1
|
Zhang X, Sheng N, Wang Z, Cao Y, Jiang X, Yan H, Cheng F, Geng T, Wei K, Zhang L, Gao M, Zhou G, Chen P. Exploring the mechanism of Carbonized Typhae Pollen in treating blood stasis syndrome through metabolic profiling: the synergistic effect of hemostasis without blood stasis. JOURNAL OF ETHNOPHARMACOLOGY 2025; 351:120124. [PMID: 40513921 DOI: 10.1016/j.jep.2025.120124] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/26/2025] [Revised: 05/31/2025] [Accepted: 06/09/2025] [Indexed: 06/16/2025]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Removing blood stasis and stopping bleeding traditional Chinese medicines (RBSB-TCM) formed a unique class of TCM, characterized by vasodilating, removing stasis and hemostatic effects. Carbonized Typhae Pollen (CTP), derived from Typhae Pollen (TP) through carbonization, has emerged as a particularly valuable therapeutic agent. It has been widely used in clinical practice to treat hemorrhagic disorders caused by blood stasis syndrome (BSS). However, the potential mechanism for CTP to achieve the dual synergistic effect of promoting blood flow and hemostasis remains unclear. AIM OF THE STUDY From the standpoint of metabolite profiles, this study attempts to investigate the fundamental mechanism of CTP in the elimination of blood stasis and the cessation of bleeding. MATERIALS AND METHODS First, chemical constituents, absorbed constituents and metabolites in rats following oral administration of CTP were identified by ultra-high performance liquid chromatography coupled with the quadrupole time-of-flight mass spectrometry (UHPLC-Q-TOF-MS) method combined with MetabolitePilot 2.0.4 software. Subsequently, the pharmacological effects of CTP were systematically investigated using rat models with BSS and zebrafish with cerebral hemorrhage. Specifically, the impact on coagulation function and histopathology in rats, as well as the effect on cerebral hemorrhage in zebrafish, were thoroughly evaluated. Untargeted metabolomics based on rat plasma was applied to analyze the metabolic profile changes, revealing the potential action mechanism. The underlying mechanism was furtherly confirmed by gut microbiome analysis and systemic molecular biology experiments. RESULTS 34 prototype chemicals and 71 metabolites from the liver, heart, spleen, lung, kidney, small intestine, uterus, and serum were found. CTP improved the abnormal coagulation system, promoted blood circulation, and reduced pathological damage caused by BSS. Plasma metabolomics revealed that BSS significantly altered bile acid (BA) metabolism and arachidonic acid (AA) metabolism. Gut microbiome analysis and fecal microbiota transplantation (FMT) experiments further demonstrated that CTP modulated the gut microbiota. This modulation promoted BA production and activated endothelial nitric oxide synthase (eNOS), leading to increased nitric oxide (NO) levels. These changes contributed to the therapeutic effect of CTP in removing blood stasis. Systemic molecular biology experiments showed that CTP activated key components of the AA metabolic pathway. It promoted PLCγ1 phosphorylation, increased intracellular Ca2+ levels, and upregulated COX-2 expression. In addition, CTP enhanced the production of AA-related metabolites, including 6-keto-prostaglandin F1α (6-keto-PGF1α), prostaglandin E2 (PGE2), and thromboxane B2 (TXB2). It also increased the transcription of AA metabolism-related genes, such as PLCγ1, PTGS2a, PTGS2b, PTGIS, PTGES, TXBAS, and vWF. CONCLUSIONS CTP could promote the generation of AA metabolites through PLCγ1/Ca2+/COX-2 to stop bleeding, while also enhancing eNOS activity and NO synthesis through gut microbiota-bile acid axis to remove blood stasis. These two effect were balanced to achieve hemostasis without blood stasis.
Collapse
Affiliation(s)
- Xingyong Zhang
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Nian Sheng
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Zhenchang Wang
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Yudan Cao
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Xuan Jiang
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Hui Yan
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Fangfang Cheng
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Ting Geng
- Nanjing University of Chinese Medicine Hanlin College, Taizhou, 225300, China
| | - Kaifeng Wei
- Grade III Laboratory of Molecular Biology of the State Administration of Traditional Chinese Medicine, Experimental Research Platform of Febrile Diseases of Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Li Zhang
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Mingliang Gao
- Key Laboratory of Acupuncture and Medicine Research of Ministry of Education, Nanjing University of Chinese Medicine, Nanjing, 210023, China.
| | - Guisheng Zhou
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, China.
| | - Peidong Chen
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, China.
| |
Collapse
|
2
|
Harrouche K, Boutaoui N, Saidi KM, Stiti MZ, Khelafi A, Khelili S. Design, Synthesis, and Biological Evaluation of New Ureido (Thioureido) Anthranilic Acid Isosteres: Molecular Docking, In Silico ADMET Predictions, and In Vivo Anti-Inflammatory Activity. Chem Biodivers 2025:e202500374. [PMID: 40128116 DOI: 10.1002/cbdv.202500374] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2025] [Revised: 03/24/2025] [Accepted: 03/24/2025] [Indexed: 03/26/2025]
Abstract
A novel series of anthranilic acid isosteres were designed and synthesized as antiinflammatory agents. The in silico absorption, distribution, metabolism, excretion, and toxicity (ADMET) study predicted a favorable pharmacokinetic profile and respect for Lipinski's rule of five. Density functional theory (DFT) calculations revealed an improvement in some target compounds' electronic parameters compared to diclofenac (DCF) and aspirin (ASA), predicting an improvement in their biological activity. Docking investigations demonstrated a strong affinity toward the cyclooxygenase (COX)-1 and COX-2 enzymes, with a relative preference for COX-2, predicting antiinflammatory activity. The MolDock scores were between -140.59 and -91.81 kcal/mol for COX-1 and between -148.10 and -108.9 kcal/mol for COX-2. The experimental pharmacological investigation confirmed these theoretical findings. Indeed, target compounds demonstrated a significant inhibition of the carrageenan-induced paw edema in rats and probable inhibition of COX. Particularly, compounds 4e and 4h devoid of COOH group, which provoke serious gastrointestinal irritation, exhibited antiinflammatory activity comparable to that of salicylic acid (ASA) and surpassed the effectiveness of DCF. Cpmpounds 4e and 4h showed 91.72% inhibition after 3h, against 91.03% and 83.44% for ASA and DCF, respectively, with a greater onset effect, and also surpassing the reference compounds after 1 and 2 h. The results also indicate good pharmacokinetic profile of the target compounds similar to ASA and DCF.
