701
|
Palau-Rodriguez M, Tulipani S, Isabel Queipo-Ortuño M, Urpi-Sarda M, Tinahones FJ, Andres-Lacueva C. Metabolomic insights into the intricate gut microbial-host interaction in the development of obesity and type 2 diabetes. Front Microbiol 2015; 6:1151. [PMID: 26579078 PMCID: PMC4621279 DOI: 10.3389/fmicb.2015.01151] [Citation(s) in RCA: 96] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2015] [Accepted: 10/05/2015] [Indexed: 12/31/2022] Open
Abstract
Gut microbiota has recently been proposed as a crucial environmental factor in the development of metabolic diseases such as obesity and type 2 diabetes, mainly due to its contribution in the modulation of several processes including host energy metabolism, gut epithelial permeability, gut peptide hormone secretion, and host inflammatory state. Since the symbiotic interaction between the gut microbiota and the host is essentially reflected in specific metabolic signatures, much expectation is placed on the application of metabolomic approaches to unveil the key mechanisms linking the gut microbiota composition and activity with disease development. The present review aims to summarize the gut microbial-host co-metabolites identified so far by targeted and untargeted metabolomic studies in humans, in association with impaired glucose homeostasis and/or obesity. An alteration of the co-metabolism of bile acids, branched fatty acids, choline, vitamins (i.e., niacin), purines, and phenolic compounds has been associated so far with the obese or diabese phenotype, in respect to healthy controls. Furthermore, anti-diabetic treatments such as metformin and sulfonylurea have been observed to modulate the gut microbiota or at least their metabolic profiles, thereby potentially affecting insulin resistance through indirect mechanisms still unknown. Despite the scarcity of the metabolomic studies currently available on the microbial-host crosstalk, the data-driven results largely confirmed findings independently obtained from in vitro and animal model studies, putting forward the mechanisms underlying the implication of a dysfunctional gut microbiota in the development of metabolic disorders.
Collapse
Affiliation(s)
- Magali Palau-Rodriguez
- Biomarkers and Nutrimetabolomic Lab., Nutrition and Food Science Department, XaRTA, INSA, Campus Torribera, Pharmacy Faculty, University of Barcelona Barcelona, Spain
| | - Sara Tulipani
- Biomarkers and Nutrimetabolomic Lab., Nutrition and Food Science Department, XaRTA, INSA, Campus Torribera, Pharmacy Faculty, University of Barcelona Barcelona, Spain ; Biomedical Research Institute (IBIMA), Service of Endocrinology and Nutrition, Malaga Hospital Complex (Virgen de la Victoria), University of Malaga Malaga, Spain
| | - Maria Isabel Queipo-Ortuño
- Biomedical Research Institute (IBIMA), Service of Endocrinology and Nutrition, Malaga Hospital Complex (Virgen de la Victoria), University of Malaga Malaga, Spain ; CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), Instituto de Salud Carlos III (ISCIII) Madrid, Spain
| | - Mireia Urpi-Sarda
- Biomarkers and Nutrimetabolomic Lab., Nutrition and Food Science Department, XaRTA, INSA, Campus Torribera, Pharmacy Faculty, University of Barcelona Barcelona, Spain
| | - Francisco J Tinahones
- Biomedical Research Institute (IBIMA), Service of Endocrinology and Nutrition, Malaga Hospital Complex (Virgen de la Victoria), University of Malaga Malaga, Spain ; CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), Instituto de Salud Carlos III (ISCIII) Madrid, Spain
| | - Cristina Andres-Lacueva
- Biomarkers and Nutrimetabolomic Lab., Nutrition and Food Science Department, XaRTA, INSA, Campus Torribera, Pharmacy Faculty, University of Barcelona Barcelona, Spain
| |
Collapse
|
702
|
Fu T, Kim YC, Byun S, Kim DH, Seok S, Suino-Powell K, Xu HE, Kemper B, Kemper JK. FXR Primes the Liver for Intestinal FGF15 Signaling by Transient Induction of β-Klotho. Mol Endocrinol 2015; 30:92-103. [PMID: 26505219 DOI: 10.1210/me.2015-1226] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
The bile acid (BA)-sensing nuclear receptor, farnesoid X receptor (FXR), regulates postprandial metabolic responses, including inhibition of BA synthesis, by inducing the intestinal hormone, fibroblast growth factor (FGF)15 (FGF19 in human). In this study, we tested a novel hypothesis that FXR not only induces intestinal FGF15 but also primes the liver for effectively responding to the signal by transcriptional induction of the obligate coreceptor for FGF15, β-Klotho (βKL). Activation of FXR by a synthetic agonist, GW4064, in mice increased occupancy of FXR and its DNA-binding partner, retinoid X receptor-α, at FGF15-signaling component genes, particularly βKL, and induced expression of these genes. Interestingly, mRNA levels of Fgfr4, the FGF15 receptor, were not increased by GW4064, but protein levels increased as a result of βKL-dependent increased protein stability. Both FGF receptor 4 and βKL protein levels were substantially decreased in FXR-knockout (KO) mice, and FGF19 signaling, monitored by phosphorylated ERK, was blunted in FXR-KO mice, FXR-KO mouse hepatocytes, and FXR-down-regulated human hepatocytes. Overexpression of βKL in FXR-lacking hepatocytes partially restored FGF19 signaling and inhibition by FGF19 of Cyp7a1, which encodes the rate-limiting BA biosynthetic enzyme. In mice, transient inductions of intestinal Fgf15 and hepatic βKL were temporally correlated after GW4064 treatment, and pretreatment of hepatocytes with GW4064 before FGF19 treatment enhanced FGF19 signaling, which was abolished by transcriptional inhibition or βKL down-regulation. This study identifies FXR as a gut-liver metabolic coordinator for FGF15/19 action that orchestrates transient induction of hepatic βKL and intestinal Fgf15/19 in a temporally correlated manner.
Collapse
Affiliation(s)
- Ting Fu
- Department of Molecular and Integrative Physiology (T.F., Y.-C.K., S.B., D.-H.K., S.S., B.K., J.K.K.), University of Illinois at Urbana-Champaign, Urbana, Illinois 61801; Laboratory of Structure Sciences (K.S.-P., H.E.X.), Van Andel Research Institute, Grand Rapids, Michigan 49503; and Van Andel Research Institute-Shanghai Institute of Materia Medica (H.E.X.), Center for Structure and Function of Drug Targets, Chinese Academy of Sciences-Key Laboratory of Receptor Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Young-Chae Kim
- Department of Molecular and Integrative Physiology (T.F., Y.-C.K., S.B., D.-H.K., S.S., B.K., J.K.K.), University of Illinois at Urbana-Champaign, Urbana, Illinois 61801; Laboratory of Structure Sciences (K.S.-P., H.E.X.), Van Andel Research Institute, Grand Rapids, Michigan 49503; and Van Andel Research Institute-Shanghai Institute of Materia Medica (H.E.X.), Center for Structure and Function of Drug Targets, Chinese Academy of Sciences-Key Laboratory of Receptor Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Sangwon Byun
- Department of Molecular and Integrative Physiology (T.F., Y.-C.K., S.B., D.-H.K., S.S., B.K., J.K.K.), University of Illinois at Urbana-Champaign, Urbana, Illinois 61801; Laboratory of Structure Sciences (K.S.-P., H.E.X.), Van Andel Research Institute, Grand Rapids, Michigan 49503; and Van Andel Research Institute-Shanghai Institute of Materia Medica (H.E.X.), Center for Structure and Function of Drug Targets, Chinese Academy of Sciences-Key Laboratory of Receptor Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Dong-Hyun Kim
- Department of Molecular and Integrative Physiology (T.F., Y.-C.K., S.B., D.-H.K., S.S., B.K., J.K.K.), University of Illinois at Urbana-Champaign, Urbana, Illinois 61801; Laboratory of Structure Sciences (K.S.-P., H.E.X.), Van Andel Research Institute, Grand Rapids, Michigan 49503; and Van Andel Research Institute-Shanghai Institute of Materia Medica (H.E.X.), Center for Structure and Function of Drug Targets, Chinese Academy of Sciences-Key Laboratory of Receptor Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Sunmi Seok
- Department of Molecular and Integrative Physiology (T.F., Y.-C.K., S.B., D.-H.K., S.S., B.K., J.K.K.), University of Illinois at Urbana-Champaign, Urbana, Illinois 61801; Laboratory of Structure Sciences (K.S.-P., H.E.X.), Van Andel Research Institute, Grand Rapids, Michigan 49503; and Van Andel Research Institute-Shanghai Institute of Materia Medica (H.E.X.), Center for Structure and Function of Drug Targets, Chinese Academy of Sciences-Key Laboratory of Receptor Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Kelly Suino-Powell
- Department of Molecular and Integrative Physiology (T.F., Y.-C.K., S.B., D.-H.K., S.S., B.K., J.K.K.), University of Illinois at Urbana-Champaign, Urbana, Illinois 61801; Laboratory of Structure Sciences (K.S.-P., H.E.X.), Van Andel Research Institute, Grand Rapids, Michigan 49503; and Van Andel Research Institute-Shanghai Institute of Materia Medica (H.E.X.), Center for Structure and Function of Drug Targets, Chinese Academy of Sciences-Key Laboratory of Receptor Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - H Eric Xu
- Department of Molecular and Integrative Physiology (T.F., Y.-C.K., S.B., D.-H.K., S.S., B.K., J.K.K.), University of Illinois at Urbana-Champaign, Urbana, Illinois 61801; Laboratory of Structure Sciences (K.S.-P., H.E.X.), Van Andel Research Institute, Grand Rapids, Michigan 49503; and Van Andel Research Institute-Shanghai Institute of Materia Medica (H.E.X.), Center for Structure and Function of Drug Targets, Chinese Academy of Sciences-Key Laboratory of Receptor Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Byron Kemper
- Department of Molecular and Integrative Physiology (T.F., Y.-C.K., S.B., D.-H.K., S.S., B.K., J.K.K.), University of Illinois at Urbana-Champaign, Urbana, Illinois 61801; Laboratory of Structure Sciences (K.S.-P., H.E.X.), Van Andel Research Institute, Grand Rapids, Michigan 49503; and Van Andel Research Institute-Shanghai Institute of Materia Medica (H.E.X.), Center for Structure and Function of Drug Targets, Chinese Academy of Sciences-Key Laboratory of Receptor Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Jongsook Kim Kemper
- Department of Molecular and Integrative Physiology (T.F., Y.-C.K., S.B., D.-H.K., S.S., B.K., J.K.K.), University of Illinois at Urbana-Champaign, Urbana, Illinois 61801; Laboratory of Structure Sciences (K.S.-P., H.E.X.), Van Andel Research Institute, Grand Rapids, Michigan 49503; and Van Andel Research Institute-Shanghai Institute of Materia Medica (H.E.X.), Center for Structure and Function of Drug Targets, Chinese Academy of Sciences-Key Laboratory of Receptor Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| |
Collapse
|
703
|
Flowers SA, Ellingrod VL. The Microbiome in Mental Health: Potential Contribution of Gut Microbiota in Disease and Pharmacotherapy Management. Pharmacotherapy 2015; 35:910-6. [DOI: 10.1002/phar.1640] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Affiliation(s)
- Stephanie A. Flowers
- College of Pharmacy, Clinical Social and Administrative Sciences; University of Michigan; Ann Arbor Michigan
| | - Vicki L. Ellingrod
- College of Pharmacy, Clinical Social and Administrative Sciences; University of Michigan; Ann Arbor Michigan
- Department of Psychiatry; School of Medicine; University of Michigan; Ann Arbor Michigan
| |
Collapse
|
704
|
Lin Y, Nishiyama T, Kurosawa M, Tamakoshi A, Kubo T, Fujino Y, Kikuchi S. Association between shift work and the risk of death from biliary tract cancer in Japanese men. BMC Cancer 2015; 15:757. [PMID: 26490349 PMCID: PMC4618019 DOI: 10.1186/s12885-015-1722-y] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2014] [Accepted: 10/08/2015] [Indexed: 12/14/2022] Open
Abstract
Background There is increasing evidence suggesting that shift work involving night work may increase cancer risk. Methods We examined the association between working rotating shifts and the risk of death from biliary tract cancer among Japanese men who participated in the Japan Collaborative Cohort Study. Of the 46,395 men recruited, 22,224 men aged 40–65 at baseline (1988–1990) who reported working full-time or were self-employed were included in the present analysis. The study subjects were followed through December 31, 2009. Information regarding occupation and lifestyle factors was collected using a self-administered questionnaire. Cox proportional hazard models were used to estimate the hazard ratio (HR) and 95 % confidence interval (CI) for the risk of death from biliary tract cancer in relation to shift work. Results During a mean 17-year follow-up, we observed 94 biliary tract cancer deaths, including 23 deaths from gallbladder cancer and 71 deaths from extrahepatic bile duct cancer. Overall, shift work was associated with a statistically non-significant increase in the risk of biliary tract cancer, with an HR of 1.50 (95 % CI: 0.81-2.77), among rotating shift workers. When the analysis was limited to extrahepatic bile duct cancer, a significant association appeared, with a multivariable-adjusted HR of 1.93 (95 % CI: 1.00-3.72) for rotating shift workers. Conclusion Our data indicate that shift work may be associated with increased risk of death from extrahepatic bile duct cancer in this cohort of Japanese men. The association with gallbladder cancer remains unclear because of the small number of deaths.
Collapse
Affiliation(s)
- Yingsong Lin
- Department of Public Health, Aichi Medical University School of Medicine, 1-1 Yazakokarimata, Nagakute, Aichi, 4980-1195, Japan.
| | - Takeshi Nishiyama
- Department of Public Health, Aichi Medical University School of Medicine, 1-1 Yazakokarimata, Nagakute, Aichi, 4980-1195, Japan.
| | - Michiko Kurosawa
- Department of Epidemiology and Environmental Medicine, Juntendo University School of Medicine, Hongo 2-1-1, Bunkyo-ku, Tokyo, 113-8421, Japan.
| | - Akiko Tamakoshi
- Department of Public Health, Hokkaido University Graduate School of Medicine, North 15, West 7, Kita-ku, Sapporo, 060-8638, Japan.
| | - Tatsuhiko Kubo
- Department of Preventive Medicine and Community Health, University of Occupational and Environmental Health, Japan, 1-1, Iseigaoka, Yahata-nishi-ku, 807-8555, Kitakyushu, Japan.
| | - Yoshihisa Fujino
- Department of Preventive Medicine and Community Health, University of Occupational and Environmental Health, Japan, 1-1, Iseigaoka, Yahata-nishi-ku, 807-8555, Kitakyushu, Japan.
| | - Shogo Kikuchi
- Department of Public Health, Aichi Medical University School of Medicine, 1-1 Yazakokarimata, Nagakute, Aichi, 4980-1195, Japan.
| | | |
Collapse
|
705
|
Shihabudeen MS, Roy D, James J, Thirumurugan K. Chenodeoxycholic acid, an endogenous FXR ligand alters adipokines and reverses insulin resistance. Mol Cell Endocrinol 2015; 414:19-28. [PMID: 26188168 DOI: 10.1016/j.mce.2015.07.012] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/25/2015] [Revised: 06/05/2015] [Accepted: 07/14/2015] [Indexed: 12/20/2022]
Abstract
Adipose tissue secretes adipokines that regulate insulin sensitivity in adipocytes and other peripheral tissues critical to glucose metabolism. Insulin resistance is associated with severe alterations in adipokines characterized by release of increased pro-inflammatory cytokines and decreased anti-inflammatory cytokines from adipose tissue. The role of Farnesoid X receptor (FXR) activation on adipokines in relation to adipose tissue inflammation and insulin resistance is not completely explored. For the first time, we have evaluated the ability of Chenodeoxycholic acid (CDCA), an endogenous FXR ligand, in restoring the disturbance in adipokine secretion and insulin resistance in palmitate treated 3T3-L1 cells and adipose tissues of High fat diet (HFD) rats. CDCA suppressed several of the tested pro-inflammatory adipokines (TNF-α, MCP-1, IL-6, Chemerin, PAI, RBP4, resistin, vaspin), and enhanced the major anti-inflammatory and insulin sensitizing adipokines (adiponectin, leptin). CDCA suppressed the activation of critical inflammatory regulators such as NF-κB and IKKβ which are activated by palmitate treatment in differentiated cells and HFD in rats. We show the altered adipokines in insulin resistance, its association with inflammatory regulators, and the role of CDCA in amelioration of insulin resistance by modulation of adipokines.
