, Urbana, IL, USA; Cancer Center at Illinois, University of Illinois Urbana-Champaign, Urbana, IL, USA; Center for Advanced Study, University of Illinois Urbana-Champaign, Urbana, IL, USA; Department of Microbiology and Immunology, Virginia Commonwealth University School of Medicine, Richmond, VA, USA.
| | - Steven L Daniel
- Department of Biological Sciences, Eastern Illinois University, Charleston, IL, USA
| | - H Rex Gaskins
- Department of Animal Sciences, University of Illinois Urbana-Champaign, Urbana, IL, USA; Division of Nutritional Sciences, University of Illinois Urbana-Champaign, Urbana, IL, USA; Carl R. Woese Institute for Genomic Biology, University of Illinois Urbana-Champaign, Urbana, IL, USA; Cancer Center at Illinois, University of Illinois Urbana-Champaign, Urbana, IL, USA; Department of Biomedical and Translational Sciences, University of Illinois Urbana-Champaign, Urbana, IL, USA; Department of Pathobiology, University of Illinois Urbana-Champaign, Urbana, IL, USA
| |
Collapse
|
38
|
Wang MX, Han J, Liu T, Wang RX, Li LT, Li ZD, Yang JC, Liu LL, Lu Y, Xie XB, Gong JY, Li SY, Zhang L, Ling V, Wang JS. Poly-hydroxylated bile acids and their prognostic roles in Alagille syndrome. World J Pediatr 2023; 19:652-662. [PMID: 36658452 DOI: 10.1007/s12519-022-00676-5] [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: 07/21/2022] [Accepted: 12/14/2022] [Indexed: 01/20/2023]
Abstract
BACKGROUND The liver manifestations of Alagille syndrome (ALGS) are highly variable, and factors affecting its prognosis are poorly understood. We asked whether the composition of bile acids in ALGS patients with good clinical outcomes differs from that in patients with poor outcomes and whether bile acids could be used as prognostic biomarkers. METHODS Blood for bile acid profiling was collected from genetically confirmed JAG1-associated ALGS patients before one year of age. A good prognosis was defined as survival with native liver and total bilirubin (TB) < 85.5 μmol/L, while a poor prognosis was defined as either liver transplantation, death from liver failure, or TB ≥ 85.5 μmol/L at the last follow-up. RESULTS We found that the concentrations of two poly-hydroxylated bile acids, tauro-2β,3α,7α,12α-tetrahydroxylated bile acid (THBA) and glyco-hyocholic acid (GHCA), were significantly increased in patients with good prognosis compared to those with poor prognosis [area under curve (AUC) = 0.836 and 0.782, respectively] in the discovery cohort. The same trend was also observed in the molar ratios of GHCA to glyco- chenodeoxycholic acid (GCDCA) and tetrahydroxylated bile acid (THCA) to tauro-chenodeoxycholic acid (TCDCA) (both AUC = 0.836). A validation cohort confirmed these findings. Notably, tauro-2β,3α,7α,12α-THBA achieved the highest prediction accuracy of 88.00% (92.31% sensitivity and 83.33% specificity); GHCA at > 607.69 nmol/L was associated with native liver survival [hazard ratio: 13.03, 95% confidence interval (CI): (2.662-63.753), P = 0.002]. CONCLUSIONS We identified two poly-hydroxylated bile acids as liver prognostic biomarkers of ALGS patients. Enhanced hydroxylation of bile acids may result in better clinical outcomes.
Collapse
Affiliation(s)
- Meng-Xuan Wang
- Department of Pediatrics, Jinshan Hospital of Fudan University, Shanghai, 201508, China
- The Center for Pediatric Liver Diseases, Children's Hospital of Fudan University, Shanghai, 201102, China
| | - Jun Han
- University of Victoria-Genome BC Proteomics Centre and Division of Medical Sciences, Victoria, British Columbia, Canada
| | - Teng Liu
- The Center for Pediatric Liver Diseases, Children's Hospital of Fudan University, Shanghai, 201102, China
| | - Ren-Xue Wang
- BC Cancer Research Centre, Vancouver, British Columbia, Canada
| | - Li-Ting Li
- The Center for Pediatric Liver Diseases, Children's Hospital of Fudan University, Shanghai, 201102, China
| | - Zhong-Die Li
- The Center for Pediatric Liver Diseases, Children's Hospital of Fudan University, Shanghai, 201102, China
| | - Jun-Cong Yang
- University of Victoria-Genome BC Proteomics Centre and Division of Medical Sciences, Victoria, British Columbia, Canada
| | - Lang-Li Liu
- The Center for Pediatric Liver Diseases, Children's Hospital of Fudan University, Shanghai, 201102, China
| | - Yi Lu
- The Center for Pediatric Liver Diseases, Children's Hospital of Fudan University, Shanghai, 201102, China
| | - Xin-Bao Xie
- The Center for Pediatric Liver Diseases, Children's Hospital of Fudan University, Shanghai, 201102, China
| | - Jing-Yu Gong
- Department of Pediatrics, Jinshan Hospital of Fudan University, Shanghai, 201508, China
| | - Shi-Yu Li
- The Center for Pediatric Liver Diseases, Children's Hospital of Fudan University, Shanghai, 201102, China
| | - Lei Zhang
- Institutes of Biomedical Sciences, Fudan University, Shanghai, 201199, China
| | - Victor Ling
- BC Cancer Research Centre, Vancouver, British Columbia, Canada
| | - Jian-She Wang
- The Center for Pediatric Liver Diseases, Children's Hospital of Fudan University, Shanghai, 201102, China.