Collapse
Affiliation(s)
- Kamel Harrouche
- Team of Chemistry of Heterocyclic Compounds, Laboratory of Phytochemistry and Pharmacology, University of Jijel, Jijel, Algeria
| | - Nassima Boutaoui
- Team of Chemistry of Heterocyclic Compounds, Laboratory of Phytochemistry and Pharmacology, University of Jijel, Jijel, Algeria
| | - Katia Mohand Saidi
- Team of Pharmaceutical Chemistry, Analysis and Drug Quality Control, Laboratory of Phytochemistry and Pharmacology, University of Jijel, Jijel, Algeria
| | - Mohamed Zakaria Stiti
- Team of Pharmaceutical Chemistry, Analysis and Drug Quality Control, Laboratory of Phytochemistry and Pharmacology, University of Jijel, Jijel, Algeria
| | - Asma Khelafi
- Team of Chemistry of Heterocyclic Compounds, Laboratory of Phytochemistry and Pharmacology, University of Jijel, Jijel, Algeria
| | - Smail Khelili
- Team of Pharmaceutical Chemistry, Analysis and Drug Quality Control, Laboratory of Phytochemistry and Pharmacology, University of Jijel, Jijel, Algeria
| |
Collapse
|
3
|
Yu T, Villalona P, Khan SH, Mikeasky N, Meinert E, Magafas J, Pulahinge T, Bader A, Okafor CD. Enhanced dynamic coupling in a nuclear receptor underlies ligand activity. J Biol Chem 2025; 301:108081. [PMID: 39675705 PMCID: PMC11783427 DOI: 10.1016/j.jbc.2024.108081] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2024] [Revised: 11/14/2024] [Accepted: 12/06/2024] [Indexed: 12/17/2024] Open
Abstract
Bile acids are signaling molecules with critical roles in cholesterol and lipid metabolism, achieved by regulating the transcriptional activity of the farnesoid X receptor (FXR, NR1H4), otherwise known as the bile acid receptor. Modifications to the C6 position of the steroidal core yield bile acid derivatives with 100× improved potency over endogenous bile acids. Prevailing hypotheses suggested increased binding affinity for FXR as the driver for this activity enhancement. Our experimental results contradict this suggestion, motivating us to investigate the underlying mechanisms of enhanced ligand activity. We combined functional assays with over 200 μs of simulations, revealing an unexpected role for helix 5 in the allosteric signaling of obeticholic acid. We uncovered dynamic coupling between adjacent helices 5 and 7, which is uniquely enhanced by the bile acid modification. Ultimately, the enhanced potency of the bile acid analog can be traced to its effect on FXR dynamics. In addition to identifying a previously unknown mechanistic role for helix 5 to helix 7 coupling in FXR, these results emphasize the inextricable linkage between the activity of nuclear receptor ligands and their effects on receptor dynamics.
Collapse
Affiliation(s)
- Tracy Yu
- Department of Biochemistry and Molecular Biology, Pennsylvania State University, University Park, Pennsylvania, USA
| | - Priscilla Villalona
- Department of Biochemistry and Molecular Biology, Pennsylvania State University, University Park, Pennsylvania, USA
| | - Sabab Hasan Khan
- Department of Biochemistry and Molecular Biology, Pennsylvania State University, University Park, Pennsylvania, USA
| | - Noriko Mikeasky
- Department of Biochemistry and Molecular Biology, Pennsylvania State University, University Park, Pennsylvania, USA
| | - Emily Meinert
- Department of Biochemistry and Molecular Biology, Pennsylvania State University, University Park, Pennsylvania, USA
| | - Jill Magafas
- Department of Biochemistry and Molecular Biology, Pennsylvania State University, University Park, Pennsylvania, USA
| | - Thilini Pulahinge
- Department of Chemistry, Pennsylvania State University, University Park, Pennsylvania, USA
| | - Ameen Bader
- Department of Chemistry, Pennsylvania State University, University Park, Pennsylvania, USA
| | - C Denise Okafor
- Department of Biochemistry and Molecular Biology, Pennsylvania State University, University Park, Pennsylvania, USA; Department of Chemistry, Pennsylvania State University, University Park, Pennsylvania, USA.