Collapse
Affiliation(s)
- Mohamed Sham Shihabudeen
- Structural Biology Lab, Centre for Biomedical Research, School of Bio Sciences and Technology, VIT University, Vellore, 632 014, Tamil Nadu, India
| | - Debasish Roy
- Structural Biology Lab, Centre for Biomedical Research, School of Bio Sciences and Technology, VIT University, Vellore, 632 014, Tamil Nadu, India
| | - Joel James
- Structural Biology Lab, Centre for Biomedical Research, School of Bio Sciences and Technology, VIT University, Vellore, 632 014, Tamil Nadu, India
| | - Kavitha Thirumurugan
- Structural Biology Lab, Centre for Biomedical Research, School of Bio Sciences and Technology, VIT University, Vellore, 632 014, Tamil Nadu, India.
| |
Collapse
|
706
|
Govindan JA, Jayamani E, Zhang X, Breen P, Larkins-Ford J, Mylonakis E, Ruvkun G. Lipid signalling couples translational surveillance to systemic detoxification in Caenorhabditis elegans. Nat Cell Biol 2015; 17:1294-303. [PMID: 26322678 PMCID: PMC4589496 DOI: 10.1038/ncb3229] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2014] [Accepted: 07/24/2015] [Indexed: 12/17/2022]
Abstract
Translation in eukaryotes is followed to detect toxins and virulence factors and coupled to the induction of defence pathways. Caenorhabditis elegans germline-specific mutations in translation components are detected by this system to induce detoxification and immune responses in distinct somatic cells. An RNA interference screen revealed gene inactivations that act at multiple steps in lipid biosynthetic and kinase pathways upstream of MAP kinase to mediate the systemic communication of translation defects to induce detoxification genes. Mammalian bile acids can rescue the defect in detoxification gene induction caused by C. elegans lipid biosynthetic gene inactivations. Extracts prepared from C. elegans with translation deficits but not from the wild type can also rescue detoxification gene induction in lipid-biosynthesis-defective strains. These eukaryotic antibacterial countermeasures are not ignored by bacteria: particular bacterial species suppress normal C. elegans detoxification responses to mutations in translation factors.
Collapse
Affiliation(s)
- J Amaranath Govindan
- Department of Molecular Biology, Massachusetts General Hospital, Boston, Massachusetts 02114, USA
- Department of Genetics, Harvard Medical School, Boston, Massachusetts 02115, USA
| | - Elamparithi Jayamani
- Department of Molecular Biology, Massachusetts General Hospital, Boston, Massachusetts 02114, USA
- Division of Infectious Diseases, Rhode Island Hospital, Alpert Medical School of Brown University, Providence, Rhode Island 02903, USA
| | - Xinrui Zhang
- Department of Molecular Biology, Massachusetts General Hospital, Boston, Massachusetts 02114, USA
| | - Peter Breen
- Department of Molecular Biology, Massachusetts General Hospital, Boston, Massachusetts 02114, USA
| | - Jonah Larkins-Ford
- Department of Molecular Biology, Massachusetts General Hospital, Boston, Massachusetts 02114, USA
| | - Eleftherios Mylonakis
- Division of Infectious Diseases, Rhode Island Hospital, Alpert Medical School of Brown University, Providence, Rhode Island 02903, USA
| | - Gary Ruvkun
- Department of Molecular Biology, Massachusetts General Hospital, Boston, Massachusetts 02114, USA
- Department of Genetics, Harvard Medical School, Boston, Massachusetts 02115, USA
| |
Collapse
|
707
|
Demir M, Lang S, Steffen HM. Nonalcoholic fatty liver disease - current status and future directions. J Dig Dis 2015; 16:541-57. [PMID: 26406351 DOI: 10.1111/1751-2980.12291] [Citation(s) in RCA: 66] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/27/2015] [Revised: 09/23/2015] [Accepted: 09/23/2015] [Indexed: 12/11/2022]
Abstract
Nonalcoholic fatty liver disease (NAFLD) has emerged as the most common chronic liver disease worldwide with a reported prevalence ranging 6-33%, depending on the studied populations. It encompasses a spectrum of liver manifestations ranging from simple steatosis (also known as nonalcoholic fatty liver, NAFL) to nonalcoholic steatohepatitis (NASH), fibrosis and cirrhosis, which may ultimately progress to hepatocellular carcinoma. NAFLD is strongly associated with the components of metabolic syndrome, mainly obesity and type 2 diabetes mellitus. NAFLD patients are at increased risk of liver-related as well as cardiovascular mortality. Current paradigm suggests a benign course for NAFL whereas NASH is considered to be the progressive phenotype. Although previously under-recognized accumulating evidence suggests that NAFL may also progress, suggesting a higher number of patients at risk than previously appreciated. Liver biopsy remains the gold standard for definitive diagnosis, but the majority of patients can be diagnosed accurately by noninvasive methods. Approved therapies for NAFLD are still lacking and lifestyle modifications aiming at weight loss remain the mainstay of NAFLD treatment. Intensive research could identify insulin resistance, lipotoxicity and dysbiosis of the gut microbiota as major pathophysiological mechanisms, leading to the development of promising targeted therapies which are currently investigated in clinical trials. In this review we summarized the current knowledge of NAFLD epidemiology, natural history, diagnosis, pathogenesis and treatment and considered future directions.
Collapse
Affiliation(s)
- Münevver Demir
- Clinic for Gastroenterology and Hepatology, University Hospital of Cologne, Cologne, Germany
| | - Sonja Lang
- Clinic for Gastroenterology and Hepatology, University Hospital of Cologne, Cologne, Germany
| | - Hans-Michael Steffen
- Clinic for Gastroenterology and Hepatology, University Hospital of Cologne, Cologne, Germany
| |
Collapse
|
708
|
Lourenco GF, Janitz M, Huang Y, Halliday GM. Long noncoding RNAs in TDP-43 and FUS/TLS-related frontotemporal lobar degeneration (FTLD). Neurobiol Dis 2015. [DOI: 10.1016/j.nbd.2015.07.011] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
|
709
|
Wang XX, Edelstein MH, Gafter U, Qiu L, Luo Y, Dobrinskikh E, Lucia S, Adorini L, D'Agati VD, Levi J, Rosenberg A, Kopp JB, Gius DR, Saleem MA, Levi M. G Protein-Coupled Bile Acid Receptor TGR5 Activation Inhibits Kidney Disease in Obesity and Diabetes. J Am Soc Nephrol 2015; 27:1362-78. [PMID: 26424786 DOI: 10.1681/asn.2014121271] [Citation(s) in RCA: 149] [Impact Index Per Article: 14.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2014] [Accepted: 08/12/2015] [Indexed: 12/31/2022] Open
Abstract
Obesity and diabetes mellitus are the leading causes of renal disease. In this study, we determined the regulation and role of the G protein-coupled bile acid receptor TGR5, previously shown to be regulated by high glucose and/or fatty acids, in obesity-related glomerulopathy (ORG) and diabetic nephropathy (DN). Treatment of diabetic db/db mice with the selective TGR5 agonist INT-777 decreased proteinuria, podocyte injury, mesangial expansion, fibrosis, and CD68 macrophage infiltration in the kidney. INT-777 also induced renal expression of master regulators of mitochondrial biogenesis, inhibitors of oxidative stress, and inducers of fatty acid β-oxidation, including sirtuin 1 (SIRT1), sirtuin 3 (SIRT3), and Nrf-1. Increased activity of SIRT3 was evidenced by normalization of the increased acetylation of mitochondrial superoxide dismutase 2 (SOD2) and isocitrate dehydrogenase 2 (IDH2) observed in untreated db/db mice. Accordingly, INT-777 decreased mitochondrial H2O2 generation and increased the activity of SOD2, which associated with decreased urinary levels of H2O2 and thiobarbituric acid reactive substances. Furthermore, INT-777 decreased renal lipid accumulation. INT-777 also prevented kidney disease in mice with diet-induced obesity. In human podocytes cultured with high glucose, INT-777 induced mitochondrial biogenesis, decreased oxidative stress, and increased fatty acid β-oxidation. Compared with normal kidney biopsy specimens, kidney specimens from patients with established ORG or DN expressed significantly less TGR5 mRNA, and levels inversely correlated with disease progression. Our results indicate that TGR5 activation induces mitochondrial biogenesis and prevents renal oxidative stress and lipid accumulation, establishing a role for TGR5 in inhibiting kidney disease in obesity and diabetes.
Collapse
Affiliation(s)
- Xiaoxin X Wang
- Department of Medicine, Division of Renal Diseases and Hypertension, University of Colorado Anschutz Medical Campus, Aurora, Colorado;
| | - Michal Herman Edelstein
- Rabin Medical Center, Department of Nephrology and Hypertension; Felsenstein Medical Research Center, Sackler School of Medicine, Tel Aviv University, Israel
| | - Uzi Gafter
- Rabin Medical Center, Department of Nephrology and Hypertension; Felsenstein Medical Research Center, Sackler School of Medicine, Tel Aviv University, Israel
| | - Liru Qiu
- Department of Medicine, Division of Renal Diseases and Hypertension, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Yuhuan Luo
- Department of Medicine, Division of Renal Diseases and Hypertension, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Evgenia Dobrinskikh
- Department of Medicine, Division of Renal Diseases and Hypertension, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Scott Lucia
- Department of Medicine, Division of Renal Diseases and Hypertension, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | | | - Vivette D D'Agati
- Department of Pathology, Columbia University, College of Physicians and Surgeons, New York, New York
| | - Jonathan Levi
- National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland
| | - Avi Rosenberg
- National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland
| | - Jeffrey B Kopp
- National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland
| | - David R Gius
- Northwestern University Feinberg School of Medicine, Chicago, Illinois; and
| | - Moin A Saleem
- University of Bristol, Southmead Hospital, Bristol, United Kingdom
| | - Moshe Levi
- Department of Medicine, Division of Renal Diseases and Hypertension, University of Colorado Anschutz Medical Campus, Aurora, Colorado;
| |
Collapse
|
710
|
Abente EJ, Subramanian M, Ramachandran V, Najafi-Shoushtari SH. MicroRNAs in obesity-associated disorders. Arch Biochem Biophys 2015; 589:108-19. [PMID: 26416722 DOI: 10.1016/j.abb.2015.09.018] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2015] [Revised: 09/17/2015] [Accepted: 09/18/2015] [Indexed: 02/08/2023]
Abstract
The emergence of a worldwide obesity epidemic has dramatically increased the prevalence of insulin resistance and metabolic syndrome, predisposing individuals to a greater risk for the development of non-alcoholic fatty liver disease, type II diabetes and atherosclerotic cardiovascular diseases. Current available pharmacological interventions combined with diet and exercise-based managements are still poorly effective for weight management, likely in part due to an incomplete understanding of regulatory mechanisms and pathways contributing to the systemic metabolic abnormalities under disturbed energy homeostasis. MicroRNAs, small non-coding RNAs that regulate posttranscriptional gene expression, have been increasingly described to influence shifts in metabolic pathways under various obesity-related disease settings. Here we review recent discoveries of the mechanistic role that microRNAs play in regulating metabolic functions in liver and adipose tissues involved in obesity associated disorders, and briefly discusses the potential candidates that are being pursued as viable therapeutic targets.
Collapse
Affiliation(s)
- Eugenio J Abente
- Department of Cell and Developmental Biology, Weill Cornell Medical College, Cornell University, New York 10021, NY, USA; Weill Cornell Medical College-Qatar, Qatar Foundation, Education City, P.O. Box 24144, Doha, Qatar
| | - Murugan Subramanian
- Department of Cell and Developmental Biology, Weill Cornell Medical College, Cornell University, New York 10021, NY, USA; Weill Cornell Medical College-Qatar, Qatar Foundation, Education City, P.O. Box 24144, Doha, Qatar
| | - Vimal Ramachandran
- Department of Cell and Developmental Biology, Weill Cornell Medical College, Cornell University, New York 10021, NY, USA; Weill Cornell Medical College-Qatar, Qatar Foundation, Education City, P.O. Box 24144, Doha, Qatar
| | - S Hani Najafi-Shoushtari
- Department of Cell and Developmental Biology, Weill Cornell Medical College, Cornell University, New York 10021, NY, USA; Weill Cornell Medical College-Qatar, Qatar Foundation, Education City, P.O. Box 24144, Doha, Qatar.
| |
Collapse
|
711
|
Cholestatic liver (dys)function during sepsis and other critical illnesses. Intensive Care Med 2015; 42:16-27. [DOI: 10.1007/s00134-015-4054-0] [Citation(s) in RCA: 83] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2015] [Accepted: 09/06/2015] [Indexed: 01/05/2023]
|
712
|
Fu ZD, Cui JY, Klaassen CD. The Role of Sirt1 in Bile Acid Regulation during Calorie Restriction in Mice. PLoS One 2015; 10:e0138307. [PMID: 26372644 PMCID: PMC4570809 DOI: 10.1371/journal.pone.0138307] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2014] [Accepted: 08/28/2015] [Indexed: 12/11/2022] Open
Abstract
Sirtuin 1 (Sirt1) is an NAD+-dependent protein deacetylase that is proposed to mediate many health-promoting effects of calorie restriction (CR). We recently reported that short-term CR increased the bile acid (BA) pool size in mice, likely due to increased BA synthesis in liver. Given the important role of Sirt1 in the regulation of glucose, lipid, as well as BA metabolism, we hypothesized that the CR-induced increase in BAs is Sirt1-dependent. To address this, the present study utilized genetically-modified mice that were Sirt1 loss of function (liver knockout, LKO) or Sirt1 gain of function (whole body-transgenic, TG). Three genotypes of mice (Sirt1-LKO, wild-type, and Sirt1-TG) were each randomly divided into ad libitum or 40% CR feeding for one month. BAs were extracted from various compartments of the enterohepatic circulation, followed by BA profiling by UPLC-MS/MS. CR increased the BA pool size and total BAs in serum, gallbladder, and small intestine. The CR-induced increase in BA pool size correlated with the tendency of increase in the expression of the rate-limiting BA-synthetic enzyme Cyp7a1. However, in contrast to the hypothesis, the CR-induced increase in BA pool size and Cyp7a1 expression was still observed with ablated expression of Sirt1 in liver, and completely suppressed with whole-body overexpression of Sirt1. Furthermore, in terms of BA composition, CR increased the ratio of 12α-hydroxylated BAs regardless of Sirt1 genotypes. In conclusion, the CR-induced alterations in BA pool size, BA profiles, and expression of BA-related genes do not appear to be dependent on Sirt1.