- Shanghai Key Laboratory of Birth Defect, Shanghai, 201102, China.
| |
Collapse
|
39
|
Luu HN, Tran CTD, Wang R, Nguyen MVT, Tran MT, Tuong TTV, Tran QH, Le LC, Pham HTT, Vu HH, Bui NC, Ha HTT, Trinh DT, Thomas CE, Adams-Haduch J, Velikokhatnaya L, Schoen RE, Xie G, Jia W, Boffetta P, Clemente JC, Yuan JM. Associations between Ileal Juice Bile Acids and Colorectal Advanced Adenoma. Nutrients 2023; 15:2930. [PMID: 37447256 PMCID: PMC10346326 DOI: 10.3390/nu15132930] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2023] [Revised: 06/23/2023] [Accepted: 06/23/2023] [Indexed: 07/15/2023] Open
Abstract
BACKGROUND There is an urgent need to identify biomarkers for advanced adenoma, an important precursor of colorectal cancer (CRC). We aimed to determine alterations in ileal juice bile acids associated with colorectal advanced adenoma. METHODS We quantified a comprehensive panel of primary and secondary bile acids and their conjugates using an ultraperformance liquid chromatography triple-quadrupole mass spectrometric assay in ileal juice collected at colonoscopy from 46 study subjects (i.e., 14 biopsy-confirmed advanced adenomas and 32 controls free of adenoma or cancer). Using analysis of covariance (ANCOVA), we examined the differences in bile acid concentrations by disease status, adjusting for age, sex, body mass index, smoking status and type 2 diabetes. RESULTS The concentrations of hyodeoxycholic acid (HCA) species in ileal juice of the advanced adenoma patients (geometric mean = 4501.9 nM) were significantly higher than those of controls (geometric mean = 1292.3 nM, p = 0.001). The relative abundance of ursodeoxycholic acid (UDCA) in total bile acids was significantly reduced in cases than controls (0.73% in cases vs. 1.33% in controls; p = 0.046). No significant difference between cases and controls was observed for concentrations of total or specific primary bile acids (i.e., cholic acid (CA), chenodeoxycholic acid (CDCA) and their glycine- and taurine-conjugates) and total and specific major secondary bile acids (i.e., deoxycholic acid and lithocholic acid). CONCLUSIONS Colorectal advanced adenoma was associated with altered bile acids in ileal juice. The HCA species may promote the development of colorectal advanced adenoma, whereas gut microbiota responsible for the conversion of CDCA to UDCA may protect against it. Our findings have important implications for the use of bile acids as biomarkers in early detection of colorectal cancer.
Collapse
Affiliation(s)
- Hung N. Luu
- UPMC Hillman Cancer Center, University of Pittsburgh, Pittsburgh, PA 15232, USA; (R.W.); (C.E.T.); (J.A.-H.); (L.V.); (J.-M.Y.)
- Department of Epidemiology, School of Public Health, University of Pittsburgh, Pittsburgh, PA 15261, USA;
| | - Chi Thi-Du Tran
- Vietnam Colorectal Cancer and Polyps Research, Vinmec Healthcare System, Hanoi 10000, Vietnam; (C.T.-D.T.); (M.V.-T.N.); (M.T.T.); (T.T.-V.T.); (Q.H.T.)
- College of Health Sciences, VinUniversity (VinUni), Hanoi 10000, Vietnam;
- Center of Applied Sciences, Regenerative Medicine and Advanced Technologies, Vinmec Healthcare System, Hanoi 10000, Vietnam
| | - Renwei Wang
- UPMC Hillman Cancer Center, University of Pittsburgh, Pittsburgh, PA 15232, USA; (R.W.); (C.E.T.); (J.A.-H.); (L.V.); (J.-M.Y.)
| | - Mai Vu-Tuyet Nguyen
- Vietnam Colorectal Cancer and Polyps Research, Vinmec Healthcare System, Hanoi 10000, Vietnam; (C.T.-D.T.); (M.V.-T.N.); (M.T.T.); (T.T.-V.T.); (Q.H.T.)
| | - Mo Thi Tran
- Vietnam Colorectal Cancer and Polyps Research, Vinmec Healthcare System, Hanoi 10000, Vietnam; (C.T.-D.T.); (M.V.-T.N.); (M.T.T.); (T.T.-V.T.); (Q.H.T.)