| |
Collapse
|
4
|
Ronen D, Rokach Y, Abedat S, Qadan A, Daana S, Amir O, Asleh R. Human Gut Microbiota in Cardiovascular Disease. Compr Physiol 2024; 14:5449-5490. [PMID: 39109979 DOI: 10.1002/cphy.c230012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
Abstract
The gut ecosystem, termed microbiota, is composed of bacteria, archaea, viruses, protozoa, and fungi and is estimated to outnumber human cells. Microbiota can affect the host by multiple mechanisms, including the synthesis of metabolites and toxins, modulating inflammation and interaction with other organisms. Advances in understanding commensal organisms' effect on human conditions have also elucidated the importance of this community for cardiovascular disease (CVD). This effect is driven by both direct CV effects and conditions known to increase CV risk, such as obesity, diabetes mellitus (DM), hypertension, and renal and liver diseases. Cardioactive metabolites, such as trimethylamine N -oxide (TMAO), short-chain fatty acids (SCFA), lipopolysaccharides, bile acids, and uremic toxins, can affect atherosclerosis, platelet activation, and inflammation, resulting in increased CV incidence. Interestingly, this interaction is bidirectional with microbiota affected by multiple host conditions including diet, bile acid secretion, and multiple diseases affecting the gut barrier. This interdependence makes manipulating microbiota an attractive option to reduce CV risk. Indeed, evolving data suggest that the benefits observed from low red meat and Mediterranean diet consumption can be explained, at least partially, by the changes that these diets may have on the gut microbiota. In this article, we depict the current epidemiological and mechanistic understanding of the role of microbiota and CVD. Finally, we discuss the potential therapeutic approaches aimed at manipulating gut microbiota to improve CV outcomes. © 2024 American Physiological Society. Compr Physiol 14:5449-5490, 2024.
Collapse
Affiliation(s)
- Daniel Ronen
- Cardiovascular Research Center, Heart Institute, Hadassah Medical Center, Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
| | - Yair Rokach
- Cardiovascular Research Center, Heart Institute, Hadassah Medical Center, Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
| | - Suzan Abedat
- Cardiovascular Research Center, Heart Institute, Hadassah Medical Center, Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
| | - Abed Qadan
- Cardiovascular Research Center, Heart Institute, Hadassah Medical Center, Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
| | - Samar Daana
- Cardiovascular Research Center, Heart Institute, Hadassah Medical Center, Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
| | - Offer Amir
- Cardiovascular Research Center, Heart Institute, Hadassah Medical Center, Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
| | - Rabea Asleh
- Cardiovascular Research Center, Heart Institute, Hadassah Medical Center, Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
| |
Collapse
|
5
|
Zhang Z, Lv T, Wang X, Wu M, Zhang R, Yang X, Fu Y, Liu Z. Role of the microbiota-gut-heart axis between bile acids and cardiovascular disease. Biomed Pharmacother 2024; 174:116567. [PMID: 38583340 DOI: 10.1016/j.biopha.2024.116567] [Citation(s) in RCA: 17] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Revised: 04/01/2024] [Accepted: 04/04/2024] [Indexed: 04/09/2024] Open
Abstract
Bile acid (BA) receptors (e.g., farnesoid X-activated receptor, muscarinic receptor) are expressed in cardiomyocytes, endothelial cells, and vascular smooth muscle cells, indicating the relevance of BAs to cardiovascular disease (CVD). Hydrophobic BAs are cardiotoxic, while hydrophilic BAs are cardioprotective. For example, fetal cardiac insufficiency in maternal intrahepatic cholestasis during pregnancy, and the degree of fetal cardiac abnormality, is closely related to the level of hydrophobic BAs in maternal blood and infant blood. However, ursodeoxycholic acid (the most hydrophilic BA) can reverse/prevent these detrimental effects of increased levels of hydrophobic BAs on the heart. The gut microbiota (GM) and GM metabolites (especially secondary BAs) have crucial roles in hypertension, atherosclerosis, unstable angina, and heart failure. Herein, we describe the relationship between CVD and the GM at the BA level. We combine the concept of the "microbiota-gut-heart axis" (MGHA) and postulate the role and mechanism of BAs in CVD development. In addition, the strategies for treating CVD with BAs under the MGHA are proposed.
Collapse
Affiliation(s)
- Ziyi Zhang
- Department of Cardiovascular Medicine, Affiliated Hospital of Shaoxing University, Shaoxing, Zhejiang, PR China; Department of Pharmacology, School of Medicine, Shaoxing University, Shaoxing, Zhejiang, PR China
| | - Tingting Lv
- Department of Pharmacology, School of Medicine, Shaoxing University, Shaoxing, Zhejiang, PR China; Department of Cardiology, Shaoxing People's Hospital (Shaoxing Hospital, Zhejiang University School of Medicine), Shaoxing, Zhejiang, PR China
| | - Xiang Wang
- Department of Pharmacology, School of Medicine, Shaoxing University, Shaoxing, Zhejiang, PR China
| | - Menglu Wu
- Department of Pharmacology, School of Medicine, Shaoxing University, Shaoxing, Zhejiang, PR China
| | - Ruolin Zhang
- Department of Pharmacology, School of Medicine, Shaoxing University, Shaoxing, Zhejiang, PR China
| | - Xiaopeng Yang
- Department of Pharmacology, School of Medicine, Shaoxing University, Shaoxing, Zhejiang, PR China
| | - Yongping Fu
- Department of Cardiovascular Medicine, Affiliated Hospital of Shaoxing University, Shaoxing, Zhejiang, PR China.
| | - Zheng Liu
- Department of Pharmacology, School of Medicine, Shaoxing University, Shaoxing, Zhejiang, PR China.