Collapse
Affiliation(s)
- Zidong Donna Fu
- Department of Pharmacology, Toxicology, and Therapeutics, University of Kansas Medical Center, Kansas City, KS, United States of America
- Department of Pharmacology, Harbin Medical University (the State-Province Key Laboratories of Biomedicine-Pharmaceutics of China), Harbin, Heilongjiang Province, People's Republic of China, 150081
| | - Julia Yue Cui
- Department of Environmental and Occupational Health Sciences, University of Washington, Seattle, WA, United States of America
| | - Curtis D. Klaassen
- Department of Internal Medicine, University of Kansas Medical Center, Kansas City, KS, United States of America
| |
Collapse
|
713
|
Fedorova OV, Zernetkina VI, Shilova VY, Grigorova YN, Juhasz O, Wei W, Marshall CA, Lakatta EG, Bagrov AY. Synthesis of an Endogenous Steroidal Na Pump Inhibitor Marinobufagenin, Implicated in Human Cardiovascular Diseases, Is Initiated by CYP27A1 via Bile Acid Pathway. ACTA ACUST UNITED AC 2015; 8:736-45. [PMID: 26374826 DOI: 10.1161/circgenetics.115.001217] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2014] [Accepted: 08/31/2015] [Indexed: 01/06/2023]
Abstract
BACKGROUND The bioactive steroid, marinobufagenin, is an endogenous Na/K-ATPase bufadienolide inhibitor that is synthesized by adrenocortical and placental cells. Marinobufagenin binding to Na/K-ATPase initiates profibrotic cell signaling, and heightened marinobufagenin levels are implicated in the pathogenesis of hypertension, preeclampsia, and chronic kidney disease. Steroids are derived from cholesterol through the traditional steroidogenesis pathway initiated by enzyme CYP11A1, and via the acidic bile acid pathway, which is controlled by enzyme CYP27A1. The mechanism of marinobufagenin biosynthesis in mammals, however, remains unknown. METHODS AND RESULTS Here, we show that post-transcriptional silencing of the CYP27A1 gene in human trophoblast and rat adrenocortical cells reduced the expression of CYP27A1 mRNA by 70%, reduced total bile acids 2-fold, and marinobufagenin levels by 67% when compared with nontreated cells or cells transfected with nontargeting siRNA. In contrast, silencing of the CYP11A1 gene did not affect marinobufagenin production in either cell culture, but suppressed production of progesterone 2-fold in human trophoblast cells and of corticosterone by 90% in rat adrenocortical cells when compared with cells transfected with nontargeting siRNA. In vivo, in a high-salt administration experiment, male and female Dahl salt-sensitive rats became hypertensive after 4 weeks on a high-NaCl diet, their plasma marinobufagenin levels doubled, and adrenocortical CYP27A1 mRNA and protein increased 1.6-fold and 2.0-fold. CONCLUSIONS Therefore, the endogenous steroidal Na/K-ATPase inhibitor, marinobufagenin, is synthesized in mammalian placenta and adrenal cortex from cholesterol through the novel acidic bile acid pathway. These findings will help to understand the role of marinobufagenin in highly prevalent human cardiovascular diseases.
Collapse
Affiliation(s)
- Olga V Fedorova
- From the Laboratory of Cardiovascular Science, National Institute on Aging, National Institutes of Health, Baltimore, MD.
| | - Valentina I Zernetkina
- From the Laboratory of Cardiovascular Science, National Institute on Aging, National Institutes of Health, Baltimore, MD
| | - Victoria Y Shilova
- From the Laboratory of Cardiovascular Science, National Institute on Aging, National Institutes of Health, Baltimore, MD
| | - Yulia N Grigorova
- From the Laboratory of Cardiovascular Science, National Institute on Aging, National Institutes of Health, Baltimore, MD
| | - Ondrej Juhasz
- From the Laboratory of Cardiovascular Science, National Institute on Aging, National Institutes of Health, Baltimore, MD
| | - Wen Wei
- From the Laboratory of Cardiovascular Science, National Institute on Aging, National Institutes of Health, Baltimore, MD
| | - Courtney A Marshall
- From the Laboratory of Cardiovascular Science, National Institute on Aging, National Institutes of Health, Baltimore, MD
| | - Edward G Lakatta
- From the Laboratory of Cardiovascular Science, National Institute on Aging, National Institutes of Health, Baltimore, MD
| | - Alexei Y Bagrov
- From the Laboratory of Cardiovascular Science, National Institute on Aging, National Institutes of Health, Baltimore, MD.
| |
Collapse
|
714
|
Liu K, Yan J, Sachar M, Zhang X, Guan M, Xie W, Ma X. A metabolomic perspective of griseofulvin-induced liver injury in mice. Biochem Pharmacol 2015; 98:493-501. [PMID: 26343413 DOI: 10.1016/j.bcp.2015.09.002] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2015] [Accepted: 09/02/2015] [Indexed: 01/09/2023]
Abstract
Griseofulvin (GSF) causes hepatic porphyria in mice, which mimics the liver injury associated with erythropoietic protoporphyria (EPP) in humans. The current study investigated the biochemical basis of GSF-induced liver injury in mice using a metabolimic approach. GSF treatment in mice resulted in significant accumulations of protoporphyrin IX (PPIX), N-methyl PPIX, bile acids, and glutathione (GSH) in the liver. Metabolomic analysis also revealed bioactivation pathways of GSF that contributed to the formation of GSF-PPIX, GSF-GSH and GSF-proline adducts. GSF-PPIX is the precursor of N-methyl PPIX. A six-fold increase of N-methyl PPIX was observed in the liver of mice after GSF treatment. N-methyl PPIX strongly inhibits ferrochelatase, the enzyme that converts PPIX to heme, and leads to PPIX accumulation. Excessive PPIX in the liver results in bile duct blockage and disturbs bile acid homeostasis. The accumulation of GSH in the liver was likely due to Nrf2-mediated upregulation of GSH synthesis. In summary, this study provides the biochemical basis of GSF-induced liver injury that can be used to understand the pathophysiology of EPP-associated liver injury in humans.
Collapse
Affiliation(s)
- Ke Liu
- Center for Pharmacogenetics, Department of Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Jiong Yan
- Center for Pharmacogenetics, Department of Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Madhav Sachar
- Center for Pharmacogenetics, Department of Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Xinju Zhang
- Department of Laboratory Medicine, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, China
| | - Ming Guan
- Department of Laboratory Medicine, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, China
| | - Wen Xie
- Center for Pharmacogenetics, Department of Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Xiaochao Ma
- Center for Pharmacogenetics, Department of Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh, Pittsburgh, Pennsylvania, USA.
| |
Collapse
|
715
|
Affiliation(s)
- Sawan Bopanna
- Department of Gastroenterology, All India Institute of Medical Sciences, New Delhi, India
| |
Collapse
|
716
|
Stinkens R, Goossens GH, Jocken JWE, Blaak EE. Targeting fatty acid metabolism to improve glucose metabolism. Obes Rev 2015; 16:715-57. [PMID: 26179344 DOI: 10.1111/obr.12298] [Citation(s) in RCA: 110] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/02/2015] [Revised: 04/23/2015] [Accepted: 05/10/2015] [Indexed: 12/15/2022]
Abstract
Disturbances in fatty acid metabolism in adipose tissue, liver, skeletal muscle, gut and pancreas play an important role in the development of insulin resistance, impaired glucose metabolism and type 2 diabetes mellitus. Alterations in diet composition may contribute to prevent and/or reverse these disturbances through modulation of fatty acid metabolism. Besides an increased fat mass, adipose tissue dysfunction, characterized by an altered capacity to store lipids and an altered secretion of adipokines, may result in lipid overflow, systemic inflammation and excessive lipid accumulation in non-adipose tissues like liver, skeletal muscle and the pancreas. These impairments together promote the development of impaired glucose metabolism, insulin resistance and type 2 diabetes mellitus. Furthermore, intrinsic functional impairments in either of these organs may contribute to lipotoxicity and insulin resistance. The present review provides an overview of fatty acid metabolism-related pathways in adipose tissue, liver, skeletal muscle, pancreas and gut, which can be targeted by diet or food components, thereby improving glucose metabolism.
Collapse
Affiliation(s)
- R Stinkens
- Department of Human Biology, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Center+, Maastricht, The Netherlands
| | - G H Goossens
- Department of Human Biology, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Center+, Maastricht, The Netherlands
| | - J W E Jocken
- Department of Human Biology, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Center+, Maastricht, The Netherlands
| | - E E Blaak
- Department of Human Biology, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Center+, Maastricht, The Netherlands
| |
Collapse
|
717
|
Masubuchi N, Sugihara M, Sugita T, Amano K, Nakano M, Matsuura T. Oxidative stress markers, secondary bile acids and sulfated bile acids classify the clinical liver injury type: Promising diagnostic biomarkers for cholestasis. Chem Biol Interact 2015; 255:83-91. [PMID: 26325587 DOI: 10.1016/j.cbi.2015.08.016] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2015] [Revised: 08/07/2015] [Accepted: 08/25/2015] [Indexed: 11/29/2022]
Abstract
Clinicians sometimes encounter difficulty in choosing a therapeutic strategy due to the uncertainty regarding the type of liver injury. In particular, cholestasis is difficult to diagnose by conventional markers at an early stage of disease. The aim of this study was to identify promising biomarkers for distinguishing the symptom-based types of liver injury (e.g. hepatocellular injury, cholestasis), which was derived from a rigorously statistical perspective. The associations between diagnostic biomarkers (e.g. bile acid components, oxidative stress markers and liver fibrosis markers) and the liver injury types were assessed by a multiple logistic regression analysis using 304 blood samples from patients with liver disease. As a result, reductions in the lithocholic acid (LCA) and deoxycholic acid (DCA) levels, and elevation of the serum sulfated bile acid (SSBA), liver fibrosis marker IV collagen (type IV collagen), hyaluronic acid (HA) and reactive oxygen species (ROS) levels were all significantly associated with cholestasis. On the other hand, elevations in the LCA and type IV collagen levels, and a reduction in the ursodeoxy cholic acid (UDCA) level, were significantly associated with hepatocellular injury. The receiver operating characteristic (ROC) analyses showed that the largest area under the ROC curve (AUC) was found for ROS, followed by DCA, HA, LCA, SSBA and type IV collagen in the cholestatic-type cases. These results indicated that ROS, the secondary bile acid levels such as DCA and LCA, and SSBA are promising biomarkers for cholestasis and for classifying the type of liver injuries. This comprehensive approach will allow for an accurate diagnosis, which will facilitate the selection of an appropriate therapy at the onset of disease.
Collapse
Affiliation(s)
- Noriko Masubuchi
- Drug Metabolism & Pharmacokinetics Research Laboratories, Daiichi Sankyo Co., Ltd., Tokyo, Japan.
| | - Masahiro Sugihara
- Clinical Data & Biostatistics Department, Daiichi Sankyo Co., Ltd., Tokyo, Japan
| | - Tomonori Sugita
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Jikei University School of Medicine, Tokyo, Japan
| | - Katsushi Amano
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Jikei University School of Medicine, Tokyo, Japan
| | - Masanori Nakano
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Jikei University School of Medicine, Tokyo, Japan
| | - Tomokazu Matsuura
- Department of Laboratory Medicine, Jikei University School of Medicine, Tokyo, Japan
| |
Collapse
|
718
|
Briere DA, Ruan X, Cheng CC, Siesky AM, Fitch TE, Dominguez C, Sanfeliciano SG, Montero C, Suen CS, Xu Y, Coskun T, Michael MD. Novel Small Molecule Agonist of TGR5 Possesses Anti-Diabetic Effects but Causes Gallbladder Filling in Mice. PLoS One 2015; 10:e0136873. [PMID: 26312995 PMCID: PMC4551797 DOI: 10.1371/journal.pone.0136873] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2015] [Accepted: 08/09/2015] [Indexed: 12/25/2022] Open
Abstract
Activation of TGR5 via bile acids or bile acid analogs leads to the release of glucagon-like peptide-1 (GLP-1) from intestine, increases energy expenditure in brown adipose tissue, and increases gallbladder filling with bile. Here, we present compound 18, a non-bile acid agonist of TGR5 that demonstrates robust GLP-1 secretion in a mouse enteroendocrine cell line yet weak GLP-1 secretion in a human enteroendocrine cell line. Acute administration of compound 18 to mice increased GLP-1 and peptide YY (PYY) secretion, leading to a lowering of the glucose excursion in an oral glucose tolerance test (OGTT), while chronic administration led to weight loss. In addition, compound 18 showed a dose-dependent increase in gallbladder filling. Lastly, compound 18 failed to show similar pharmacological effects on GLP-1, PYY, and gallbladder filling in Tgr5 knockout mice. Together, these results demonstrate that compound 18 is a mouse-selective TGR5 agonist that induces GLP-1 and PYY secretion, and lowers the glucose excursion in an OGTT, but only at doses that simultaneously induce gallbladder filling. Overall, these data highlight the benefits and potential risks of using TGR5 agonists to treat diabetes and metabolic diseases.
Collapse
Affiliation(s)
- Daniel A. Briere
- Lilly Research Laboratories, Eli Lilly and Co., Indianapolis, Indiana, United States of America
| | - Xiaoping Ruan
- Lilly Research Laboratories, Eli Lilly and Co., Indianapolis, Indiana, United States of America
| | - Christine C. Cheng
- Lilly Research Laboratories, Eli Lilly and Co., Indianapolis, Indiana, United States of America
| | - Angela M. Siesky
- Lilly Research Laboratories, Eli Lilly and Co., Indianapolis, Indiana, United States of America
| | - Thomas E. Fitch
- Lilly Research Laboratories, Eli Lilly and Co., Indianapolis, Indiana, United States of America
| | - Carmen Dominguez
- Centro de Investigación, Eli Lilly and Company, Alcobendas, Spain
| | | | - Carlos Montero
- Centro de Investigación, Eli Lilly and Company, Alcobendas, Spain
| | - Chen S. Suen
- Lilly Research Laboratories, Eli Lilly and Co., Indianapolis, Indiana, United States of America
| | - Yanping Xu
- Lilly Research Laboratories, Eli Lilly and Co., Indianapolis, Indiana, United States of America
| | - Tamer Coskun
- Lilly Research Laboratories, Eli Lilly and Co., Indianapolis, Indiana, United States of America
| | - M. Dodson Michael
- Lilly Research Laboratories, Eli Lilly and Co., Indianapolis, Indiana, United States of America
- * E-mail:
| |
Collapse
|
719
|
Sawitza I, Kordes C, Götze S, Herebian D, Häussinger D. Bile acids induce hepatic differentiation of mesenchymal stem cells. Sci Rep 2015; 5:13320. [PMID: 26304833 PMCID: PMC4548444 DOI: 10.1038/srep13320] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2015] [Accepted: 07/23/2015] [Indexed: 12/25/2022] Open
Abstract
Mesenchymal stem cells (MSC) have the potential to differentiate into multiple cell lineages and their therapeutic potential has become obvious. In the liver, MSC are represented by stellate cells which have the potential to differentiate into hepatocytes after stimulation with growth factors. Since bile acids can promote liver regeneration, their influence on liver-resident and bone marrow-derived MSC was investigated. Physiological concentrations of bile acids such as tauroursodeoxycholic acid were able to initiate hepatic differentiation of MSC via the farnesoid X receptor and transmembrane G-protein-coupled bile acid receptor 5 as investigated with knockout mice. Notch, hedgehog, transforming growth factor-β/bone morphogenic protein family and non-canonical Wnt signalling were also essential for bile acid-mediated differentiation, whereas β-catenin-dependent Wnt signalling was able to attenuate this process. Our findings reveal bile acid-mediated signalling as an alternative way to induce hepatic differentiaion of stem cells and highlight bile acids as important signalling molecules during liver regeneration.
Collapse
Affiliation(s)
- Iris Sawitza
- Clinic of Gastroenterology, Hepatology and Infectious Diseases
| | - Claus Kordes
- Clinic of Gastroenterology, Hepatology and Infectious Diseases
| | - Silke Götze
- Clinic of Gastroenterology, Hepatology and Infectious Diseases
| | - Diran Herebian
- Department of General Pediatrics, Neonatology and Pediatric Cardiology, Heinrich Heine University, Moorenstraße 5, 40225 Düsseldorf, Germany
| | | |
Collapse
|
720
|
Billeter AT, Fischer L, Wekerle AL, Senft J, Müller-Stich B. Malabsorption as a Therapeutic Approach in Bariatric Surgery. VISZERALMEDIZIN 2015; 30:198-204. [PMID: 26288594 PMCID: PMC4513825 DOI: 10.1159/000363480] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Background The increasing prevalence of obese patients will lead to a more frequent use of bariatric procedures in the future. Compared to conservative medical therapy, bariatric procedures achieve greater weight loss and superior control of comorbidities, resulting in improved overall mortality. Methods A search for current literature regarding mechanisms, indications, and outcomes of bariatric surgery was performed. Results In order to care for patients after bariatric surgery properly, it is important to understand its mechanisms of action and effects on gastrointestinal physiology. Recent investigations indicate that the beneficial effects of bariatric procedures are much more complex than simply limiting food intake or an associated malabsorption. Changes in gastrointestinal hormone secretion, energy expenditure, intestinal bacterial colonization, bile acid metabolism, and epigenetic modifications resulting in altered gene expression are likely responsible for the majority of the beneficial effects of bariatric surgery. Malabsorptive bariatric procedures divert the flow of bile and pancreatic enzymes from food and therefore limit the digestion and absorption of nutrients, resulting in reduced calorie intake and subsequent weight loss. Essential micronutrients such as vitamins and trace elements are also absorbed to a lesser extent, potentially leading to severe side effects. Conclusion To prevent malnutrition, dietary supplementation and regular control of micronutrient levels are mandatory for patients undergoing malabsorptive bariatric procedures, in whom the fat-soluble vitamins A and D are commonly deficient.