| | - Thuy Thi-Van Tuong
- Vietnam Colorectal Cancer and Polyps Research, Vinmec Healthcare System, Hanoi 10000, Vietnam; (C.T.-D.T.); (M.V.-T.N.); (M.T.T.); (T.T.-V.T.); (Q.H.T.)
| | - Quang Hong Tran
- Vietnam Colorectal Cancer and Polyps Research, Vinmec Healthcare System, Hanoi 10000, Vietnam; (C.T.-D.T.); (M.V.-T.N.); (M.T.T.); (T.T.-V.T.); (Q.H.T.)
| | - Linh Cu Le
- College of Health Sciences, VinUniversity (VinUni), Hanoi 10000, Vietnam;
| | - Huong Thi-Thu Pham
- Department of Gastroenterology, Vinmec Healthcare System, Hanoi 10000, Vietnam; (H.T.-T.P.); (H.H.V.); (N.C.B.)
| | - Hien Huy Vu
- Department of Gastroenterology, Vinmec Healthcare System, Hanoi 10000, Vietnam; (H.T.-T.P.); (H.H.V.); (N.C.B.)
| | - Nam Chi Bui
- Department of Gastroenterology, Vinmec Healthcare System, Hanoi 10000, Vietnam; (H.T.-T.P.); (H.H.V.); (N.C.B.)
| | - Hien Thi-Thu Ha
- Department of Cytopathology, Vinmec Healthcare System, Hanoi 10000, Vietnam; (H.T.-T.H.); (D.T.T.)
| | - Dung Tuan Trinh
- Department of Cytopathology, Vinmec Healthcare System, Hanoi 10000, Vietnam; (H.T.-T.H.); (D.T.T.)
- Department of Cytopathology, Tam Anh General Hospital, Hanoi 10000, Vietnam
| | - Claire E. Thomas
- UPMC Hillman Cancer Center, University of Pittsburgh, Pittsburgh, PA 15232, USA; (R.W.); (C.E.T.); (J.A.-H.); (L.V.); (J.-M.Y.)
- Department of Epidemiology, School of Public Health, University of Pittsburgh, Pittsburgh, PA 15261, USA;
| | - Jennifer Adams-Haduch
- UPMC Hillman Cancer Center, University of Pittsburgh, Pittsburgh, PA 15232, USA; (R.W.); (C.E.T.); (J.A.-H.); (L.V.); (J.-M.Y.)
| | - Liudmilla Velikokhatnaya
- UPMC Hillman Cancer Center, University of Pittsburgh, Pittsburgh, PA 15232, USA; (R.W.); (C.E.T.); (J.A.-H.); (L.V.); (J.-M.Y.)
| | - Robert E. Schoen
- Department of Epidemiology, School of Public Health, University of Pittsburgh, Pittsburgh, PA 15261, USA;
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Medicine, University of Pittsburgh Medical Center, Pittsburgh, PA 15213, USA
| | - Guoxiang Xie
- University of Hawai’i Cancer Center, University of Hawaii, Honolulu, HI 96813, USA; (G.X.); (W.J.)
| | - Wei Jia
- University of Hawai’i Cancer Center, University of Hawaii, Honolulu, HI 96813, USA; (G.X.); (W.J.)
- School of Chinese Medicine, Hong Kong Baptist University, Kowloon, Hong Kong
| | - Paolo Boffetta
- Stony Brook Cancer Center, Stony Brook University, Stony Brook, NY 11794, USA;
- Department of Medical and Surgical Sciences, University of Bologna, 40138 Bologna, Italy
| | - Jose C. Clemente
- Icahn Institute for Genomics & Multiscale Biology, Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA;
- Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Jian-Min Yuan
- UPMC Hillman Cancer Center, University of Pittsburgh, Pittsburgh, PA 15232, USA; (R.W.); (C.E.T.); (J.A.-H.); (L.V.); (J.-M.Y.)