| |
Collapse
|
6
|
Bansal S, Li Y, Bansal S, Klotzbier W, Singh B, Jayatilake M, Sridharan V, Fernández JA, Griffin JH, Weiler H, Boerma M, Cheema AK. Genetic Upregulation of Activated Protein C Mitigates Delayed Effects of Acute Radiation Exposure in the Mouse Plasma. Metabolites 2024; 14:245. [PMID: 38786722 PMCID: PMC11122730 DOI: 10.3390/metabo14050245] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Revised: 04/17/2024] [Accepted: 04/22/2024] [Indexed: 05/25/2024] Open
Abstract
Exposure to ionizing radiation, accidental or intentional, may lead to delayed effects of acute radiation exposure (DEARE) that manifest as injury to organ systems, including the kidney, heart, and brain. This study examines the role of activated protein C (APC), a known mitigator of radiation-induced early toxicity, in long-term plasma metabolite and lipid panels that may be associated with DEARE in APCHi mice. The APCHi mouse model used in the study was developed in a C57BL/6N background, expressing the D168F/N173K mouse analog of the hyper-activatable human D167F/D172K protein C variant. This modification enables increased circulating APC levels throughout the mouse's lifetime. Male and female cohorts of C57BL/6N wild-type and APCHi transgenic mice were exposed to 9.5 Gy γ-rays with their hind legs shielded to allow long-term survival that is necessary to monitor DEARE, and plasma was collected at 6 months for LC-MS-based metabolomics and lipidomics. We observed significant dyslipidemia, indicative of inflammatory phenotype, upon radiation exposure. Additionally, observance of several other metabolic dysregulations was suggestive of gut damage, perturbations in TriCarboxylic Acid (TCA) and urea cycles, and arginine metabolism. We also observed gender- and genotype-modulated metabolic perturbations post radiation exposure. The APCHi mice showed near-normal abundance for several lipids. Moreover, restoration of plasma levels of some metabolites, including amino acids, citric acid, and hypoxanthine, in APCHi mice is indicative of APC-mediated protection from radiation injuries. With the help of these findings, the role of APC in plasma molecular events after acute γ-radiation exposure in a gender-specific manner can be established for the first time.
Collapse
Affiliation(s)
- Shivani Bansal
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC 20057, USA; (S.B.); (S.B.); (W.K.); (B.S.); (M.J.)
| | - Yaoxiang Li
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC 20057, USA; (S.B.); (S.B.); (W.K.); (B.S.); (M.J.)
| | - Sunil Bansal
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC 20057, USA; (S.B.); (S.B.); (W.K.); (B.S.); (M.J.)
| | - William Klotzbier
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC 20057, USA; (S.B.); (S.B.); (W.K.); (B.S.); (M.J.)
| | - Baldev Singh
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC 20057, USA; (S.B.); (S.B.); (W.K.); (B.S.); (M.J.)
| | - Meth Jayatilake
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC 20057, USA; (S.B.); (S.B.); (W.K.); (B.S.); (M.J.)
| | - Vijayalakshmi Sridharan
- Division of Radiation Health, Department of Pharmaceutical Sciences, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA; (V.S.); (M.B.)
| | - José A. Fernández
- Department of Molecular Medicine, Scripps Research Institute, La Jolla, CA 92037, USA; (J.A.F.); (J.H.G.)
| | - John H. Griffin
- Department of Molecular Medicine, Scripps Research Institute, La Jolla, CA 92037, USA; (J.A.F.); (J.H.G.)
| | - Hartmut Weiler
- Versiti Blood Research Institute, Medical College of Wisconsin, Milwaukee, WI 53233, USA;
| | - Marjan Boerma
- Division of Radiation Health, Department of Pharmaceutical Sciences, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA; (V.S.); (M.B.)
| | - Amrita K. Cheema
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC 20057, USA; (S.B.); (S.B.); (W.K.); (B.S.); (M.J.)
- Departments of Biochemistry, Molecular and Cellular Biology, Georgetown University Medical Center, Washington, DC 20057, USA
| |
Collapse
|
7
|
Caparrós-Martín JA, Maher P, Ward NC, Saladié M, Agudelo-Romero P, Stick SM, Chan DC, Watts GF, O’Gara F. An Analysis of the Gut Microbiota and Related Metabolites following PCSK9 Inhibition in Statin-Treated Patients with Elevated Levels of Lipoprotein(a). Microorganisms 2024; 12:170. [PMID: 38257996 PMCID: PMC10818477 DOI: 10.3390/microorganisms12010170] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2023] [Revised: 12/26/2023] [Accepted: 01/08/2024] [Indexed: 01/24/2024] Open
Abstract
BACKGROUND Atherosclerotic cardiovascular disease (ASCVD) is a leading cause of global mortality, often associated with high blood levels of LDL cholesterol (LDL-c). Medications like statins and PCSK9 inhibitors, are used to manage LDL-c levels and reduce ASCVD risk. Recent findings connect the gut microbiota and its metabolites to ASCVD development. We showed that statins modulate the gut microbiota including the production of microbial metabolites involved in the regulation of cholesterol metabolism such as short chain fatty acids (SCFAs) and bile acids (BAs). Whether this pleiotropic effect of statins is associated with their antimicrobial properties or it is secondary to the modulation of cholesterol metabolism in the host is unknown. In this observational study, we evaluated whether alirocumab, a PCSK9 inhibitor administered subcutaneously, alters the stool-associated microbiota and the profiles of SCFAs and BAs. METHODS We used stool and plasma collected from patients enrolled in a single-sequence study using alirocumab. Microbial DNA was extracted from stool, and the bacterial component of the gut microbiota profiled following an amplicon sequencing strategy targeting the V3-V4 region of the 16S rRNA gene. Bile acids and SCFAs were profiled and quantified in stool and plasma using mass spectrometry. RESULTS Treatment with alirocumab did not alter bacterial alpha (Shannon index, p = 0.74) or beta diversity (PERMANOVA, p = 0.89) in feces. Similarly, circulating levels of SCFAs (mean difference (95% confidence interval (CI)), 8.12 [-7.15-23.36] µM, p = 0.25) and BAs (mean difference (95% CI), 0.04 [-0.11-0.19] log10(nmol mg-1 feces), p = 0.56) were equivalent regardless of PCSK9 inhibition. Alirocumab therapy was associated with increased concentration of BAs in feces (mean difference (95% CI), 0.20 [0.05-0.34] log10(nmol mg-1 feces), p = 0.01). CONCLUSION In statin-treated patients, the use of alirocumab to inhibit PCSK9 leads to elevated levels of fecal BAs without altering the bacterial population of the gut microbiota. The association of alirocumab with increased fecal BA concentration suggests an additional mechanism for the cholesterol-lowering effect of PCSK9 inhibition.