Collapse
Affiliation(s)
- Adrian T Billeter
- Department of General, Visceral, and Transplantation Surgery, University of Heidelberg, Germany
| | - Lars Fischer
- Department of General, Visceral, and Transplantation Surgery, University of Heidelberg, Germany
| | - Anna-Laura Wekerle
- Department of General, Visceral, and Transplantation Surgery, University of Heidelberg, Germany
| | - Jonas Senft
- Department of General, Visceral, and Transplantation Surgery, University of Heidelberg, Germany
| | - Beat Müller-Stich
- Department of General, Visceral, and Transplantation Surgery, University of Heidelberg, Germany
| |
Collapse
|
721
|
Zou A, Lehn S, Magee N, Zhang Y. New Insights into Orphan Nuclear Receptor SHP in Liver Cancer. NUCLEAR RECEPTOR RESEARCH 2015; 2. [PMID: 26504773 PMCID: PMC4618403 DOI: 10.11131/2015/101162] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Small heterodimer partner (SHP; NR0B2) is a unique orphan nuclear receptor (NR) that contains a putative ligand-binding domain but lacks a DNA-binding domain. SHP is a transcriptional corepressor affecting diverse metabolic processes including bile acid synthesis, cholesterol and lipid metabolism, glucose and energy homeostasis, and reproductive biology via interaction with multiple NRs and transcriptional factors (TFs). Hepatocellular carcinoma (HCC) is one of the most deadly human cancers worldwide with few therapeutic options and poor prognosis. Recently, it is becoming clear that SHP plays an antitumor role in the development of liver cancer. In this review, we summarize the most recent findings regarding the new SHP interaction partners, new structural insights into SHP’s gene repressing activity, and SHP protein posttranslational modifications by bile acids. We also discuss the pleiotropic role of SHP in regulating cell proliferation, apoptosis, DNA methylation, and inflammation that are related to antitumor role of SHP in HCC. Improving our understanding of SHP’s antitumor role in the development of liver cancer will provide new insights into developing novel treatments or prevention strategies. Future research will focus on developing more efficacious and specific synthetic SHP ligands for pharmaceutical applications in liver cancer and several metabolic diseases such as hypercholesterolemia, obesity, diabetes, and fatty liver disease.
Collapse
Affiliation(s)
- An Zou
- Department of Pharmacology, Toxicology & Therapeutics, University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Sarah Lehn
- Department of Pharmacology, Toxicology & Therapeutics, University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Nancy Magee
- Department of Pharmacology, Toxicology & Therapeutics, University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Yuxia Zhang
- Department of Pharmacology, Toxicology & Therapeutics, University of Kansas Medical Center, Kansas City, KS 66160, USA
| |
Collapse
|
722
|
Tran CD, Grice DM, Wade B, Kerr CA, Bauer DC, Li D, Hannan GN. Gut permeability, its interaction with gut microflora and effects on metabolic health are mediated by the lymphatics system, liver and bile acid. Future Microbiol 2015; 10:1339-53. [PMID: 26234760 DOI: 10.2217/fmb.15.54] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
There is evidence to link obesity (and metabolic syndrome) with alterations in gut permeability and microbiota. The underlying mechanisms have been questioned and have prompted this review. We propose that the gut barrier function is a primary driver in maintaining metabolic health with poor health being linked to ‘gut leakiness'. This review will highlight changes in intestinal permeability and how it may change gut microflora and subsequently affect metabolic health by influencing the functioning of major bodily organs/organ systems: the lymphatic system, liver and pancreas. We also discuss the likelihood that metabolic syndrome undergoes a cyclic worsening facilitated by an increase in intestinal permeability leading to gut dysbiosis, culminating in ongoing poor health leading to further exacerbated gut leakiness.
Collapse
Affiliation(s)
- Cuong D Tran
- CSIRO Food & Nutrition Flagship, Adelaide, SA 5000, Australia
| | - Desma M Grice
- CSIRO Food & Nutrition Flagship, North Ryde, NSW 2113, Australia
| | - Ben Wade
- CSIRO Biosecurity Flagship, Geelong, VIC 3219, Australia
| | - Caroline A Kerr
- CSIRO Food & Nutrition Flagship, North Ryde, NSW 2113, Australia
| | - Denis C Bauer
- CSIRO Digital Productivity & Services Flagship, North Ryde, NSW 1670, Australia
| | - Dongmei Li
- CSIRO Food & Nutrition Flagship, North Ryde, NSW 2113, Australia
| | - Garry N Hannan
- CSIRO Food & Nutrition Flagship, North Ryde, NSW 2113, Australia
| |
Collapse
|
723
|
Lim DW, Wales PW, Mi S, Yap JYK, Curtis JM, Mager DR, Mazurak VC, Wizzard PR, Sigalet DL, Turner JM. Glucagon-Like Peptide-2 Alters Bile Acid Metabolism in Parenteral Nutrition--Associated Liver Disease. JPEN J Parenter Enteral Nutr 2015. [PMID: 26220199 DOI: 10.1177/0148607115595596] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
BACKGROUND We aim to study the mechanisms underlying our previous finding that exogenous glucagon-like peptide-2 (GLP-2) treatment in a preclinical model of neonatal parenteral nutrition-associated liver disease (PNALD) improves cholestasis. METHODS Neonatal piglets received 17 days of parenteral nutrition (PN) therapy and either saline control (PN/Saline n = 8) or GLP-2 treatment at 11 nmol/kg/d (PN/GLP-2, n = 7). At terminal laparotomy, bile and liver samples were collected. The relative gene expression of enzymes involved in bile acid synthesis, regulation, and transport was measured in liver by reverse-transcriptase quantitative polymerase chain reaction. Bile acid composition in bile was determined using tandem mass spectrometry. Data were analyzed using 1-way analysis of variance (ANOVA) or Kruskal-Wallis ANOVA. RESULTS GLP-2 increased the expression of bile acid export genes: multidrug resistance-associated proteins 2 (MRP2) (P = .002) and 3 (MRP3) (P = .037) over saline control. GLP-2 increased expression of Farnesoid X receptor (FXR) (P < .001) and CYP7A1 (cytochrome P450, family 7, subfamily A, polypeptide 1) (P = .03). GLP-2 treatment was associated with decreased concentrations of taurohyocholic acid and conjugates of toxic lithocholic acid (P < .01). GLP-2 treatment increased the liver bile acid content. CONCLUSIONS GLP-2 treatment was associated with alterations in the hepatic expression of genes involved in bile acid metabolism. The transcriptomic results indicate the mechanisms at the transcriptional level acting to regulate bile acid synthesis and increase bile acid export. Differences in bile acid profiles further support a beneficial role for GLP-2 therapy in PNALD.
Collapse
Affiliation(s)
- David W Lim
- Department of Surgery, University of Alberta, Edmonton, Alberta, Canada
| | - Paul W Wales
- Department of Surgery, University of Alberta, Edmonton, Alberta, Canada Department of Pediatrics, University of Alberta, Edmonton, Alberta, Canada Department of Surgery, Hospital for Sick Children & University of Toronto, Toronto, Ontario, Canada
| | - Si Mi
- Department of Agricultural, Food and Nutritional Science, University of Alberta, Edmonton, Alberta, Canada
| | - Jason Y K Yap
- Department of Pediatrics, University of Alberta, Edmonton, Alberta, Canada
| | - Jonathan M Curtis
- Department of Agricultural, Food and Nutritional Science, University of Alberta, Edmonton, Alberta, Canada
| | - Diana R Mager
- Department of Agricultural, Food and Nutritional Science, University of Alberta, Edmonton, Alberta, Canada
| | - Vera C Mazurak
- Department of Agricultural, Food and Nutritional Science, University of Alberta, Edmonton, Alberta, Canada
| | - Pamela R Wizzard
- Department of Pediatrics, University of Alberta, Edmonton, Alberta, Canada
| | - David L Sigalet
- Department of Surgery, University of Calgary, Calgary, Alberta, Canada
| | - Justine M Turner
- Department of Pediatrics, University of Alberta, Edmonton, Alberta, Canada Department of Agricultural, Food and Nutritional Science, University of Alberta, Edmonton, Alberta, Canada
| |
Collapse
|
724
|
Devlin AS, Fischbach MA. A biosynthetic pathway for a prominent class of microbiota-derived bile acids. Nat Chem Biol 2015; 11:685-90. [PMID: 26192599 PMCID: PMC4543561 DOI: 10.1038/nchembio.1864] [Citation(s) in RCA: 299] [Impact Index Per Article: 29.9] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2014] [Accepted: 05/27/2015] [Indexed: 12/26/2022]
Abstract
The gut bile acid pool is millimolar in concentration, varies widely in composition among individuals, and is linked to metabolic disease and cancer. Although these molecules derive almost exclusively from the microbiota, remarkably little is known about which bacterial species and genes are responsible for their biosynthesis. Here, we report a biosynthetic pathway for the second most abundant class in the gut, iso (3β-hydroxy) bile acids, whose levels exceed 300 µM in some humans and are absent in others. We show, for the first time, that iso bile acids are produced by Ruminococcus gnavus, a far more abundant commensal than previously known producers; and that the iso bile acid pathway detoxifies deoxycholic acid, favoring the growth of the keystone genus Bacteroides. By revealing the biosynthetic genes for an abundant class of bile acids, our work sets the stage for predicting and rationally altering the composition of the bile acid pool.
Collapse
Affiliation(s)
- A Sloan Devlin
- 1] Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, San Francisco, California, USA. [2] California Institute for Quantitative Biosciences, University of California, San Francisco, San Francisco, California, USA
| | - Michael A Fischbach
- 1] Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, San Francisco, California, USA. [2] California Institute for Quantitative Biosciences, University of California, San Francisco, San Francisco, California, USA
| |
Collapse
|
725
|
Poša M, Bjedov S, Škorić D, Sakač M. Micellization parameters (number average, aggregation number and critical micellar concentration) of bile salt 3 and 7 ethylidene derivatives: Role of the steroidal skeleton II. Biochim Biophys Acta Gen Subj 2015; 1850:1345-53. [DOI: 10.1016/j.bbagen.2015.03.010] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2014] [Revised: 03/23/2015] [Accepted: 03/24/2015] [Indexed: 01/22/2023]
|
726
|
Pan X, Lee YK, Jeong H. Farnesoid X Receptor Agonist Represses Cytochrome P450 2D6 Expression by Upregulating Small Heterodimer Partner. Drug Metab Dispos 2015; 43:1002-7. [PMID: 25926433 PMCID: PMC4468439 DOI: 10.1124/dmd.115.064758] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2015] [Accepted: 04/29/2015] [Indexed: 01/05/2023] Open
Abstract
Cytochrome P450 2D6 (CYP2D6) is a major drug-metabolizing enzyme responsible for eliminating approximately 20% of marketed drugs. Studies have shown that differential transcriptional regulation of CYP2D6 may contribute to large interindividual variability in CYP2D6-mediated drug metabolism. However, the factors governing CYP2D6 transcription are largely unknown. We previously demonstrated small heterodimer partner (SHP) as a novel transcriptional repressor of CYP2D6 expression. SHP is a representative target gene of the farnesoid X receptor (FXR). The objective of this study is to investigate whether an agonist of FXR, 3-(2,6-dichlorophenyl)-4-(3'-carboxy-2-chlorostilben-4-yl)oxymethyl-5-isopropylisoxazole (GW4064), alters CYP2D6 expression and activity. In CYP2D6-humanized transgenic mice, GW4064 decreased hepatic CYP2D6 expression and activity (by 2-fold) while increasing SHP expression (by 2-fold) and SHP recruitment to the CYP2D6 promoter. CYP2D6 repression by GW4064 was abrogated in Shp(-/-);CYP2D6 mice, indicating a critical role of SHP in CYP2D6 regulation by GW4064. Also, GW4064 decreased CYP2D6 expression (by 2-fold) in primary human hepatocytes, suggesting that the results obtained in CYP2D6-humanized transgenic mice can be translated to humans. This proof of concept study provides evidence for CYP2D6 regulation by an inducer of SHP expression, namely, the FXR agonist GW4064.
Collapse
Affiliation(s)
- Xian Pan
- Departments of Pharmacy Practice (H.J.) and Biopharmaceutical Sciences (X.P., H.J.), College of Pharmacy, University of Illinois at Chicago, Chicago, Illinois; and Department of Integrative Medical Sciences, Northeast Ohio Medical University, Rootstown, Ohio (Y.K.L.)
| | - Yoon-Kwang Lee
- Departments of Pharmacy Practice (H.J.) and Biopharmaceutical Sciences (X.P., H.J.), College of Pharmacy, University of Illinois at Chicago, Chicago, Illinois; and Department of Integrative Medical Sciences, Northeast Ohio Medical University, Rootstown, Ohio (Y.K.L.)
| | - Hyunyoung Jeong
- Departments of Pharmacy Practice (H.J.) and Biopharmaceutical Sciences (X.P., H.J.), College of Pharmacy, University of Illinois at Chicago, Chicago, Illinois; and Department of Integrative Medical Sciences, Northeast Ohio Medical University, Rootstown, Ohio (Y.K.L.)
| |
Collapse
|
727
|
Abstract
TGR5 (Takeda G-protein-coupled receptor 5) [also known as GPBAR1 (G-protein-coupled bile acid receptor 1), M-BAR (membrane-type receptor for bile acids) or GPR131 (G-protein-coupled receptor 131)] is a G-protein-coupled receptor that was discovered as a bile acid receptor. TGR5 has specific roles in several tissues, among which are the regulation of energy expenditure, GLP-1 (glucagon-like peptide 1) secretion and gall bladder filling. An accumulating body of evidence now demonstrates that TGR5 also acts in a number of processes important in inflammation. Most striking in this context are several observations that TGR5 signalling curbs the inflammatory response of macrophages via interfering with NF-κB (nuclear factor κB) activity. In line with this, recent animal studies also suggest that TGR5 could be exploited as a potential target for intervention in a number of inflammation-driven diseases, including atherosclerosis. In the present paper, I review our current understanding of TGR5 with a strong focus on its potential as target for intervention in inflammation-driven diseases.
Collapse
|
728
|
Pereira K, Salsamendi J, Casillas J. The Global Nonalcoholic Fatty Liver Disease Epidemic: What a Radiologist Needs to Know. J Clin Imaging Sci 2015; 5:32. [PMID: 26167390 PMCID: PMC4485197 DOI: 10.4103/2156-7514.157860] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2015] [Accepted: 04/26/2015] [Indexed: 01/10/2023] Open
Abstract
Nonalcoholic fatty liver disease (NAFLD) represents a spectrum of disorders from a benign steatosis to hepatocellular carcinoma (HCC). Metabolic syndrome, mainly obesity, plays an important role, both as an independent risk factor and in the pathogenesis of NAFLD. With the progressive epidemics of obesity and diabetes mellitus, the prevalence of NAFLD and its associated complications is expected to increase dramatically. Therapeutic strategies for treating NAFLD and metabolic syndrome, particularly obesity, are continuously being refined. Their goal is the prevention of NAFLD by the management of risk factors, prevention of progression of the disease, as well as management of complications, ultimately preventing morbidity and mortality. Optimal management of NAFLD and metabolic syndrome requires a multidisciplinary collaboration between the government as well as the health system including the nutritionist, primary care physician, radiologist, hepatologist, oncologist, and transplant surgeon. An awareness of the clinical presentation, risk factors, pathogenesis, diagnosis, and management is of paramount importance to a radiologist, both from the clinical perspective as well as from the imaging standpoint. With expertise in imaging modalities as well as minimally invasive percutaneous endovascular therapies, radiologists play an essential role in the comprehensive management, which is highlighted in this article, with cases from our practice. We also briefly discuss transarterial embolization of the left gastric artery (LGA), a novel method that promises to have an enormous potential in the minimally invasive management of obesity, with details of a case from our practice.