- Department of Epidemiology, School of Public Health, University of Pittsburgh, Pittsburgh, PA 15261, USA;
| |
Collapse
|
40
|
Ying Z, van Eenige R, Ge X, van Marwijk C, Lambooij JM, Guigas B, Giera M, de Boer JF, Coskun T, Qu H, Wang Y, Boon MR, Rensen PCN, Kooijman S. Combined GIP receptor and GLP1 receptor agonism attenuates NAFLD in male APOE∗3-Leiden.CETP mice. EBioMedicine 2023; 93:104684. [PMID: 37379656 DOI: 10.1016/j.ebiom.2023.104684] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2023] [Revised: 06/13/2023] [Accepted: 06/14/2023] [Indexed: 06/30/2023] Open
Abstract
BACKGROUND Combined glucose-dependent insulinotropic polypeptide receptor (GIPR) and glucagon-like peptide-1 receptor (GLP1R) agonism is superior to single GLP1R agonism with respect to glycemic control and weight loss in obese patients with or without type 2 diabetes. As insulin resistance and obesity are strong risk factors for nonalcoholic fatty liver disease (NAFLD), in the current study we investigated the effects of combined GIPR/GLP1R agonism on NAFLD development. METHODS Male APOE∗3-Leiden.CETP mice, a humanized model for diabetic dyslipidemia and NAFLD when fed a high-fat high-cholesterol diet, received subcutaneous injections with either vehicle, a GIPR agonist, a GLP1R agonist, or both agonists combined every other day. FINDINGS GIPR and GLP1R agonism reduced body weight and additively lowered fasting plasma levels of glucose, triglycerides and total cholesterol. Strikingly, we report an additive reduction in hepatic steatosis as evidenced by lower hepatic lipid content and NAFLD scores. Underlying the lipid-lowering effects were a reduced food intake and intestinal lipid absorption and an increased uptake of glucose and triglyceride-derived fatty acids by energy-combusting brown adipose tissue. Combined GIPR/GLP1R agonism also attenuated hepatic inflammation as evidenced by a decreased number of monocyte-derived Kupffer cells and a reduced expression of inflammatory markers. Together, the reduced hepatic steatosis and inflammation coincided with lowered markers of liver injury. INTERPRETATION We interpretate that GIPR and GLP1R agonism additively attenuate hepatic steatosis, lower hepatic inflammation, ameliorate liver injury, together preventing NAFLD development in humanized APOE∗3-Leiden.CETP mice. We anticipate that combined GIPR/GLP1R agonism is a promising strategy to attenuate NAFLD progression in humans. FUNDING This work was supported by a grant from the Netherlands CardioVascular Research Initiative: the Dutch Heart Foundation, Dutch Federation of University Medical Centers, the Netherlands Organization for Health Research and Development, and the Royal Netherlands Academy of Sciences [CVON-GENIUS-II] to P.C.N.R., a Lilly Research Award Program [LRAP] Award to P.C.N.R. and S.K., a Dutch Heart Foundation [2017T016] grant to S.K., and an NWO-VENI grant [09150161910073] to M.R.B.; J.F.D.B. is supported by the Nutrition and Health initiative of the University of Groningen; Z.Y. is supported by a full-time PhD scholarship from the China Scholarship Council (201806850094 to Z.Y.).
Collapse
Affiliation(s)
- Zhixiong Ying
- Division of Endocrinology, Department of Medicine, Leiden University Medical Center, Leiden, the Netherlands; Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Leiden, the Netherlands
| | - Robin van Eenige
- Division of Endocrinology, Department of Medicine, Leiden University Medical Center, Leiden, the Netherlands; Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Leiden, the Netherlands
| | - Xiaoke Ge
- Division of Endocrinology, Department of Medicine, Leiden University Medical Center, Leiden, the Netherlands; Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Leiden, the Netherlands
| | - Christy van Marwijk
- Division of Endocrinology, Department of Medicine, Leiden University Medical Center, Leiden, the Netherlands; Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Leiden, the Netherlands
| | - Joost M Lambooij
- Department of Parasitology, Leiden University Medical Center, Leiden, the Netherlands; Department of Cell and Chemical Biology, Leiden University Medical Center, Leiden, the Netherlands
| | - Bruno Guigas
- Department of Parasitology, Leiden University Medical Center, Leiden, the Netherlands
| | - Martin Giera
- The Center for Proteomics and Metabolomics, Leiden University Medical Center, Leiden, the Netherlands
| | - Jan Freark de Boer
- Departments of Pediatrics and Laboratory Medicine, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Tamer Coskun
- Department of Diabetes/Endocrine, Lilly Research Laboratories, Lilly Corporate Center, Indianapolis, IN, United States
| | - Hongchang Qu
- Department of Diabetes/Endocrine, Lilly Research Laboratories, Lilly Corporate Center, Indianapolis, IN, United States
| | - Yanan Wang
- Division of Endocrinology, Department of Medicine, Leiden University Medical Center, Leiden, the Netherlands; Med-X Institute, Center for Immunological and Metabolic Diseases and Department of Endocrinology, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an Jiaotong University, Xi'an, China
| | - Mariëtte R Boon
- Division of Endocrinology, Department of Medicine, Leiden University Medical Center, Leiden, the Netherlands; Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Leiden, the Netherlands
| | - Patrick C N Rensen
- Division of Endocrinology, Department of Medicine, Leiden University Medical Center, Leiden, the Netherlands; Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Leiden, the Netherlands
| | - Sander Kooijman
- Division of Endocrinology, Department of Medicine, Leiden University Medical Center, Leiden, the Netherlands; Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Leiden, the Netherlands.