Collapse
Affiliation(s)
- Jose A. Caparrós-Martín
- Wal-yan Respiratory Research Centre, Telethon Kids Institute, Perth, WA 6009, Australia
- Curtin Health Innovation Research Institute (CHIRI), Curtin University, Perth, WA 6102, Australia
| | - Patrice Maher
- Curtin Health Innovation Research Institute (CHIRI), Curtin University, Perth, WA 6102, Australia
| | - Natalie C. Ward
- Dobney Hypertension Centre, Medical School, The University of Western Australia, Perth, WA 6009, Australia
| | - Montserrat Saladié
- Curtin Health Innovation Research Institute (CHIRI), Curtin University, Perth, WA 6102, Australia
| | - Patricia Agudelo-Romero
- Wal-yan Respiratory Research Centre, Telethon Kids Institute, Perth, WA 6009, Australia
- The University of Western Australia, Perth, WA 6009, Australia
| | - Stephen M. Stick
- Wal-yan Respiratory Research Centre, Telethon Kids Institute, Perth, WA 6009, Australia
- The University of Western Australia, Perth, WA 6009, Australia
- Department of Respiratory Medicine, Princess Margaret Hospital for Children, Perth, WA 6008, Australia
| | - Dick C. Chan
- Medical School, Faculty of Health and Medical Sciences, The University of Western Australia, Perth, WA 6009, Australia
| | - Gerald F. Watts
- Medical School, Faculty of Health and Medical Sciences, The University of Western Australia, Perth, WA 6009, Australia
- Cardiometabolic Service, Departments of Cardiology and Internal Medicine, Royal Perth Hospital, Perth, WA 6000, Australia
| | - Fergal O’Gara
- Wal-yan Respiratory Research Centre, Telethon Kids Institute, Perth, WA 6009, Australia
- Curtin Health Innovation Research Institute (CHIRI), Curtin University, Perth, WA 6102, Australia
- BIOMERIT Research Centre, School of Microbiology, University College Cork, T12 XF62 Cork, Ireland
| |
Collapse
|
8
|
Chattaraj B, Nandi A, Lin WY. Role of the gallbladder in our metabolism and immune system. GALLSTONE FORMATION, DIAGNOSIS, TREATMENT AND PREVENTION 2024:23-38. [DOI: 10.1016/b978-0-443-16098-1.00008-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
|
9
|
Cheraghi S, Babataheri S, Soraya H. The Detrimental Effect of Pre-Treatment with Ivermectin on Myocardial Ischemia. Pharmacology 2023; 109:1-9. [PMID: 37879298 DOI: 10.1159/000534206] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2023] [Accepted: 09/18/2023] [Indexed: 10/27/2023]
Abstract
INTRODUCTION Ivermectin (IVM) is a broad-spectrum anti-parasitic agent with potential antibacterial, antiviral, and anti-cancer effects. There are limited studies on the effects of IVM on cardiovascular diseases, so the present study sought to determine the effects of pre-treatment with IVM on myocardial ischemia in both ex vivo and in vivo. METHODS In the ex vivo part, two groups of control and treated rats with IVM (0.2 mg/kg) were examined for cardiac function and arrhythmias by isolated heart perfusion. In the in vivo part, four groups, namely, control, IVM, Iso (MI), and Iso + IVM 0.2 mg/kg were used. Subcutaneous injection of isoproterenol (100 mg/kg/day) for 2 consecutive days was used for the induction of myocardial infarction (MI) in male Wistar rats. Then electrocardiogram, hemodynamic factors, cardiac hypertrophy, and malondialdehyde (MDA) levels were investigated. RESULTS The ex vivo results showed that administration of IVM induces cardiac arrhythmia and decreases the left ventricular maximal rate of pressure increase (contractility) and maximal rate of pressure decline (relaxation). The isoproterenol-induced MI model used as an in vivo model showed that cardiac hypertrophy were increased with no improvement in the hemodynamic and electrocardiogram pattern in the IVM-treated group in comparison to MI (Iso) group. However, the MDA level was lower in the IVM-treated group. CONCLUSION IVM pre-treatment demonstrates detrimental effects in cardiac ischemia through exacerbation of cardiac arrhythmia, myocardial dysfunction, and increased cardiac hypertrophy. Therefore, the use of IVM in ischemic heart patients should be done with great caution.
Collapse
Affiliation(s)
- Sara Cheraghi
- Department of Pharmacology and Toxicology, School of Pharmacy, Urmia University of Medical Sciences, Urmia, Iran
| | - Shabnam Babataheri
- Department of Pharmacology and Toxicology, School of Pharmacy, Urmia University of Medical Sciences, Urmia, Iran
| | - Hamid Soraya
- Department of Pharmacology and Toxicology, School of Pharmacy, Urmia University of Medical Sciences, Urmia, Iran
| |
Collapse
|
10
|
Jing J, Guo J, Dai R, Zhu C, Zhang Z. Targeting gut microbiota and immune crosstalk: potential mechanisms of natural products in the treatment of atherosclerosis. Front Pharmacol 2023; 14:1252907. [PMID: 37719851 PMCID: PMC10504665 DOI: 10.3389/fphar.2023.1252907] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2023] [Accepted: 08/21/2023] [Indexed: 09/19/2023] Open
Abstract
Atherosclerosis (AS) is a chronic inflammatory reaction that primarily affects large and medium-sized arteries. It is a major cause of cardiovascular disease and peripheral arterial occlusive disease. The pathogenesis of AS involves specific structural and functional alterations in various populations of vascular cells at different stages of the disease. The immune response is involved throughout the entire developmental stage of AS, and targeting immune cells presents a promising avenue for its treatment. Over the past 2 decades, studies have shown that gut microbiota (GM) and its metabolites, such as trimethylamine-N-oxide, have a significant impact on the progression of AS. Interestingly, it has also been reported that there are complex mechanisms of action between GM and their metabolites, immune responses, and natural products that can have an impact on AS. GM and its metabolites regulate the functional expression of immune cells and have potential impacts on AS. Natural products have a wide range of health properties, and researchers are increasingly focusing on their role in AS. Now, there is compelling evidence that natural products provide an alternative approach to improving immune function in the AS microenvironment by modulating the GM. Natural product metabolites such as resveratrol, berberine, curcumin, and quercetin may improve the intestinal microenvironment by modulating the relative abundance of GM, which in turn influences the accumulation of GM metabolites. Natural products can delay the progression of AS by regulating the metabolism of GM, inhibiting the migration of monocytes and macrophages, promoting the polarization of the M2 phenotype of macrophages, down-regulating the level of inflammatory factors, regulating the balance of Treg/Th17, and inhibiting the formation of foam cells. Based on the above, we describe recent advances in the use of natural products that target GM and immune cells crosstalk to treat AS, which may bring some insights to guide the treatment of AS.