Collapse
Affiliation(s)
- Keith Pereira
- Department of Interventional Radiology, Jackson Memorial Hospital, University of Miami Hospital, Miami, Florida, USA
| | - Jason Salsamendi
- Department of Interventional Radiology, Jackson Memorial Hospital, University of Miami Hospital, Miami, Florida, USA
| | - Javier Casillas
- Department of Diagnostic Radiology (Body Imaging), Jackson Memorial Hospital, University of Miami Hospital, Miami, Florida, USA
| |
Collapse
|
729
|
González-Peña D, Dudzik D, Colina-Coca C, de Ancos B, García A, Barbas C, Sánchez-Moreno C. Multiplatform metabolomic fingerprinting as a tool for understanding hypercholesterolemia in Wistar rats. Eur J Nutr 2015; 55:997-1010. [DOI: 10.1007/s00394-015-0914-1] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2014] [Accepted: 04/25/2015] [Indexed: 02/08/2023]
|
730
|
Olesen NE, Westh P, Holm R. Determination of thermodynamic potentials and the aggregation number for micelles with the mass-action model by isothermal titration calorimetry: A case study on bile salts. J Colloid Interface Sci 2015; 453:79-89. [PMID: 25978555 DOI: 10.1016/j.jcis.2015.03.069] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2015] [Revised: 03/28/2015] [Accepted: 03/30/2015] [Indexed: 01/04/2023]
Abstract
The aggregation number (n), thermodynamic potentials (ΔG, ΔH, ΔS) and critical micelle concentration (CMC) for 6 natural bile salts were determined on the basis of both original and previously published isothermal titration calorimetry (ITC) data. Different procedures to estimate parameters of micelles with ITC were compared to a mass-action model (MAM) of reaction type: n⋅S⇌Mn. This analysis can provide guidelines for future ITC studies of systems behaving in accordance with this model such as micelles and proteins that undergo self-association to oligomers. Micelles with small aggregation numbers, as those of bile salts, are interesting because such small aggregates cannot be characterized as a separate macroscopic phase and the widely applied pseudo-phase model (PPM) is inaccurate. In the present work it was demonstrated that the aggregation number of micelles was constant at low concentrations enabling determination of the thermodynamic potentials by the MAM. A correlation between the aggregation number and the heat capacity was found, which implies that the dehydrated surface area of bile salts increases with the aggregation number. This is in accordance with Tanford's principles of opposing forces where neighbouring molecules in the aggregate are better able to shield from the surrounding hydrophilic environment when the aggregation number increases.
Collapse
Affiliation(s)
- Niels Erik Olesen
- Biologics and Pharmaceutical Science, H. Lundbeck A/S, Ottiliavej 9, DK-2500 Valby, Denmark; NSM, Research Unit for Functional Biomaterials, Roskilde University, Universitetsvej 1, DK-4000 Roskilde, Denmark
| | - Peter Westh
- NSM, Research Unit for Functional Biomaterials, Roskilde University, Universitetsvej 1, DK-4000 Roskilde, Denmark
| | - René Holm
- Biologics and Pharmaceutical Science, H. Lundbeck A/S, Ottiliavej 9, DK-2500 Valby, Denmark.
| |
Collapse
|
731
|
Zhang S, Pan X, Jeong H. GW4064, an agonist of farnesoid X receptor, represses CYP3A4 expression in human hepatocytes by inducing small heterodimer partner expression. Drug Metab Dispos 2015; 43:743-8. [PMID: 25725071 PMCID: PMC4407707 DOI: 10.1124/dmd.114.062836] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2014] [Accepted: 02/27/2015] [Indexed: 12/20/2022] Open
Abstract
Farnesoid X receptor (FXR) functions as a regulator of bile acid and lipid homeostasis and is recognized as a promising therapeutic target for metabolic diseases. The biologic function of FXR is mediated in part by a small heterodimer partner (SHP); ligand-activated FXR enhances SHP expression, and SHP in turn represses the activity of multiple transcription factors. This study aimed to investigate the effect of FXR activation on expression of the major drug-metabolizing enzyme CYP3A4. The effects of 3-(2,6-dichlorophenyl)-4-(3'-carboxy-2-chlorostilben-4-yl)oxymethyl-5-isopropylisoxazole (GW4064), a synthetic agonist of FXR, on the expression and activity of CYP3A4 were examined in primary human hepatocytes by using quantitative real-time polymerase chain reaction and S9 phenotyping. In human hepatocytes, treatment of GW4064 (1 μM) for 48 hours resulted in a 75% decrease in CYP3A4 mRNA expression and a 25% decrease in CYP3A4 activity, accompanied by ∼3-fold increase in SHP mRNA expression. In HepG2 cells, SHP repressed transactivation of CYP3A4 promoter by pregnane X receptor (PXR), constitutive androstane receptor (CAR), and glucocorticoid receptor. Interestingly, GW4064 did not repress expression of CYP2B6, another target gene of PXR and CAR; GW4064 enhanced CYP2B6 promoter activity. In conclusion, GW4064 represses CYP3A4 expression in human hepatocytes, potentially through upregulation of SHP expression and subsequent repression of CYP3A4 promoter activity. Clinically significant drug-drug interaction involving FXR agonists and CYP3A4 substrates may occur.
Collapse
Affiliation(s)
- Shu Zhang
- Department of Pharmacy Practice (H.J.) and Department of Biopharmaceutical Sciences (S.Z., X.P., H.J.), College of Pharmacy, University of Illinois at Chicago, Chicago, Illinois
| | - Xian Pan
- Department of Pharmacy Practice (H.J.) and Department of Biopharmaceutical Sciences (S.Z., X.P., H.J.), College of Pharmacy, University of Illinois at Chicago, Chicago, Illinois
| | - Hyunyoung Jeong
- Department of Pharmacy Practice (H.J.) and Department of Biopharmaceutical Sciences (S.Z., X.P., H.J.), College of Pharmacy, University of Illinois at Chicago, Chicago, Illinois
| |
Collapse
|
732
|
Nagahashi M, Takabe K, Liu R, Peng K, Wang X, Wang Y, Hait NC, Wang X, Allegood JC, Yamada A, Aoyagi T, Liang J, Pandak WM, Spiegel S, Hylemon PB, Zhou H. Conjugated bile acid-activated S1P receptor 2 is a key regulator of sphingosine kinase 2 and hepatic gene expression. Hepatology 2015; 61:1216-26. [PMID: 25363242 PMCID: PMC4376566 DOI: 10.1002/hep.27592] [Citation(s) in RCA: 162] [Impact Index Per Article: 16.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/23/2014] [Accepted: 10/26/2014] [Indexed: 12/17/2022]
Abstract
UNLABELLED Bile acids are important hormones during the feed/fast cycle, allowing the liver to coordinately regulate nutrient metabolism. How they accomplish this has not been fully elucidated. Conjugated bile acids activate both the ERK1/2 and AKT signaling pathways via sphingosine 1-phosphate receptor 2 (S1PR2) in rodent hepatocytes and in vivo. Here, we report that feeding mice a high-fat diet, infusion of taurocholate into the chronic bile fistula rat, or overexpression of the gene encoding S1PR2 in mouse hepatocytes significantly upregulated hepatic sphingosine kinase 2 (SphK2) but not SphK1. Key genes encoding nuclear receptors/enzymes involved in nutrient metabolism were significantly downregulated in livers of S1PR2(-/-) and SphK2(-/-) mice. In contrast, overexpression of the gene encoding S1PR2 in primary mouse hepatocytes differentially increased SphK2, but not SphK1, and mRNA levels of key genes involved in nutrient metabolism. Nuclear levels of sphingosine-1-phosphate, an endogenous inhibitor of histone deacetylases 1 and 2, as well as the acetylation of histones H3K9, H4K5, and H2BK12 were significantly decreased in hepatocytes prepared from S1PR2(-/-) and SphK2(-/-) mice. CONCLUSION Both S1PR2(-/-) and SphK2(-/-) mice rapidly developed fatty livers on a high-fat diet, suggesting the importance of conjugated bile acids, S1PR2, and SphK2 in regulating hepatic lipid metabolism.
Collapse
Affiliation(s)
- Masayuki Nagahashi
- Department of Surgery, Medical College of Virginia Campus, Virginia Commonwealth University, Richmond, Virginia 23298,Department of Biochemistry and Molecular Biology, Medical College of Virginia Campus, Virginia Commonwealth University, Richmond, Virginia 23298,Division of Digestive and General Surgery, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan, 951-8510
| | - Kazuaki Takabe
- Department of Surgery, Medical College of Virginia Campus, Virginia Commonwealth University, Richmond, Virginia 23298,Department of Biochemistry and Molecular Biology, Medical College of Virginia Campus, Virginia Commonwealth University, Richmond, Virginia 23298,Address correspondence to: Huiping Zhou, Ph.D, Department of Microbiology & Immunology, Virginia Commonwealth University, P.O. Box 980678, Richmond, VA 23298-0678, Tel: 804-828-6817; Fax: 804-828-0676, Or Kazuaki Takabbe, M.D., Ph.D., FACS, Department of Surgery, VCU, P.O. Box 980011, Richmond, VA 23298-0011, Tel. 804-828-9322, Fax. 804-828-4809, Or Phillip B. Hylemon, Ph.D., Department of Microbiology and Immunology, Medical College of Virginia Campus-VCU, PO Box 908678, Richmond, VA 23298-0678, Tel: (804) 347-1752; Fax. (804) 828-0676,
| | - Runping Liu
- Department of Microbiology and Immunology, Medical College of Virginia Campus, Virginia Commonwealth University, Richmond, Virginia 23298,McGuire Veterans Affairs Medical Center, Medical College of Virginia Campus, Virginia Commonwealth University, Richmond, Virginia 23298
| | - Kesong Peng
- Department of Microbiology and Immunology, Medical College of Virginia Campus, Virginia Commonwealth University, Richmond, Virginia 23298
| | - Xiang Wang
- Department of Microbiology and Immunology, Medical College of Virginia Campus, Virginia Commonwealth University, Richmond, Virginia 23298
| | - Yun Wang
- Department of Microbiology and Immunology, Medical College of Virginia Campus, Virginia Commonwealth University, Richmond, Virginia 23298,McGuire Veterans Affairs Medical Center, Medical College of Virginia Campus, Virginia Commonwealth University, Richmond, Virginia 23298
| | - Nitai C. Hait
- Department of Biochemistry and Molecular Biology, Medical College of Virginia Campus, Virginia Commonwealth University, Richmond, Virginia 23298
| | - Xuan Wang
- Department of Microbiology and Immunology, Medical College of Virginia Campus, Virginia Commonwealth University, Richmond, Virginia 23298,McGuire Veterans Affairs Medical Center, Medical College of Virginia Campus, Virginia Commonwealth University, Richmond, Virginia 23298
| | - Jeremy C. Allegood
- Department of Biochemistry and Molecular Biology, Medical College of Virginia Campus, Virginia Commonwealth University, Richmond, Virginia 23298
| | - Akimitsu Yamada
- Department of Surgery, Medical College of Virginia Campus, Virginia Commonwealth University, Richmond, Virginia 23298,Department of Biochemistry and Molecular Biology, Medical College of Virginia Campus, Virginia Commonwealth University, Richmond, Virginia 23298
| | - Tomoyoshi Aoyagi
- Department of Surgery, Medical College of Virginia Campus, Virginia Commonwealth University, Richmond, Virginia 23298,Department of Biochemistry and Molecular Biology, Medical College of Virginia Campus, Virginia Commonwealth University, Richmond, Virginia 23298
| | - Jie Liang
- Department of Biochemistry and Molecular Biology, Medical College of Virginia Campus, Virginia Commonwealth University, Richmond, Virginia 23298
| | - William M. Pandak
- McGuire Veterans Affairs Medical Center, Medical College of Virginia Campus, Virginia Commonwealth University, Richmond, Virginia 23298
| | - Sarah Spiegel
- Department of Biochemistry and Molecular Biology, Medical College of Virginia Campus, Virginia Commonwealth University, Richmond, Virginia 23298
| | - Phillip B. Hylemon
- Department of Microbiology and Immunology, Medical College of Virginia Campus, Virginia Commonwealth University, Richmond, Virginia 23298,McGuire Veterans Affairs Medical Center, Medical College of Virginia Campus, Virginia Commonwealth University, Richmond, Virginia 23298,Address correspondence to: Huiping Zhou, Ph.D, Department of Microbiology & Immunology, Virginia Commonwealth University, P.O. Box 980678, Richmond, VA 23298-0678, Tel: 804-828-6817; Fax: 804-828-0676, Or Kazuaki Takabbe, M.D., Ph.D., FACS, Department of Surgery, VCU, P.O. Box 980011, Richmond, VA 23298-0011, Tel. 804-828-9322, Fax. 804-828-4809, Or Phillip B. Hylemon, Ph.D., Department of Microbiology and Immunology, Medical College of Virginia Campus-VCU, PO Box 908678, Richmond, VA 23298-0678, Tel: (804) 347-1752; Fax. (804) 828-0676,
| | - Huiping Zhou
- Department of Microbiology and Immunology, Medical College of Virginia Campus, Virginia Commonwealth University, Richmond, Virginia 23298,McGuire Veterans Affairs Medical Center, Medical College of Virginia Campus, Virginia Commonwealth University, Richmond, Virginia 23298,Address correspondence to: Huiping Zhou, Ph.D, Department of Microbiology & Immunology, Virginia Commonwealth University, P.O. Box 980678, Richmond, VA 23298-0678, Tel: 804-828-6817; Fax: 804-828-0676, Or Kazuaki Takabbe, M.D., Ph.D., FACS, Department of Surgery, VCU, P.O. Box 980011, Richmond, VA 23298-0011, Tel. 804-828-9322, Fax. 804-828-4809, Or Phillip B. Hylemon, Ph.D., Department of Microbiology and Immunology, Medical College of Virginia Campus-VCU, PO Box 908678, Richmond, VA 23298-0678, Tel: (804) 347-1752; Fax. (804) 828-0676,
| |
Collapse
|
733
|
Heinzmann SS, Schmitt-Kopplin P. Deep Metabotyping of the Murine Gastrointestinal Tract for the Visualization of Digestion and Microbial Metabolism. J Proteome Res 2015; 14:2267-77. [DOI: 10.1021/acs.jproteome.5b00034] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Affiliation(s)
- Silke S. Heinzmann
- Research
Unit Analytical BioGeoChemistry, Helmholtz Zentrum München, 85764 Neuherberg, Germany
- German Center for Diabetes Research (DZD), 85764 Neuherberg, Germany
| | - Philippe Schmitt-Kopplin
- Research
Unit Analytical BioGeoChemistry, Helmholtz Zentrum München, 85764 Neuherberg, Germany
- German Center for Diabetes Research (DZD), 85764 Neuherberg, Germany
- Chair
of Analytical Food Chemistry, Technische Universität München, 85354 Freising, Germany
| |
Collapse
|
734
|
Schaffner CA, Mwinyi J, Gai Z, Thasler WE, Eloranta JJ, Kullak-Ublick GA. The organic solute transporters alpha and beta are induced by hypoxia in human hepatocytes. Liver Int 2015; 35:1152-61. [PMID: 24703425 PMCID: PMC4407926 DOI: 10.1111/liv.12558] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/21/2014] [Accepted: 03/30/2014] [Indexed: 12/12/2022]
Abstract
BACKGROUND & AIMS The organic solute transporters alpha and beta (OSTα-OSTβ) form a heterodimeric transporter located at the basolateral membrane of intestinal epithelial cells and hepatocytes. Liver injury caused by ischaemia-reperfusion, cancer, inflammation or cholestasis can induce a state of hypoxia in hepatocytes. Here, we studied the effect of hypoxia on the expression of OSTα-OSTβ. METHODS OSTα-OSTβ expression was measured in Huh7 cells and primary human hepatocytes (PHH) exposed to chenodeoxycholic acid (CDCA), hypoxia or both. OSTα-OSTβ promoter activity was analysed in luciferase reporter gene assays. Binding of hypoxia-inducible factor-1 alpha (HIF-1α) to the OSTα-OSTβ gene promoters was studied in electrophoretic mobility shift assays (EMSA). RESULTS Expression of OSTα and OSTβ increased in PHH under conditions of hypoxia. Exposure of Huh7 cells or PHH to CDCA (50 μM) enhanced the effect of hypoxia on OSTα mRNA levels. In luciferase assays and EMSA, the inducing effect of low oxygen could be assigned to HIF-1α, which binds to hypoxia responsive elements (HRE) in the OSTα and OSTβ gene promoters. Site-directed mutagenesis of either the predicted HRE or the bile acid responsive FXR binding site abolished inducibility of the OSTα promoter, indicating that both elements need to be intact for induction by hypoxia and CDCA. In a rat model of chronic renal failure, the known increase in hepatic OSTα expression was associated with an increase in HIF-1α protein levels. CONCLUSION OSTα-OSTβ expression is induced by hypoxia. FXR and HIF-1α bind in close proximity to the OSTα gene promoter and produce synergistic effects on OSTα expression.