| |
Collapse
|
41
|
Li W, Chen W, Niu X, Zhao C, Tu P, Li J, Liu W, Song Y. Characterization of Metabolic Correlations of Ursodeoxycholic Acid with Other Bile Acid Species through In Vitro Sequential Metabolism and Isomer-Focused Identification. Molecules 2023; 28:4801. [PMID: 37375356 DOI: 10.3390/molecules28124801] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2023] [Revised: 06/10/2023] [Accepted: 06/13/2023] [Indexed: 06/29/2023] Open
Abstract
As a first-line agent for cholestasis treatment in a clinic, ursodeoxycholic acid rectifies the perturbed bile acids (BAs) submetabolome in a holistic manner. Considering the endogenous distribution of ursodeoxycholic acid and extensive occurrences of isomeric metabolites, it is challenging to point out whether a given bile acid species is impacted by ursodeoxycholic acid in a direct or indirect manner, thus hindering the therapeutic mechanism clarification. Here, an in-depth exploration of the metabolism pattern of ursodeoxycholic acid was attempted. Sequential metabolism in vitro with enzyme-enriched liver microsomes was implemented to simulate the step-wise metabolism and to capture the metabolically labile intermediates in the absence of endogenous BAs. Squared energy-resolved mass spectrometry (ER2-MS) was utilized to achieve isomeric identification of the conjugated metabolites. As a result, 20 metabolites (M1-M20) in total were observed and confirmatively identified. Of those, eight metabolites were generated by hydroxylation, oxidation, and epimerization, which were further metabolized to nine glucuronides and three sulfates by uridine diphosphate-glycosyltransferases and sulfotransferases, respectively. Regarding a given phase II metabolite, the conjugation sites were correlated with first-generation breakdown graphs corresponding to the linkage fission mediated by collision-induced dissociation, and the structural nuclei were identified by matching second-generation breakdown graphs with the known structures. Together, except for intestinal-bacteria-involved biotransformation, the current study characterized BA species directly influenced by ursodeoxycholic acid administration. Moreover, sequential metabolism in vitro should be a meaningful way of characterizing the metabolic pathways of endogenous substances, and squared energy-resolved mass spectrometry is a legitimate tool for structurally identifying phase II metabolites.
Collapse
Affiliation(s)
- Wei Li
- Modern Research Center for Traditional Chinese Medicine, Beijing Research Institute of Chinese Medicine, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Wei Chen
- Modern Research Center for Traditional Chinese Medicine, Beijing Research Institute of Chinese Medicine, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Xiaoya Niu
- Modern Research Center for Traditional Chinese Medicine, Beijing Research Institute of Chinese Medicine, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Chen Zhao
- Zhangzhou Pien Tze Huang Pharmaceutical Co., Ltd., Zhangzhou 363000, China
| | - Pengfei Tu
- Modern Research Center for Traditional Chinese Medicine, Beijing Research Institute of Chinese Medicine, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Jun Li
- Modern Research Center for Traditional Chinese Medicine, Beijing Research Institute of Chinese Medicine, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Wenjing Liu
- School of Pharmacy, Henan University of Chinese Medicine, Zhengzhou 450046, China
| | - Yuelin Song
- Modern Research Center for Traditional Chinese Medicine, Beijing Research Institute of Chinese Medicine, Beijing University of Chinese Medicine, Beijing 100029, China
| |
Collapse
|
42
|
Kakiyama G, Rodriguez-Agudo D, Pandak WM. Mitochondrial Cholesterol Metabolites in a Bile Acid Synthetic Pathway Drive Nonalcoholic Fatty Liver Disease: A Revised "Two-Hit" Hypothesis. Cells 2023; 12:1434. [PMID: 37408268 PMCID: PMC10217489 DOI: 10.3390/cells12101434] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2023] [Revised: 05/15/2023] [Accepted: 05/17/2023] [Indexed: 07/07/2023] Open
Abstract
The rising prevalence of nonalcoholic fatty liver disease (NAFLD)-related cirrhosis highlights the need for a better understanding of the molecular mechanisms responsible for driving the transition of hepatic steatosis (fatty liver; NAFL) to steatohepatitis (NASH) and fibrosis/cirrhosis. Obesity-related insulin resistance (IR) is a well-known hallmark of early NAFLD progression, yet the mechanism linking aberrant insulin signaling to hepatocyte inflammation has remained unclear. Recently, as a function of more distinctly defining the regulation of mechanistic pathways, hepatocyte toxicity as mediated by hepatic free cholesterol and its metabolites has emerged as fundamental to the subsequent necroinflammation/fibrosis characteristics of NASH. More specifically, aberrant hepatocyte insulin signaling, as found with IR, leads to dysregulation in bile acid biosynthetic pathways with the subsequent intracellular accumulation of mitochondrial CYP27A1-derived cholesterol metabolites, (25R)26-hydroxycholesterol and 3β-Hydroxy-5-cholesten-(25R)26-oic acid, which appear to be responsible for driving hepatocyte toxicity. These findings bring forth a "two-hit" interpretation as to how NAFL progresses to NAFLD: abnormal hepatocyte insulin signaling, as occurs with IR, develops as a "first hit" that sequentially drives the accumulation of toxic CYP27A1-driven cholesterol metabolites as the "second hit". In the following review, we examine the mechanistic pathway by which mitochondria-derived cholesterol metabolites drive the development of NASH. Insights into mechanistic approaches for effective NASH intervention are provided.