Collapse
Affiliation(s)
- Jinpeng Jing
- Graduate School, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Jing Guo
- Graduate School, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Rui Dai
- Graduate School, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Chaojun Zhu
- Institute of TCM Ulcers, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- Surgical Department of Traditional Chinese Medicine, Second Affiliated Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Zhaohui Zhang
- Institute of TCM Ulcers, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- Surgical Department of Traditional Chinese Medicine, Second Affiliated Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
| |
Collapse
|
11
|
Yang Y, Karampoor S, Mirzaei R, Borozdkin L, Zhu P. The interplay between microbial metabolites and macrophages in cardiovascular diseases: A comprehensive review. Int Immunopharmacol 2023; 121:110546. [PMID: 37364331 DOI: 10.1016/j.intimp.2023.110546] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2023] [Revised: 06/11/2023] [Accepted: 06/18/2023] [Indexed: 06/28/2023]
Abstract
The gut microbiome has emerged as a crucial player in developing and progressing cardiovascular diseases (CVDs). Recent studies have highlighted the role of microbial metabolites in modulating immune cell function and their impact on CVD. Macrophages, which have a significant function in the pathogenesis of CVD, are very vulnerable to the effects of microbial metabolites. Microbial metabolites, such as short-chain fatty acids (SCFAs) and trimethylamine-N-oxide (TMAO), have been linked to atherosclerosis and the regulation of immune functions. Butyrate has been demonstrated to reduce monocyte migration and inhibit monocyte attachment to injured endothelial cells, potentially contributing to the attenuation of the inflammatory response and the progression of atherosclerosis. On the other hand, TMAO, another compound generated by gut bacteria, has been linked to atherosclerosis due to its impact on lipid metabolism and the accumulation of cholesterol in macrophages. Indole-3-propionic acid, a tryptophan metabolite produced solely by microbes, has been found to promote the development of atherosclerosis by stimulating macrophage reverse cholesterol transport (RCT) and raising the expression of ABCA1. This review comprehensively discusses how various microbiota-produced metabolites affect macrophage polarization, inflammation, and foam cell formation in CVD. We also highlight the mechanisms underlying these effects and the potential therapeutic applications of targeting microbial metabolites in treating CVD.
Collapse
Affiliation(s)
- Yongzheng Yang
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou, China
| | - Sajad Karampoor
- Gastrointestinal and Liver Diseases Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Rasoul Mirzaei
- Venom and Biotherapeutics Molecules Lab, Medical Biotechnology Department, Biotechnology Research Center, Pasteur Institute of Iran, Tehran, Iran
| | - Leonid Borozdkin
- Department of Maxillofacial Surgery, I. M. Sechenov First Moscow State Medical University (Sechenov University), Moscow, Russia.
| | - Ping Zhu
- Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong 510100, China.
| |
Collapse
|
12
|
Peng J, Fan M, Huang KX, Huang LA, Wang Y, Yin R, Zhao H, Xu S, Li H, Agua A, Xie J, Horne DA, Kandeel F, Huang W, Li J. Design, Synthesis, Computational and Biological Evaluation of Novel Structure Fragments Based on Lithocholic Acid (LCA). Molecules 2023; 28:5332. [PMID: 37513205 PMCID: PMC10383687 DOI: 10.3390/molecules28145332] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2023] [Revised: 06/30/2023] [Accepted: 07/07/2023] [Indexed: 07/30/2023] Open
Abstract
The regulation of bile acid pathways has become a particularly promising therapeutic strategy for a variety of metabolic disorders, cancers, and diseases. However, the hydrophobicity of bile acids has been an obstacle to clinical efficacy due to off-target effects from rapid drug absorption. In this report, we explored a novel strategy to design new structure fragments based on lithocholic acid (LCA) with improved hydrophilicity by introducing a polar "oxygen atom" into the side chain of LCA, then (i) either retaining the carboxylic acid group or replacing the carboxylic acid group with (ii) a diol group or (iii) a vinyl group. These novel fragments were evaluated using luciferase-based reporter assays and the MTS assay. Compared to LCA, the result revealed that the two lead compounds 1a-1b were well tolerated in vitro, maintaining similar potency and efficacy to LCA. The MTS assay results indicated that cell viability was not affected by dose dependence (under 25 µM). Additionally, computational model analysis demonstrated that compounds 1a-1b formed more extensive hydrogen bond networks with Takeda G protein-coupled receptor 5 (TGR5) than LCA. This strategy displayed a potential approach to explore the development of novel endogenous bile acids fragments. Further evaluation on the biological activities of the two lead compounds is ongoing.