Collapse
Affiliation(s)
- Carlos A Schaffner
- Department of Clinical Pharmacology and Toxicology, University Hospital Zurich, Zurich, Switzerland
| | | | | | | | | | | |
Collapse
|
735
|
Sombetzki M, Fuchs CD, Fickert P, Österreicher CH, Mueller M, Claudel T, Loebermann M, Engelmann R, Langner C, Sahin E, Schwinge D, Guenther ND, Schramm C, Mueller-Hilke B, Reisinger EC, Trauner M. 24-nor-ursodeoxycholic acid ameliorates inflammatory response and liver fibrosis in a murine model of hepatic schistosomiasis. J Hepatol 2015; 62:871-8. [PMID: 25463533 PMCID: PMC4368108 DOI: 10.1016/j.jhep.2014.11.020] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/19/2013] [Revised: 11/09/2014] [Accepted: 11/11/2014] [Indexed: 12/21/2022]
Abstract
BACKGROUND & AIMS Intrahepatic granuloma formation and fibrosis characterize the pathological features of Schistosoma mansoni infection. Based on previously observed substantial anti-fibrotic effects of 24-nor-ursodeoxycholic acid (norUDCA) in Abcb4/Mdr2(-/-) mice with cholestatic liver injury and biliary fibrosis, we hypothesized that norUDCA improves inflammation-driven liver fibrosis in S. mansoni infection. METHODS Adult NMRI mice were infected with 50 S. mansoni cercariae and after 12 weeks received either norUDCA- or ursodeoxycholic acid (UDCA)-enriched diet (0.5% wt/wt) for 4 weeks. Bile acid effects on liver histology, serum biochemistry, key regulatory cytokines, hepatic hydroxyproline content as well as granuloma formation were compared to naive mice and infected controls. In addition, effects of norUDCA on primary T-cell activation/proliferation and maturation of the antigen-presenting-cells (dendritic cells, macrophages) were determined in vitro. RESULTS UDCA as well as norUDCA attenuated the inflammatory response in livers of S. mansoni infected mice, but exclusively norUDCA changed cellular composition and reduced size of hepatic granulomas as well as TH2-mediated hepatic fibrosis in vivo. Moreover, norUDCA affected surface expression level of major histocompatibility complex (MHC) class II of macrophages and dendritic cells as well as activation/proliferation of T-lymphocytes in vitro, whereas UDCA had no effect. CONCLUSIONS This study demonstrates pronounced anti-inflammatory and anti-fibrotic effects of norUDCA compared to UDCA in S. mansoni induced liver injury, and indicates that norUDCA directly represses antigen presentation of antigen presenting cells and subsequent T-cell activation in vitro. Therefore, norUDCA represents a promising drug for the treatment of this important cause of liver fibrosis.
Collapse
Affiliation(s)
- Martina Sombetzki
- Division of Tropical Medicine and Infectious Diseases, Department of Internal Medicine, University of Rostock, Germany
| | - Claudia D Fuchs
- Hans Popper Laboratory of Molecular Hepatology, Division of Gastroenterology and Hepatology, Department of Internal Medicine III, Medical University of Vienna, Austria
| | - Peter Fickert
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Medical University of Graz, Austria; Institute of Pathology, Medical University Graz, Austria
| | - Christoph H Österreicher
- Institute of Pharmacology, Center for Physiology and Pharmacology, Medical University of Vienna, Austria
| | - Michaela Mueller
- Hans Popper Laboratory of Molecular Hepatology, Division of Gastroenterology and Hepatology, Department of Internal Medicine III, Medical University of Vienna, Austria
| | - Thierry Claudel
- Hans Popper Laboratory of Molecular Hepatology, Division of Gastroenterology and Hepatology, Department of Internal Medicine III, Medical University of Vienna, Austria
| | - Micha Loebermann
- Division of Tropical Medicine and Infectious Diseases, Department of Internal Medicine, University of Rostock, Germany
| | | | - Cord Langner
- Institute of Pathology, Medical University Graz, Austria
| | - Emine Sahin
- Institute for Physiology, Center for Physiology and Pharmacology, Medical University of Vienna, Austria
| | - Dorothee Schwinge
- Department of Medicine I, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Nina D Guenther
- Department of Medicine I, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Christoph Schramm
- Department of Medicine I, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | | | - Emil C Reisinger
- Division of Tropical Medicine and Infectious Diseases, Department of Internal Medicine, University of Rostock, Germany
| | - Michael Trauner
- Hans Popper Laboratory of Molecular Hepatology, Division of Gastroenterology and Hepatology, Department of Internal Medicine III, Medical University of Vienna, Austria.
| |
Collapse
|
736
|
Yu DD, Sousa KM, Mattern DL, Wagner J, Fu X, Vaidehi N, Forman BM, Huang W. Stereoselective synthesis, biological evaluation, and modeling of novel bile acid-derived G-protein coupled Bile acid receptor 1 (GP-BAR1, TGR5) agonists. Bioorg Med Chem 2015; 23:1613-28. [DOI: 10.1016/j.bmc.2015.01.048] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2014] [Revised: 01/19/2015] [Accepted: 01/27/2015] [Indexed: 12/31/2022]
|
737
|
Shih DM, Yu JM, Vergnes L, Dali-Youcef N, Champion MD, Devarajan A, Zhang P, Castellani LW, Brindley DN, Jamey C, Auwerx J, Reddy ST, Ford DA, Reue K, Lusis AJ. PON3 knockout mice are susceptible to obesity, gallstone formation, and atherosclerosis. FASEB J 2015; 29:1185-97. [PMID: 25477283 PMCID: PMC4396607 DOI: 10.1096/fj.14-260570] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2014] [Accepted: 11/07/2014] [Indexed: 11/11/2022]
Abstract
We report the engineering and characterization of paraoxonase-3 knockout mice (Pon3KO). The mice were generally healthy but exhibited quantitative alterations in bile acid metabolism and a 37% increased body weight compared to the wild-type mice on a high fat diet. PON3 was enriched in the mitochondria-associated membrane fraction of hepatocytes. PON3 deficiency resulted in impaired mitochondrial respiration, increased mitochondrial superoxide levels, and increased hepatic expression of inflammatory genes. PON3 deficiency did not influence atherosclerosis development on an apolipoprotein E null hyperlipidemic background, but it did lead to a significant 60% increase in atherosclerotic lesion size in Pon3KO mice on the C57BL/6J background when fed a cholate-cholesterol diet. On the diet, the Pon3KO had significantly increased plasma intermediate-density lipoprotein/LDL cholesterol and bile acid levels. They also exhibited significantly elevated levels of hepatotoxicity markers in circulation, a 58% increase in gallstone weight, a 40% increase in hepatic cholesterol level, and increased mortality. Furthermore, Pon3KO mice exhibited decreased hepatic bile acid synthesis and decreased bile acid levels in the small intestine compared with wild-type mice. Our study suggests a role for PON3 in the metabolism of lipid and bile acid as well as protection against atherosclerosis, gallstone disease, and obesity.
Collapse
Affiliation(s)
- Diana M Shih
- *Division of Cardiology, Department of Medicine, Department of Microbiology, Immunology, and Molecular Genetics, Department of Human Genetics, Department of Molecular and Medical Pharmacology, and Department of Medicine and Molecular Biology Institute, University of California at Los Angeles, Los Angeles, California, USA; IGBMC, Illkirch and Hôpitaux Universitaires de Strasbourg, and **Laboratoire de Toxicologie, Universitaires de Strasbourg, Strasbourg, France; Department of Biochemistry and Molecular Biology, and Center for Cardiovascular Research, St. Louis University School of Medicine, St. Louis, Missouri, USA; University of Alberta, Edmonton, Alberta, Canada; and Laboratory for Integrative and Systems Physiology, School of Life Sciences, Ecole Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
| | - Janet M Yu
- *Division of Cardiology, Department of Medicine, Department of Microbiology, Immunology, and Molecular Genetics, Department of Human Genetics, Department of Molecular and Medical Pharmacology, and Department of Medicine and Molecular Biology Institute, University of California at Los Angeles, Los Angeles, California, USA; IGBMC, Illkirch and Hôpitaux Universitaires de Strasbourg, and **Laboratoire de Toxicologie, Universitaires de Strasbourg, Strasbourg, France; Department of Biochemistry and Molecular Biology, and Center for Cardiovascular Research, St. Louis University School of Medicine, St. Louis, Missouri, USA; University of Alberta, Edmonton, Alberta, Canada; and Laboratory for Integrative and Systems Physiology, School of Life Sciences, Ecole Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
| | - Laurent Vergnes
- *Division of Cardiology, Department of Medicine, Department of Microbiology, Immunology, and Molecular Genetics, Department of Human Genetics, Department of Molecular and Medical Pharmacology, and Department of Medicine and Molecular Biology Institute, University of California at Los Angeles, Los Angeles, California, USA; IGBMC, Illkirch and Hôpitaux Universitaires de Strasbourg, and **Laboratoire de Toxicologie, Universitaires de Strasbourg, Strasbourg, France; Department of Biochemistry and Molecular Biology, and Center for Cardiovascular Research, St. Louis University School of Medicine, St. Louis, Missouri, USA; University of Alberta, Edmonton, Alberta, Canada; and Laboratory for Integrative and Systems Physiology, School of Life Sciences, Ecole Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
| | - Nassim Dali-Youcef
- *Division of Cardiology, Department of Medicine, Department of Microbiology, Immunology, and Molecular Genetics, Department of Human Genetics, Department of Molecular and Medical Pharmacology, and Department of Medicine and Molecular Biology Institute, University of California at Los Angeles, Los Angeles, California, USA; IGBMC, Illkirch and Hôpitaux Universitaires de Strasbourg, and **Laboratoire de Toxicologie, Universitaires de Strasbourg, Strasbourg, France; Department of Biochemistry and Molecular Biology, and Center for Cardiovascular Research, St. Louis University School of Medicine, St. Louis, Missouri, USA; University of Alberta, Edmonton, Alberta, Canada; and Laboratory for Integrative and Systems Physiology, School of Life Sciences, Ecole Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
| | - Matthew D Champion
- *Division of Cardiology, Department of Medicine, Department of Microbiology, Immunology, and Molecular Genetics, Department of Human Genetics, Department of Molecular and Medical Pharmacology, and Department of Medicine and Molecular Biology Institute, University of California at Los Angeles, Los Angeles, California, USA; IGBMC, Illkirch and Hôpitaux Universitaires de Strasbourg, and **Laboratoire de Toxicologie, Universitaires de Strasbourg, Strasbourg, France; Department of Biochemistry and Molecular Biology, and Center for Cardiovascular Research, St. Louis University School of Medicine, St. Louis, Missouri, USA; University of Alberta, Edmonton, Alberta, Canada; and Laboratory for Integrative and Systems Physiology, School of Life Sciences, Ecole Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
| | - Asokan Devarajan
- *Division of Cardiology, Department of Medicine, Department of Microbiology, Immunology, and Molecular Genetics, Department of Human Genetics, Department of Molecular and Medical Pharmacology, and Department of Medicine and Molecular Biology Institute, University of California at Los Angeles, Los Angeles, California, USA; IGBMC, Illkirch and Hôpitaux Universitaires de Strasbourg, and **Laboratoire de Toxicologie, Universitaires de Strasbourg, Strasbourg, France; Department of Biochemistry and Molecular Biology, and Center for Cardiovascular Research, St. Louis University School of Medicine, St. Louis, Missouri, USA; University of Alberta, Edmonton, Alberta, Canada; and Laboratory for Integrative and Systems Physiology, School of Life Sciences, Ecole Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
| | - Peixiang Zhang
- *Division of Cardiology, Department of Medicine, Department of Microbiology, Immunology, and Molecular Genetics, Department of Human Genetics, Department of Molecular and Medical Pharmacology, and Department of Medicine and Molecular Biology Institute, University of California at Los Angeles, Los Angeles, California, USA; IGBMC, Illkirch and Hôpitaux Universitaires de Strasbourg, and **Laboratoire de Toxicologie, Universitaires de Strasbourg, Strasbourg, France; Department of Biochemistry and Molecular Biology, and Center for Cardiovascular Research, St. Louis University School of Medicine, St. Louis, Missouri, USA; University of Alberta, Edmonton, Alberta, Canada; and Laboratory for Integrative and Systems Physiology, School of Life Sciences, Ecole Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
| | - Lawrence W Castellani
- *Division of Cardiology, Department of Medicine, Department of Microbiology, Immunology, and Molecular Genetics, Department of Human Genetics, Department of Molecular and Medical Pharmacology, and Department of Medicine and Molecular Biology Institute, University of California at Los Angeles, Los Angeles, California, USA; IGBMC, Illkirch and Hôpitaux Universitaires de Strasbourg, and **Laboratoire de Toxicologie, Universitaires de Strasbourg, Strasbourg, France; Department of Biochemistry and Molecular Biology, and Center for Cardiovascular Research, St. Louis University School of Medicine, St. Louis, Missouri, USA; University of Alberta, Edmonton, Alberta, Canada; and Laboratory for Integrative and Systems Physiology, School of Life Sciences, Ecole Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
| | - David N Brindley
- *Division of Cardiology, Department of Medicine, Department of Microbiology, Immunology, and Molecular Genetics, Department of Human Genetics, Department of Molecular and Medical Pharmacology, and Department of Medicine and Molecular Biology Institute, University of California at Los Angeles, Los Angeles, California, USA; IGBMC, Illkirch and Hôpitaux Universitaires de Strasbourg, and **Laboratoire de Toxicologie, Universitaires de Strasbourg, Strasbourg, France; Department of Biochemistry and Molecular Biology, and Center for Cardiovascular Research, St. Louis University School of Medicine, St. Louis, Missouri, USA; University of Alberta, Edmonton, Alberta, Canada; and Laboratory for Integrative and Systems Physiology, School of Life Sciences, Ecole Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
| | - Carole Jamey
- *Division of Cardiology, Department of Medicine, Department of Microbiology, Immunology, and Molecular Genetics, Department of Human Genetics, Department of Molecular and Medical Pharmacology, and Department of Medicine and Molecular Biology Institute, University of California at Los Angeles, Los Angeles, California, USA; IGBMC, Illkirch and Hôpitaux Universitaires de Strasbourg, and **Laboratoire de Toxicologie, Universitaires de Strasbourg, Strasbourg, France; Department of Biochemistry and Molecular Biology, and Center for Cardiovascular Research, St. Louis University School of Medicine, St. Louis, Missouri, USA; University of Alberta, Edmonton, Alberta, Canada; and Laboratory for Integrative and Systems Physiology, School of Life Sciences, Ecole Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
| | - Johan Auwerx