Collapse
Affiliation(s)
- Genta Kakiyama
- Department of Internal Medicine, Virginia Commonwealth University School of Medicine, Richmond, VA 23298, USA; (D.R.-A.); (W.M.P.)
- Research Services, Central Virginia Veterans Affairs Healthcare System, Richmond, VA 23249, USA
| | - Daniel Rodriguez-Agudo
- Department of Internal Medicine, Virginia Commonwealth University School of Medicine, Richmond, VA 23298, USA; (D.R.-A.); (W.M.P.)
- Research Services, Central Virginia Veterans Affairs Healthcare System, Richmond, VA 23249, USA
| | - William M. Pandak
- Department of Internal Medicine, Virginia Commonwealth University School of Medicine, Richmond, VA 23298, USA; (D.R.-A.); (W.M.P.)
- Research Services, Central Virginia Veterans Affairs Healthcare System, Richmond, VA 23249, USA
| |
Collapse
|
43
|
Lei K, Yuan M, Li S, Zhou Q, Li M, Zeng D, Guo Y, Guo L. Performance evaluation of E-nose and E-tongue combined with machine learning for qualitative and quantitative assessment of bear bile powder. Anal Bioanal Chem 2023:10.1007/s00216-023-04740-5. [PMID: 37199792 DOI: 10.1007/s00216-023-04740-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2023] [Revised: 05/06/2023] [Accepted: 05/09/2023] [Indexed: 05/19/2023]
Abstract
Bear bile powder (BBP) is a valuable animal-derived product with a huge adulteration problem on market. It is a crucially important task to identify BBP and its counterfeit. Electronic sensory technologies are the inheritance and development of traditional empirical identification. Considering that each drug has its own specific odor and taste characteristics, electronic tongue (E-tongue), electronic nose (E-nose) and GC-MS were used to evaluate the aroma and taste of BBP and its common counterfeit. Two active components of BBP, namely tauroursodeoxycholic acid (TUDCA) and taurochenodeoxycholic acid (TCDCA) were measured and linked with the electronic sensory data. The results showed that bitterness was the main flavor of TUDCA in BBP, saltiness and umami were the main flavor of TCDCA. The volatiles detected by E-nose and GC-MS were mainly aldehydes, ketones, alcohols, hydrocarbons, carboxylic acids, heterocyclic, lipids, and amines, mainly earthy, musty, coffee, bitter almond, burnt, pungent odor descriptions. Four different machine learning algorithms (backpropagation neural network, support vector machine, K-nearest neighbor, and random forest) were used to identify BBP and its counterfeit, and the regression performance of these four algorithms was also evaluated. For qualitative identification, the algorithm of random forest has shown the best performance, with 100% accuracy, precision, recall and F1-score. Also, the random forest algorithm has the best R2 and the lowest RMSE in terms of quantitative prediction.
Collapse
Affiliation(s)
- Kelu Lei
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, No. 1166, Liutai Avenue, Chengdu, 611137, China
| | - Minghao Yuan
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, No. 1166, Liutai Avenue, Chengdu, 611137, China
| | - Sihui Li
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, No. 1166, Liutai Avenue, Chengdu, 611137, China
| | - Qiang Zhou
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, No. 1166, Liutai Avenue, Chengdu, 611137, China
| | - Meifeng Li
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, No. 1166, Liutai Avenue, Chengdu, 611137, China
- School of Public Health, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Dafu Zeng
- Chengdu Jingbo Biotechnology Co., Ltd, No.39 Renhe Street, Chengdu, 611731, China
| | - Yiping Guo
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, No. 1166, Liutai Avenue, Chengdu, 611137, China.
| | - Li Guo
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, No. 1166, Liutai Avenue, Chengdu, 611137, China.
| |
Collapse
|
44
|
Affiliation(s)
- Robert A Quinn
- Department of Biochemistry and Molecular Biology, Michigan State University, East Lansing, MI, USA.