Collapse
Affiliation(s)
- Jiangling Peng
- Department of Translational Research and Cellular Therapeutics, Arthur Riggs Diabetes and Metabolism Research Institute, Beckman Research Institute, City of Hope National Medical Center, Duarte, CA 91010, USA
| | - Mingjie Fan
- Department of Diabetes Complications & Metabolism, Arthur Riggs Diabetes and Metabolism Research Institute, Beckman Research Institute, City of Hope National Medical Center, Duarte, CA 91010, USA
| | - Kelly X Huang
- Department of Translational Research and Cellular Therapeutics, Arthur Riggs Diabetes and Metabolism Research Institute, Beckman Research Institute, City of Hope National Medical Center, Duarte, CA 91010, USA
| | - Lina A Huang
- Department of Translational Research and Cellular Therapeutics, Arthur Riggs Diabetes and Metabolism Research Institute, Beckman Research Institute, City of Hope National Medical Center, Duarte, CA 91010, USA
| | - Yangmeng Wang
- Department of Diabetes Complications & Metabolism, Arthur Riggs Diabetes and Metabolism Research Institute, Beckman Research Institute, City of Hope National Medical Center, Duarte, CA 91010, USA
| | - Runkai Yin
- Department of Translational Research and Cellular Therapeutics, Arthur Riggs Diabetes and Metabolism Research Institute, Beckman Research Institute, City of Hope National Medical Center, Duarte, CA 91010, USA
| | - Hanyi Zhao
- Department of Translational Research and Cellular Therapeutics, Arthur Riggs Diabetes and Metabolism Research Institute, Beckman Research Institute, City of Hope National Medical Center, Duarte, CA 91010, USA
| | - Senlin Xu
- Department of Diabetes Complications & Metabolism, Arthur Riggs Diabetes and Metabolism Research Institute, Beckman Research Institute, City of Hope National Medical Center, Duarte, CA 91010, USA
| | - Hongzhi Li
- Department of Molecular Medicine, Beckman Research Institute, City of Hope National Medical Center, Duarte, CA 91010, USA
| | - Alon Agua
- Department of Diabetes Complications & Metabolism, Arthur Riggs Diabetes and Metabolism Research Institute, Beckman Research Institute, City of Hope National Medical Center, Duarte, CA 91010, USA
| | - Jun Xie
- Department of Molecular Medicine, Beckman Research Institute, City of Hope National Medical Center, Duarte, CA 91010, USA
| | - David A Horne
- Department of Molecular Medicine, Beckman Research Institute, City of Hope National Medical Center, Duarte, CA 91010, USA
| | - Fouad Kandeel
- Department of Translational Research and Cellular Therapeutics, Arthur Riggs Diabetes and Metabolism Research Institute, Beckman Research Institute, City of Hope National Medical Center, Duarte, CA 91010, USA
| | - Wendong Huang
- Department of Diabetes Complications & Metabolism, Arthur Riggs Diabetes and Metabolism Research Institute, Beckman Research Institute, City of Hope National Medical Center, Duarte, CA 91010, USA
| | - Junfeng Li
- Department of Translational Research and Cellular Therapeutics, Arthur Riggs Diabetes and Metabolism Research Institute, Beckman Research Institute, City of Hope National Medical Center, Duarte, CA 91010, USA
| |
Collapse
|
13
|
Hepatic and renal improvements with FXR agonist vonafexor in individuals with suspected fibrotic NASH. J Hepatol 2023; 78:479-492. [PMID: 36334688 DOI: 10.1016/j.jhep.2022.10.023] [Citation(s) in RCA: 45] [Impact Index Per Article: 22.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/22/2022] [Revised: 10/04/2022] [Accepted: 10/21/2022] [Indexed: 11/11/2022]
Abstract
BACKGROUND & AIMS The LIVIFY trial investigated the safety, tolerability, and efficacy of vonafexor, a second-generation, non-bile acid farnesoid X receptor agonist in patients with suspected fibrotic non-alcoholic steatohepatitis (NASH). METHODS This double-blind phase IIa study was conducted in two parts. Patients were randomised (1:1:1:1) to receive placebo, vonafexor 100 mg twice daily (VONA-100BID), vonafexor 200 mg once daily (VONA-200QD), or 400 mg vonafexor QD (VONA-400QD) in Part A (safety run-in, pharmacokinetics/pharmacodynamics) or placebo, vonafexor 100 mg QD (VONA-100QD), or VONA-200QD (1:1:1) in Part B. The primary efficacy endpoint was a reduction in liver fat content (LFC) by MRI-proton density fat fraction, while secondary endpoints included reduced corrected T1 values and liver enzymes, from baseline to Week 12. RESULTS One hundred and twenty patients were randomised (Part A, n = 24; Part B, n = 96). In Part B, there was a significant reduction in least-square mean (SE) absolute change in LFC from baseline to Week 12 for VONA-100QD (-6.3% [0.9]) and VONA-200QD (-5.4% [0.9]), vs. placebo (-2.3% [0.9], p = 0.002 and 0.012, respectively). A >30% relative LFC reduction was achieved by 50.0% and 39.3% of patients in the VONA-100QD and VONA-200QD arms, respectively, but only in 12.5% in the placebo arm. Reductions in body weight, liver enzymes, and corrected T1 were also observed with vonafexor. Creatinine-based glomerular filtration rate improved in the active arms but not the placebo arm. Mild to moderate generalised pruritus was reported in 6.3%, 9.7%, and 18.2% of participants in the placebo, VONA-100QD, and VONA-200QD arms, respectively. CONCLUSIONS In patients with suspected fibrotic NASH, vonafexor was safe and induced potent liver fat reduction, improvement in liver enzymes, weight loss, and a possible renal benefit. CLINICAL TRIAL NUMBER (EUDRACT) 2018-003119-22. CLINICALTRIALS GOV IDENTIFIER NCT03812029. IMPACT AND IMPLICATIONS Non-alcoholic steatohepatitis (NASH) has become a leading cause of chronic liver disease worldwide. Affected patients are also at higher risk of developing chronic kidney disease. There are no approved therapies and only few options to treat this population. The phase IIa LIVIFY trial results show that single daily administration of oral vonafexor, an FXR agonist, leads in the short term to a reduction in liver fat, liver enzymes, fibrosis biomarkers, body weight and abdominal circumference, and a possible improvement in kidney function, while possible mild moderate pruritus (a peripheral FXR class effect) and an LDL-cholesterol increase are manageable with lower doses and statins. These results support exploration in longer and larger trials, with the aim of addressing the unmet medical need in NASH.