- *Division of Cardiology, Department of Medicine, Department of Microbiology, Immunology, and Molecular Genetics, Department of Human Genetics, Department of Molecular and Medical Pharmacology, and Department of Medicine and Molecular Biology Institute, University of California at Los Angeles, Los Angeles, California, USA; IGBMC, Illkirch and Hôpitaux Universitaires de Strasbourg, and **Laboratoire de Toxicologie, Universitaires de Strasbourg, Strasbourg, France; Department of Biochemistry and Molecular Biology, and Center for Cardiovascular Research, St. Louis University School of Medicine, St. Louis, Missouri, USA; University of Alberta, Edmonton, Alberta, Canada; and Laboratory for Integrative and Systems Physiology, School of Life Sciences, Ecole Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
| | - Srinivasa T Reddy
- *Division of Cardiology, Department of Medicine, Department of Microbiology, Immunology, and Molecular Genetics, Department of Human Genetics, Department of Molecular and Medical Pharmacology, and Department of Medicine and Molecular Biology Institute, University of California at Los Angeles, Los Angeles, California, USA; IGBMC, Illkirch and Hôpitaux Universitaires de Strasbourg, and **Laboratoire de Toxicologie, Universitaires de Strasbourg, Strasbourg, France; Department of Biochemistry and Molecular Biology, and Center for Cardiovascular Research, St. Louis University School of Medicine, St. Louis, Missouri, USA; University of Alberta, Edmonton, Alberta, Canada; and Laboratory for Integrative and Systems Physiology, School of Life Sciences, Ecole Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
| | - David A Ford
- *Division of Cardiology, Department of Medicine, Department of Microbiology, Immunology, and Molecular Genetics, Department of Human Genetics, Department of Molecular and Medical Pharmacology, and Department of Medicine and Molecular Biology Institute, University of California at Los Angeles, Los Angeles, California, USA; IGBMC, Illkirch and Hôpitaux Universitaires de Strasbourg, and **Laboratoire de Toxicologie, Universitaires de Strasbourg, Strasbourg, France; Department of Biochemistry and Molecular Biology, and Center for Cardiovascular Research, St. Louis University School of Medicine, St. Louis, Missouri, USA; University of Alberta, Edmonton, Alberta, Canada; and Laboratory for Integrative and Systems Physiology, School of Life Sciences, Ecole Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
| | - Karen Reue
- *Division of Cardiology, Department of Medicine, Department of Microbiology, Immunology, and Molecular Genetics, Department of Human Genetics, Department of Molecular and Medical Pharmacology, and Department of Medicine and Molecular Biology Institute, University of California at Los Angeles, Los Angeles, California, USA; IGBMC, Illkirch and Hôpitaux Universitaires de Strasbourg, and **Laboratoire de Toxicologie, Universitaires de Strasbourg, Strasbourg, France; Department of Biochemistry and Molecular Biology, and Center for Cardiovascular Research, St. Louis University School of Medicine, St. Louis, Missouri, USA; University of Alberta, Edmonton, Alberta, Canada; and Laboratory for Integrative and Systems Physiology, School of Life Sciences, Ecole Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
| | - Aldons J Lusis
- *Division of Cardiology, Department of Medicine, Department of Microbiology, Immunology, and Molecular Genetics, Department of Human Genetics, Department of Molecular and Medical Pharmacology, and Department of Medicine and Molecular Biology Institute, University of California at Los Angeles, Los Angeles, California, USA; IGBMC, Illkirch and Hôpitaux Universitaires de Strasbourg, and **Laboratoire de Toxicologie, Universitaires de Strasbourg, Strasbourg, France; Department of Biochemistry and Molecular Biology, and Center for Cardiovascular Research, St. Louis University School of Medicine, St. Louis, Missouri, USA; University of Alberta, Edmonton, Alberta, Canada; and Laboratory for Integrative and Systems Physiology, School of Life Sciences, Ecole Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
| |
Collapse
|
738
|
Proteomic and metabonomic biomarkers for hepatocellular carcinoma: a comprehensive review. Br J Cancer 2015; 112:1141-56. [PMID: 25826224 PMCID: PMC4385954 DOI: 10.1038/bjc.2015.38] [Citation(s) in RCA: 95] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2014] [Revised: 11/04/2014] [Accepted: 12/20/2014] [Indexed: 12/12/2022] Open
Abstract
Hepatocellular carcinoma (HCC) ranks third in overall global cancer-related mortality. Symptomatic presentation often means advanced disease where potentially curative treatment options become very limited. Numerous international guidelines propose the routine monitoring of those with the highest risk factors for the condition in order to diagnose potential tumourigenesis early. To aid this, the fields of metabonomic- and proteomic-based biomarker discovery have applied advanced tools to identify early changes in protein and metabolite expression in HCC patients vs controls. With robust validation, it is anticipated that from these candidates will rise a high-performance non-invasive test able to diagnose early HCC and related conditions. This review gathers the numerous markers proposed by studies using mass spectrometry and proton nuclear magnetic resonance spectroscopy and evaluates areas of consistency as well as discordance.
Collapse
|
739
|
|
740
|
Gitto S, Vitale G, Villa E, Andreone P. Treatment of nonalcoholic steatohepatitis in adults: present and future. Gastroenterol Res Pract 2015; 2015:732870. [PMID: 25866507 PMCID: PMC4381725 DOI: 10.1155/2015/732870] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/04/2015] [Accepted: 03/05/2015] [Indexed: 02/08/2023] Open
Abstract
Nonalcoholic steatohepatitis has become one of the most common liver-related health problems. This condition has been linked to an unhealthy diet and weight gain, but it can also be observed in nonobese people. The standard of care is represented by the lifestyle intervention. However, because this approach has several limitations, such as a lack of compliance, the use of many drugs has been proposed. The first-line pharmacological choices are vitamin E and pioglitazone, both showing a positive effect on transaminases, fat accumulation, and inflammation. Nevertheless, vitamin E has no proven effect on fibrosis and on long-term morbidity and mortality and pioglitazone has a negative impact on weight. Other drugs have been studied such as metformin, ursodeoxycholic acid, statins, pentoxiphylline, and orlistat with only partially positive results. Among the emerging treatments, telmisartan is particularly interesting as it seems to have an impact on insulin resistance, liver steatosis, inflammation, and fibrosis. However, the pathogenesis of steatohepatitis is highly complex and is determined by different parallel hits; indeed, the association of different drugs that act on various levels has been suggested. In conclusion, lifestyle intervention should be optimised and the associations of different drugs should be tested in large studies with long-term outcomes.
Collapse
Affiliation(s)
- S. Gitto
- Dipartimento di Gastroenterologia, Azienda Ospedaliero-Universitaria and Università di Modena e Reggio Emilia, 41125 Modena, Italy
| | - G. Vitale
- Dipartimento di Scienze Mediche e Chirurgiche, Università di Bologna e Dipartimento dell'Apparato Digerente, Azienda Ospedaliero-Universitaria di Bologna, Policlinico Sant'Orsola Malpighi, 40138 Bologna, Italy
| | - E. Villa
- Dipartimento di Gastroenterologia, Azienda Ospedaliero-Universitaria and Università di Modena e Reggio Emilia, 41125 Modena, Italy
| | - P. Andreone
- Dipartimento di Scienze Mediche e Chirurgiche, Università di Bologna e Dipartimento dell'Apparato Digerente, Azienda Ospedaliero-Universitaria di Bologna, Policlinico Sant'Orsola Malpighi, 40138 Bologna, Italy
| |
Collapse
|
741
|
Li P, Ruan X, Yang L, Kiesewetter K, Zhao Y, Luo H, Chen Y, Gucek M, Zhu J, Cao H. A liver-enriched long non-coding RNA, lncLSTR, regulates systemic lipid metabolism in mice. Cell Metab 2015; 21:455-67. [PMID: 25738460 PMCID: PMC4350020 DOI: 10.1016/j.cmet.2015.02.004] [Citation(s) in RCA: 238] [Impact Index Per Article: 23.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/20/2014] [Revised: 12/12/2014] [Accepted: 02/06/2015] [Indexed: 02/01/2023]
Abstract
Long non-coding RNAs (lncRNAs) constitute a significant portion of mammalian genome, yet the physiological importance of lncRNAs is largely unknown. Here, we identify a liver-enriched lncRNA in mouse that we term liver-specific triglyceride regulator (lncLSTR). Mice with a liver-specific depletion of lncLSTR exhibit a marked reduction in plasma triglyceride levels. We show that lncLSTR depletion enhances apoC2 expression, leading to robust lipoprotein lipase activation and increased plasma triglyceride clearance. We further demonstrate that the regulation of apoC2 expression occurs through an FXR-mediated pathway. LncLSTR forms a molecular complex with TDP-43 to regulate expression of Cyp8b1, a key enzyme in the bile acid synthesis pathway, and engenders an in vivo bile pool that induces apoC2 expression through FXR. Finally, we demonstrate that lncLSTR depletion can reduce triglyceride levels in a hyperlipidemia mouse model. Taken together, these data support a model in which lncLSTR regulates a TDP-43/FXR/apoC2-dependent pathway to maintain systemic lipid homeostasis.
Collapse
Affiliation(s)
- Ping Li
- Center for Molecular Medicine, National Heart, Lung and Blood Institute, NIH, Bethesda, MD 20892, USA
| | - Xiangbo Ruan
- Center for Molecular Medicine, National Heart, Lung and Blood Institute, NIH, Bethesda, MD 20892, USA
| | - Ling Yang
- Center for Molecular Medicine, National Heart, Lung and Blood Institute, NIH, Bethesda, MD 20892, USA
| | - Kurtis Kiesewetter
- Center for Molecular Medicine, National Heart, Lung and Blood Institute, NIH, Bethesda, MD 20892, USA
| | - Yi Zhao
- Key Laboratory of Intelligent Information Processing, Institute of Computing Technology, Chinese Academy of Sciences, Beijing, PR China
| | - Haitao Luo
- Key Laboratory of Intelligent Information Processing, Institute of Computing Technology, Chinese Academy of Sciences, Beijing, PR China
| | - Yong Chen
- Proteomics Core, National Heart, Lung and Blood Institute, NIH, Bethesda, MD 20892, USA
| | - Marjan Gucek
- Proteomics Core, National Heart, Lung and Blood Institute, NIH, Bethesda, MD 20892, USA
| | - Jun Zhu
- Systems Biology Center, National Heart, Lung and Blood Institute, NIH, Bethesda, MD 20892, USA
| | - Haiming Cao
- Center for Molecular Medicine, National Heart, Lung and Blood Institute, NIH, Bethesda, MD 20892, USA.
| |
Collapse
|
742
|
Ferrebee CB, Dawson PA. Metabolic effects of intestinal absorption and enterohepatic cycling of bile acids. Acta Pharm Sin B 2015; 5:129-34. [PMID: 26579438 PMCID: PMC4629214 DOI: 10.1016/j.apsb.2015.01.001] [Citation(s) in RCA: 88] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2014] [Revised: 12/30/2014] [Accepted: 01/04/2015] [Indexed: 12/13/2022] Open
Abstract
The classical functions of bile acids include acting as detergents to facilitate the digestion and absorption of nutrients in the gut. In addition, bile acids also act as signaling molecules to regulate glucose homeostasis, lipid metabolism and energy expenditure. The signaling potential of bile acids in compartments such as the systemic circulation is regulated in part by an efficient enterohepatic circulation that functions to conserve and channel the pool of bile acids within the intestinal and hepatobiliary compartments. Changes in hepatobiliary and intestinal bile acid transport can alter the composition, size, and distribution of the bile acid pool. These alterations in turn can have significant effects on bile acid signaling and their downstream metabolic targets. This review discusses recent advances in our understanding of the inter-relationship between the enterohepatic cycling of bile acids and the metabolic consequences of signaling via bile acid-activated receptors, such as farnesoid X nuclear receptor (FXR) and the G-protein-coupled bile acid receptor (TGR5).
Collapse
Key Words
- ACCII, acetyl-CoA carboxylase 2
- APO, apolipoproteins
- ASBT, apical sodium-dependent bile acid transporter
- BSEP, bile salt export pump
- Bile acids
- CYP7A1, cholesterol 7α-hydroxylase
- DIO2, deiodinase 2
- Energy homeostasis
- FAS, fatty acid synthase
- FGF, fibroblast growth factor
- FGFR4, fibroblast growth factor receptor 4
- FOXO1, forkhead box protein O1
- FXR, farnesoid X-receptor
- G6Pase, glucose-6-phosphatase
- GLP-1, glucagon-like polypeptide-1
- HNF4α, hepatocyte nuclear factor 4 alpha
- IBABP, ileal bile acid binding protein
- Intestine
- LDL, low density lipoprotein
- Lipid metabolism
- Liver
- NTCP, Na+-taurocholate transporting polypeptide
- OATP, organic anion transporting polypeptide
- OST, organic solute transporter
- PEPCK, phosphoenolpyruvate carboxykinase
- PGC1α, peroxisome proliferator-activated receptor gamma coactivator 1 alpha
- PPAR, peroxisome proliferator-activated receptor
- SHP, small heterodimer partner
- SREBP1c, sterol regulatory element binding protein-1c
- T4, thyroid hormone
- TGR5, G-protein-coupled bile acid receptor
- Transporters
- VLDL, very low density lipoprotein
Collapse
|
743
|
Wirth J, Giuseppe RD, Wientzek A, Katzke VA, Kloss M, Kaaks R, Boeing H, Weikert C. Presence of gallstones and the risk of cardiovascular diseases: The EPIC-Germany cohort study. Eur J Prev Cardiol 2015; 22:326-334. [DOI: 10.1177/2047487313512218] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 08/30/2023]
Affiliation(s)
- Janine Wirth
- Department of Epidemiology, German Institute of Human Nutrition Potsdam-Rehbruecke, Germany
| | - Romina di Giuseppe
- Department of Epidemiology, German Institute of Human Nutrition Potsdam-Rehbruecke, Germany
| | - Angelika Wientzek
- Department of Epidemiology, German Institute of Human Nutrition Potsdam-Rehbruecke, Germany
| | - Verena A Katzke
- Division of Cancer Epidemiology, German Cancer Research Center Heidelberg, Germany
| | - Manja Kloss
- Department of Neurology, University of Heidelberg, Germany
| | - Rudolf Kaaks
- Division of Cancer Epidemiology, German Cancer Research Center Heidelberg, Germany
| | - Heiner Boeing
- Department of Epidemiology, German Institute of Human Nutrition Potsdam-Rehbruecke, Germany
| | - Cornelia Weikert
- Department of Epidemiology, German Institute of Human Nutrition Potsdam-Rehbruecke, Germany
- Institute of Social Medicine, Epidemiology, and Health Economics, Charité University, Medical Center, Berlin, Germany
| |
Collapse
|
744
|
Ding L, Yang L, Wang Z, Huang W. Bile acid nuclear receptor FXR and digestive system diseases. Acta Pharm Sin B 2015; 5:135-44. [PMID: 26579439 PMCID: PMC4629217 DOI: 10.1016/j.apsb.2015.01.004] [Citation(s) in RCA: 306] [Impact Index Per Article: 30.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2014] [Revised: 12/31/2014] [Accepted: 01/05/2015] [Indexed: 12/14/2022] Open
Abstract
Bile acids (BAs) are not only digestive surfactants but also important cell signaling molecules, which stimulate several signaling pathways to regulate some important biological processes. The bile-acid-activated nuclear receptor, farnesoid X receptor (FXR), plays a pivotal role in regulating bile acid, lipid and glucose homeostasis as well as in regulating the inflammatory responses, barrier function and prevention of bacterial translocation in the intestinal tract. As expected, FXR is involved in the pathophysiology of a wide range of diseases of gastrointestinal tract, including inflammatory bowel disease, colorectal cancer and type 2 diabetes. In this review, we discuss current knowledge of the roles of FXR in physiology of the digestive system and the related diseases. Better understanding of the roles of FXR in digestive system will accelerate the development of FXR ligands/modulators for the treatment of digestive system diseases.