| | - Christian Martin
- Department of Biochemistry and Molecular Biology, Michigan State University, East Lansing, MI, USA
| | - Douglas V Guzior
- Department of Biochemistry and Molecular Biology, Michigan State University, East Lansing, MI, USA
- Department of Microbiology and Molecular Genetics, Michigan State University, East Lansing, MI, USA
| |
Collapse
|
45
|
Lee D, Iwasaki W, Hori S, Kubota N, Ishizuka S. Ingesting a fermented milk product reduces liver triacylglycerol accumulation and normalizes gut permeability in rats even under a cholic acid-fed condition. J Funct Foods 2023. [DOI: 10.1016/j.jff.2023.105523] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/03/2023] Open
|
46
|
Akl MG, Li L, Baccetto R, Phanse S, Zhang Q, Trites MJ, McDonald S, Aoki H, Babu M, Widenmaier SB. Complementary gene regulation by NRF1 and NRF2 protects against hepatic cholesterol overload. Cell Rep 2023; 42:112399. [PMID: 37060561 DOI: 10.1016/j.celrep.2023.112399] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2022] [Revised: 10/04/2022] [Accepted: 03/30/2023] [Indexed: 04/16/2023] Open
Abstract
Hepatic cholesterol overload promotes steatohepatitis. Insufficient understanding of liver stress defense impedes therapy development. Here, we elucidate the role of stress defense transcription factors, nuclear factor erythroid 2 related factor-1 (NRF1) and -2 (NRF2), in counteracting cholesterol-linked liver stress. Using a diet that increases liver cholesterol storage, expression profiles and phenotypes of liver from mice with hepatocyte deficiency of NRF1, NRF2, or both are compared with controls, and chromatin immunoprecipitation sequencing is undertaken to identify target genes. Results show NRF1 and NRF2 co-regulate genes that eliminate cholesterol and mitigate inflammation and oxidative damage. Combined deficiency, but not deficiency of either alone, results in severe steatohepatitis, hepatic cholesterol overload and crystallization, altered bile acid metabolism, and decreased biliary cholesterol. Moreover, therapeutic effects of NRF2-activating drug bardoxolone require NRF1 and are supplemented by NRF1 overexpression. Thus, we discover complementary gene programming by NRF1 and NRF2 that counteract cholesterol-associated fatty liver disease progression.
Collapse
Affiliation(s)
- May G Akl
- Department of Anatomy, Physiology, and Pharmacology, University of Saskatchewan, Saskatoon, SK, Canada; Department of Physiology, Faculty of Medicine, University of Alexandria, Alexandria, Egypt
| | - Lei Li
- Department of Anatomy, Physiology, and Pharmacology, University of Saskatchewan, Saskatoon, SK, Canada
| | - Raquel Baccetto
- Department of Anatomy, Physiology, and Pharmacology, University of Saskatchewan, Saskatoon, SK, Canada
| | - Sadhna Phanse
- Department of Chemistry and Biochemistry, University of Regina, Regina, SK, Canada
| | - Qingzhou Zhang
- Department of Chemistry and Biochemistry, University of Regina, Regina, SK, Canada
| | - Michael J Trites
- Department of Anatomy, Physiology, and Pharmacology, University of Saskatchewan, Saskatoon, SK, Canada
| | - Sherin McDonald
- Department of Anatomy, Physiology, and Pharmacology, University of Saskatchewan, Saskatoon, SK, Canada
| | - Hiroyuki Aoki
- Department of Chemistry and Biochemistry, University of Regina, Regina, SK, Canada
| | - Mohan Babu
- Department of Chemistry and Biochemistry, University of Regina, Regina, SK, Canada
| | - Scott B Widenmaier
- Department of Anatomy, Physiology, and Pharmacology, University of Saskatchewan, Saskatoon, SK, Canada.
| |
Collapse
|
47
|
Fickert P. Bacteria as key players in primary sclerosing cholangitis? Gut 2023; 72:607-608. [PMID: 35764378 DOI: 10.1136/gutjnl-2022-327876] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Accepted: 06/16/2022] [Indexed: 12/08/2022]
Affiliation(s)
- Peter Fickert
- Division of Gastroenterology and Hepatology, Department of Medicine, Medical University of Graz, Graz, Austria
| |
Collapse
|
48
|
Wan L, Li L, Zhang X, Li B, Harro JM, Shirtliff ME. Effects of palm olein and palm stearin on cecal and fecal microbiota of C57BL/6J mice under low and high fat intakes. Food Chem 2023; 404:134693. [DOI: 10.1016/j.foodchem.2022.134693] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Revised: 10/14/2022] [Accepted: 10/17/2022] [Indexed: 11/22/2022]
|
49
|
Parekh PY, Patel VI, Khimani MR, Bahadur P. Self-assembly of bile salts and their mixed aggregates as building blocks for smart aggregates. Adv Colloid Interface Sci 2023; 312:102846. [PMID: 36736167 DOI: 10.1016/j.cis.2023.102846] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2022] [Revised: 01/11/2023] [Accepted: 01/22/2023] [Indexed: 01/27/2023]
Abstract
The present communication offers a comprehensive overview of the self-assembly of bile salts emphasizing their mixed smart aggregates with a variety of amphiphiles. Using an updated literature survey, we have explored the dissimilar interactions of bile salts with different types of surfactants, phospholipids, ionic liquids, drugs, and a variety of natural and synthetic polymers. While assembling this review, special attention was also provided to the potency of bile salts to alter the size/shape of aggregates formed by several amphiphiles to use these aggregates for solubility improvement of medicinally important compounds, active pharmaceutical ingredients, and also to develop their smart delivery vehicles. A fundamental understanding of bile salt mixed aggregates will enable the development of new strategies for improving the bioavailability of drugs solubilized in newly developed potential hosts and to formulate smart aggregates of desired morphology for specific targeted applications. It enriches our existing knowledge of the distinct interactions exerted in mixed systems of bile salts with variety of amphiphiles. By virtue of this, researchers can get innovative ideas to construct novel nanoaggregates from bile salts by incorporating various amphiphiles that serve as a building block for smart aggregates for their numerous industrial applications.
Collapse
Affiliation(s)
- Paresh Y Parekh
- Department of Chemistry, Veer Narmad South Gujarat University, Surat 395007, Gujarat, India
| | - Vijay I Patel
- Department of Chemistry, Navyug Science College, Rander Road, Surat 395009, Gujarat, India.
| | - Mehul R Khimani
- Countryside International School, Nr. Bhesan Railway Crossing, CIS Barbodhan Road, Surat 394125, Gujarat, India
| | - Pratap Bahadur
- Department of Chemistry, Veer Narmad South Gujarat University, Surat 395007, Gujarat, India
| |
Collapse
|
50
|
Tacchi F, Orozco-Aguilar J, Valero-Breton M, Cabello-Verrugio C. Bile Acids Alter the Autophagy and Mitogenesis in Skeletal Muscle Cells. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2023; 1408:183-199. [PMID: 37093428 DOI: 10.1007/978-3-031-26163-3_10] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/25/2023]
Abstract
Muscle atrophy decreases muscle mass with the subsequent loss of muscle function. Among the mechanisms that trigger sarcopenia is mitochondrial dysfunction. Mitochondria, whose primary function is to produce ATP, are dynamic organelles that present the process of formation (mitogenesis) and elimination (mitophagy). Failure of any of these processes contributes to mitochondrial malfunction. Mitogenesis is mainly controlled by Peroxisome proliferator-activated receptor-gamma coactivator-1alpha (PGC-1α), a transcriptional coactivator that regulates the expression of TFAM, which participates in mitogenesis. Mitophagy is a process of selective autophagy. Autophagy corresponds to a degradative pathway of protein complexes and organelles. Liver disease caused sarcopenia and increased bile acids in the blood. We demonstrated that the treatment with cholic (CA) or deoxycholic (DCA) bile acids generates mitochondrial dysfunction and loss of biomass. This work assessed whether CA and DCA alter autophagy and mitogenesis. For this, western blot evaluated the autophagy process by determining the protein levels of the LC3II/LC3I ratio. In addition, we assessed mitogenesis using a luciferase-coupled plasmid reporter for the PGC-1α promoter and the protein levels of TFAM by western blot. Our results indicate that treatment with CA or DCA induces autophagy, represented by an increase in the LC3II/LC3I ratio. In addition, a decreased autophagic flux was observed. On the other hand, when treated with CA or DCA, a decrease in the activity of the PGC-1α promoter was observed. However, the levels of TFAM increased in myotubes incubated with CA and DCA. Our results demonstrate that CA and DCA modulate autophagy ad mitogenesis in C2C12 myotubes.
Collapse
Affiliation(s)
- Franco Tacchi
- Laboratory of Muscle Pathology, Fragility and Aging, Faculty of Life Sciences, Universidad Andres Bello, Santiago, 8370146, Chile
- Millennium Institute on Immunology and Immunotherapy, Faculty of Life Sciences, Universidad Andres Bello, Santiago, Chile
- Center for the Development of Nanoscience and Nanotechnology (CEDENNA), Universidad de Santiago de Chile, Santiago, Chile
| | - Josué Orozco-Aguilar
- Laboratory of Muscle Pathology, Fragility and Aging, Faculty of Life Sciences, Universidad Andres Bello, Santiago, 8370146, Chile
- Millennium Institute on Immunology and Immunotherapy, Faculty of Life Sciences, Universidad Andres Bello, Santiago, Chile
- Center for the Development of Nanoscience and Nanotechnology (CEDENNA), Universidad de Santiago de Chile, Santiago, Chile
- Laboratorio de Ensayos Biológicos (LEBi), Universidad de Costa Rica, San José, Costa Rica
- Facultad de Farmacia, Universidad de Costa Rica, San José, Costa Rica
| | - Mayalen Valero-Breton
- Laboratory of Muscle Pathology, Fragility and Aging, Faculty of Life Sciences, Universidad Andres Bello, Santiago, 8370146, Chile
- Millennium Institute on Immunology and Immunotherapy, Faculty of Life Sciences, Universidad Andres Bello, Santiago, Chile
- Center for the Development of Nanoscience and Nanotechnology (CEDENNA), Universidad de Santiago de Chile, Santiago, Chile
| | - Claudio Cabello-Verrugio
- Laboratory of Muscle Pathology, Fragility and Aging, Faculty of Life Sciences, Universidad Andres Bello, Santiago, 8370146, Chile.
- Millennium Institute on Immunology and Immunotherapy, Faculty of Life Sciences, Universidad Andres Bello, Santiago, Chile.
- Center for the Development of Nanoscience and Nanotechnology (CEDENNA), Universidad de Santiago de Chile, Santiago, Chile.
| |
Collapse
|