Collapse
Key Words
- 6-ECDCA, 6α-ethyl-chenodeoxycholic acid
- AF2, activation domain
- ANGTPL3, angiopoietin-like protein 3
- AOM, azoxymethane
- AP-1, activator protein-1
- ASBT, apical sodium-dependent bile salt transporter
- Apo, apolipoprotein
- BAAT, bile acid-CoA amino acid N-acetyltransferase
- BACS, bile acid-CoA synthetase
- BAs, bile acids
- BMI, body mass index
- BSEP, bile salt export pump
- Bile acids
- CA, cholic acid
- CD, Crohn׳s disease
- CDCA, chenodeoxycholic acid
- CREB, cAMP regulatory element-binding protein
- CYP7A1, cholesterol 7α-hydroxylase
- Colorectal cancer
- DBD, DNA binding domain
- DCA, deoxycholic acid
- DSS, dextrane sodium sulfate
- ERK, extracellular signal-regulated kinase
- FABP6, fatty acid-binding protein subclass 6
- FFAs, free fatty acids
- FGF19, fibroblast growth factor 19
- FGFR4, fibroblast growth factor receptor 4
- FXR, farnesoid X receptor
- FXRE, farnesoid X receptor response element
- Farnesoid X receptor
- G6Pase, glucose-6-phosphatase
- GLP-1, glucagon-like peptide 1
- GLUT2, glucose transporter type 2
- GPBAR, G protein-coupled BA receptor
- GPCRs, G protein-coupled receptors
- GSK3, glycogen synthase kinase 3
- Gastrointestinal tract
- HDL-C, high density lipoprotein cholesterol
- HNF4α, hepatic nuclear factor 4α
- I-BABP, intestinal bile acid-binding protein
- IBD, inflammatory bowel disease
- IL-1, interleukin 1
- Inflammatory bowel disease
- KLF11, Krüppel-like factor 11
- KRAS, Kirsten rat sarcoma viral oncogene homolog
- LBD, ligand binding domain
- LCA, lithocholic acid
- LPL, lipoprotein lipase
- LRH-1, liver receptor homolog-1
- MCA, muricholicacid
- MRP2, multidrug resistance-associated protein 2
- NF-κB, nuclear factor-kappa B
- NOD, non-obese diabetic
- NRs, nuclear receptors
- OSTα, organic solute transporter alpha
- OSTβ, organic solute transporter beta
- PEPCK, phosphoenol pyruvate carboxykinase
- PGC-1α, peroxisome proliferators-activated receptor γ coactivator protein-1α
- SHP, small heterodimer partner
- SREBP-1c, sterol regulatory element-binding protein 1c
- STAT3, signal transducers and activators of transcription 3
- T2D, type 2 diabetes
- TLCA, taurolithocholic acid
- TNBS, trinitrobenzensulfonic acid
- TNFα, tumor necrosis factors α
- Type 2 diabetes
- UC, ulcerative colitis
- UDCA, ursodeoxycholic acid
- VSG, vertical sleeve gastrectomy
- db/db, diabetic mice
Collapse
|
745
|
Manley S, Ding W. Role of farnesoid X receptor and bile acids in alcoholic liver disease. Acta Pharm Sin B 2015; 5:158-67. [PMID: 26579442 PMCID: PMC4629219 DOI: 10.1016/j.apsb.2014.12.011] [Citation(s) in RCA: 81] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2014] [Revised: 12/20/2014] [Accepted: 12/29/2014] [Indexed: 02/07/2023] Open
Abstract
Alcoholic liver disease (ALD) is one of the major causes of liver morbidity and mortality worldwide. Chronic alcohol consumption leads to development of liver pathogenesis encompassing steatosis, inflammation, fibrosis, cirrhosis, and in extreme cases, hepatocellular carcinoma. Moreover, ALD may also associate with cholestasis. Emerging evidence now suggests that farnesoid X receptor (FXR) and bile acids also play important roles in ALD. In this review, we discuss the effects of alcohol consumption on FXR, bile acids and gut microbiome as well as their impacts on ALD. Moreover, we summarize the findings on FXR, FoxO3a (forkhead box-containing protein class O3a) and PPARα (peroxisome proliferator-activated receptor alpha) in regulation of autophagy-related gene transcription program and liver injury in response to alcohol exposure.
Collapse
Key Words
- 6ECDCA, 6α-ethyl-chenodeoxycholic acid
- ADH, alcohol dehydrogenase
- AF, activation function
- AKT, protein kinase B
- ALD, alcoholic liver disease
- ALT, alanine aminotransferase
- ASBT, apical sodium dependent bile acid transporter
- Alcoholic liver disease
- Atg, autophagy-related
- Autophagy
- BAAT, bile acid CoA:amino acid N-acyltransferase
- BACS, bile acid CoA synthetase
- BSEP, bile salt export pump
- Bile acids
- CA, cholic acid
- CB1R, cannabinoid receptor type 1
- CDCA, chenodeoxycholic acid
- CREB, cAMP response element-binding protein
- CREBH, cAMP response element-binding protein, hepatocyte specific
- CRTC2, CREB regulated transcription coactivator 2
- CYP, cytochrome P450
- DCA, deoxycholic acid
- DR1, direct repeat 1
- FGF15/19, fibroblast growth factor 15/19
- FGFR4, fibroblast growth factor receptor 4
- FXR, farnesoid X receptor
- Farnesoid X receptor
- FoxO3
- FoxO3a, forkhead box-containing protein class O3a
- GGT, gamma-glutamyltranspeptidase
- HCC, hepatocellular carcinoma
- IR-1, inverted repeat-1
- KO, knockout
- LC3, light chain 3
- LRH-1, liver receptor homolog 1
- LXR, liver X receptor
- MRP4, multidrug resistance protein 4
- NAD+, nicotinamide adenine dinucleotide
- NTCP, sodium taurocholate cotransporting polypeptide
- OSTα/β, organic solute transporter α/β
- PE, phosphatidylethanolamine
- PPARα, peroxisome proliferator-activated receptor alpha
- ROS, reactive oxygen species
- RXRα, retinoid X receptor-alpha
- SHP, small heterodimer partner
- SQSTM, sequestome-1
- SREBP1, sterol regulatory element-binding protein 1
- Sirt1, sirtuin 1
- TCA, taurocholic acid
- TFEB, transcription factor EB
- TLR4, toll-like receptor 4
- TUDCA, tauro-ursodeoxycholic acid
- UDCA, ursodeoxycholic acid
- WAY, WAY-362450
- WT, wild type
Collapse
Affiliation(s)
| | - Wenxing Ding
- Corresponding author. Tel.: +1 913 5889813; fax: +1 913 5887501.
| |
Collapse
|
746
|
Deciphering metabolic abnormalities associated with Alzheimer's disease in the APP/PS1 mouse model using integrated metabolomic approaches. Biochimie 2015; 110:119-128. [DOI: 10.1016/j.biochi.2015.01.005] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2014] [Accepted: 01/05/2015] [Indexed: 01/01/2023]
|
747
|
Abstract
Itching is commonly reported by pregnant women and may be due to physiologic changes of pregnancy or could indicate a more serious health concern. Intrahepatic cholestasis of pregnancy, while classified as a pregnancy dermatosis, is actually a liver disease of pregnancy associated with significant fetal mortality and morbidity, as well as lifelong health risks for the offspring. In these challenging cases, nurses must understand the differential diagnoses and be prepared to provide comprehensive care, education and support to women with this condition. A case example is included.
Collapse
|
748
|
Magotti P, Bauer I, Igarashi M, Babagoli M, Marotta R, Piomelli D, Garau G. Structure of human N-acylphosphatidylethanolamine-hydrolyzing phospholipase D: regulation of fatty acid ethanolamide biosynthesis by bile acids. Structure 2015; 23:598-604. [PMID: 25684574 DOI: 10.1016/j.str.2014.12.018] [Citation(s) in RCA: 73] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2014] [Revised: 12/16/2014] [Accepted: 12/18/2014] [Indexed: 12/31/2022]
Abstract
The fatty acid ethanolamides (FAEs) are lipid mediators present in all organisms and involved in highly conserved biological functions, such as innate immunity, energy balance, and stress control. They are produced from membrane N-acylphosphatidylethanolamines (NAPEs) and include agonists for G protein-coupled receptors (e.g., cannabinoid receptors) and nuclear receptors (e.g., PPAR-α). Here, we report the crystal structure of human NAPE-hydrolyzing phospholipase D (NAPE-PLD) at 2.65 Å resolution, a membrane enzyme that catalyzes FAE formation in mammals. NAPE-PLD forms homodimers partly separated by an internal ∼ 9-Å-wide channel and uniquely adapted to associate with phospholipids. A hydrophobic cavity provides an entryway for NAPE into the active site, where a binuclear Zn(2+) center orchestrates its hydrolysis. Bile acids bind with high affinity to selective pockets in this cavity, enhancing dimer assembly and enabling catalysis. These elements offer multiple targets for the design of small-molecule NAPE-PLD modulators with potential applications in inflammation and metabolic disorders.
Collapse
Affiliation(s)
- Paola Magotti
- Department of Drug Discovery and Development, Fondazione Istituto Italiano di Tecnologia, Via Morego 30, 16163 Genoa, Italy
| | - Inga Bauer
- Department of Drug Discovery and Development, Fondazione Istituto Italiano di Tecnologia, Via Morego 30, 16163 Genoa, Italy
| | - Miki Igarashi
- Department of Anatomy & Neurobiology, University of California - Irvine, Gillespie NRF 3101, Irvine, CA 92697, USA
| | - Masih Babagoli
- Department of Anatomy & Neurobiology, University of California - Irvine, Gillespie NRF 3101, Irvine, CA 92697, USA
| | - Roberto Marotta
- Nanochemistry, Fondazione Istituto Italiano di Tecnologia, Via Morego 30, 16163 Genoa, Italy
| | - Daniele Piomelli
- Department of Drug Discovery and Development, Fondazione Istituto Italiano di Tecnologia, Via Morego 30, 16163 Genoa, Italy; Department of Anatomy & Neurobiology, University of California - Irvine, Gillespie NRF 3101, Irvine, CA 92697, USA.
| | - Gianpiero Garau
- Department of Drug Discovery and Development, Fondazione Istituto Italiano di Tecnologia, Via Morego 30, 16163 Genoa, Italy.
| |
Collapse
|
749
|
Xie G, Wang Y, Wang X, Zhao A, Chen T, Ni Y, Wong L, Zhang H, Zhang J, Liu C, Liu P, Jia W. Profiling of serum bile acids in a healthy Chinese population using UPLC-MS/MS. J Proteome Res 2015; 14:850-859. [PMID: 25581415 DOI: 10.1021/pr500920q] [Citation(s) in RCA: 127] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Bile acids (BAs) are a group of important physiological agents for cholesterol metabolism, intestinal nutrient absorption, and biliary secretion of lipids, toxic metabolites, and xenobiotics. Extensive research in the last two decades has unveiled new functions of BAs as signaling molecules and metabolic regulators that modulate hepatic lipid, glucose, and energy homeostasis through the activation of nuclear receptors and G-protein-coupled receptor signaling in gut-liver metabolic axis involving host-gut microbial co-metabolism. Therefore, investigation of serum BA profiles, in healthy human male and female subjects with a wide range of age and body mass index (BMI), will provide important baseline information on the BA physiology as well as metabolic homeostasis among human subjects that are regulated by two sets of genome, host genome, and symbiotic microbiome. Previous reports on age- or gender-related changes on BA profiles in animals and human showed inconsistent results, and the information acquired from various studies was highly fragmentary. Here we profiled the serum BAs in a large population of healthy participants (n = 502) and examined the impact of age, gender, and BMI on serum BA concentrations and compositions using a targeted metabonomics approach with ultraperformance liquid chromatography triple-quadrupole mass spectrometry. We found that the BA profiles were dependent on gender, age, and BMI among study subjects. The total BAs were significantly higher in males than in females (p < 0.05) and higher in obese females than in lean females (p < 0.05). The difference in BA profiles between male and female subjects was decreased at age of 50-70 years, while the difference in BA profiles between lean and obese increased for subjects aged 50-70 years. The study provides a comprehensive understanding of the BA profiles in healthy subjects and highlights the need to take into account age, gender, and BMI differences when investigating pathophysiological changes of BAs resulting from gastrointestinal diseases.
Collapse
Affiliation(s)
- Guoxiang Xie
- Shanghai Key Laboratory of Diabetes Mellitus and Center for Translational Medicine, Shanghai Jiao Tong University Affiliated Sixth People's Hospital , 600 Yishan Road, Shanghai 200233, China
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
750
|
Bathena SPR, Thakare R, Gautam N, Mukherjee S, Olivera M, Meza J, Alnouti Y. Urinary bile acids as biomarkers for liver diseases I. Stability of the baseline profile in healthy subjects. Toxicol Sci 2015; 143:296-307. [PMID: 25344562 DOI: 10.1093/toxsci/kfu227] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
The role of bile acids (BAs) as biomarkers for liver injury has been proposed for decades. However, the large inter- and intra-individual variability of the BA profile has prevented its clinical application. To this end, we investigated the effect of covariates such as food, gender, age, BMI, and moderate alcohol consumption on the BA profile in healthy human subjects. The BA profile was characterized by the calculation of indices that describe the composition, sulfation, and amidation of total and individual BAs. Both inter- and intra-individual variabilities of BA indices were low in serum and even lower in urine compared with those of absolute concentrations of BAs. Serum BA concentrations increased with consumption of food, whereas urinary BA concentrations were mildly affected by food. Gender differences in the urinary and serum BA profile were minimal. The serum and urinary BA profiles were also not affected by age. BMI showed minimal effect on the urine and serum BA profile. Moderate alcohol consumption did not have a significant effect on the BA profile in both urine and serum. When the effect of the type of alcohol was studied, the results indicate that moderate drinking of beer does not affect BA concentrations and has minimal effect on BA indices, whereas moderate wine consumption slightly increases BA concentrations without affecting the BA indices. In summary, urinary BA indices showed lower variability and higher stability than absolute BA concentrations in serum and showed minimal changes to covariate effects suggesting their utility as biomarkers in clinic.
Collapse
Affiliation(s)
- Sai Praneeth R Bathena
- *Department of Pharmaceutical Sciences, College of Pharmacy, Department of Internal Medicine, College of Medicine and Department of Biostatistics, College of Public Health, University of Nebraska Medical Center, Omaha, Nebraska 68198
| | - Rhishikesh Thakare
- *Department of Pharmaceutical Sciences, College of Pharmacy, Department of Internal Medicine, College of Medicine and Department of Biostatistics, College of Public Health, University of Nebraska Medical Center, Omaha, Nebraska 68198
| | - Nagsen Gautam
- *Department of Pharmaceutical Sciences, College of Pharmacy, Department of Internal Medicine, College of Medicine and Department of Biostatistics, College of Public Health, University of Nebraska Medical Center, Omaha, Nebraska 68198
| | - Sandeep Mukherjee
- *Department of Pharmaceutical Sciences, College of Pharmacy, Department of Internal Medicine, College of Medicine and Department of Biostatistics, College of Public Health, University of Nebraska Medical Center, Omaha, Nebraska 68198
| | - Marco Olivera
- *Department of Pharmaceutical Sciences, College of Pharmacy, Department of Internal Medicine, College of Medicine and Department of Biostatistics, College of Public Health, University of Nebraska Medical Center, Omaha, Nebraska 68198
| | - Jane Meza
- *Department of Pharmaceutical Sciences, College of Pharmacy, Department of Internal Medicine, College of Medicine and Department of Biostatistics, College of Public Health, University of Nebraska Medical Center, Omaha, Nebraska 68198
| | - Yazen Alnouti
- *Department of Pharmaceutical Sciences, College of Pharmacy, Department of Internal Medicine, College of Medicine and Department of Biostatistics, College of Public Health, University of Nebraska Medical Center, Omaha, Nebraska 68198 *Department of Pharmaceutical Sciences, College of Pharmacy, Department of Internal Medicine, College of Medicine and Department of Biostatistics, College of Public Health, University of Nebraska Medical Center, Omaha, Nebraska 68198
| |
Collapse
|