Collapse
|
14
|
Zhang Q, Zhang L, Chen C, Li P, Lu B. The gut microbiota-artery axis: A bridge between dietary lipids and atherosclerosis? Prog Lipid Res 2023; 89:101209. [PMID: 36473673 DOI: 10.1016/j.plipres.2022.101209] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2022] [Revised: 11/30/2022] [Accepted: 12/02/2022] [Indexed: 12/09/2022]
Abstract
Atherosclerotic cardiovascular disease is one of the major leading global causes of death. Growing evidence has demonstrated that gut microbiota (GM) and its metabolites play a pivotal role in the onset and progression of atherosclerosis (AS), now known as GM-artery axis. There are interactions between dietary lipids and GM, which ultimately affect GM and its metabolites. Given these two aspects, the GM-artery axis may play a mediating role between dietary lipids and AS. Diets rich in saturated fatty acids (SFAs), omega-6 polyunsaturated fatty acids (n-6 PUFAs), industrial trans fatty acids (TFAs), and cholesterol can increase the levels of atherogenic microbes and metabolites, whereas monounsaturated fatty acids (MUFAs), ruminant TFAs, and phytosterols (PS) can increase the levels of antiatherogenic microbes and metabolites. Actually, dietary phosphatidylcholine (PC), sphingomyelin (SM), and omega-3 polyunsaturated fatty acids (n-3 PUFAs) have been demonstrated to affect AS via the GM-artery axis. Therefore, that GM-artery axis acts as a communication bridge between dietary lipids and AS. Herein, we will describe the molecular mechanism of GM-artery axis in AS and discuss the complex interactions between dietary lipids and GM. In particular, we will highlight the evidence and potential mechanisms of dietary lipids affecting AS via GM-artery axis.
Collapse
Affiliation(s)
- Qinjun Zhang
- College of Biosystems Engineering and Food Science, National-Local Joint Engineering Laboratory of Intelligent Food Technology and Equipment, Key Laboratory for Agro-Products Nutritional Evaluation of Ministry of Agriculture and Rural Affairs, Key Laboratory of Agro-Products Postharvest Handling of Ministry of Agriculture and Rural Affairs, Zhejiang Key Laboratory for Agro-Food Processing, Zhejiang International Scientific and Technological Cooperation Base of Health Food Manufacturing and Quality Control, Zhejiang University, Hangzhou, China; Hangzhou Global Scientific and Technological Innovation Center, Zhejiang University, Hangzhou, China
| | - Liangxiao Zhang
- Oil Crops Research Institute, Chinese Academy of Agricultural Sciences, Wubhan, China
| | - Cheng Chen
- Center for Ultrasound Molecular Imaging and Therapeutics, University of Pittsburgh, Pittsburgh, PA, USA
| | - Peiwu Li
- Oil Crops Research Institute, Chinese Academy of Agricultural Sciences, Wubhan, China
| | - Baiyi Lu
- College of Biosystems Engineering and Food Science, National-Local Joint Engineering Laboratory of Intelligent Food Technology and Equipment, Key Laboratory for Agro-Products Nutritional Evaluation of Ministry of Agriculture and Rural Affairs, Key Laboratory of Agro-Products Postharvest Handling of Ministry of Agriculture and Rural Affairs, Zhejiang Key Laboratory for Agro-Food Processing, Zhejiang International Scientific and Technological Cooperation Base of Health Food Manufacturing and Quality Control, Zhejiang University, Hangzhou, China; Hangzhou Global Scientific and Technological Innovation Center, Zhejiang University, Hangzhou, China.
| |
Collapse
|
15
|
Sorption of bile salts from aqueous solutions by MCM-41 silica with chemically immobilized steroid groups. APPLIED NANOSCIENCE 2022. [DOI: 10.1007/s13204-022-02677-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
|
16
|
Shulpekova Y, Zharkova M, Tkachenko P, Tikhonov I, Stepanov A, Synitsyna A, Izotov A, Butkova T, Shulpekova N, Lapina N, Nechaev V, Kardasheva S, Okhlobystin A, Ivashkin V. The Role of Bile Acids in the Human Body and in the Development of Diseases. MOLECULES (BASEL, SWITZERLAND) 2022; 27:molecules27113401. [PMID: 35684337 PMCID: PMC9182388 DOI: 10.3390/molecules27113401] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/14/2022] [Revised: 05/13/2022] [Accepted: 05/23/2022] [Indexed: 11/28/2022]
Abstract
Bile acids are specific and quantitatively important organic components of bile, which are synthesized by hepatocytes from cholesterol and are involved in the osmotic process that ensures the outflow of bile. Bile acids include many varieties of amphipathic acid steroids. These are molecules that play a major role in the digestion of fats and the intestinal absorption of hydrophobic compounds and are also involved in the regulation of many functions of the liver, cholangiocytes, and extrahepatic tissues, acting essentially as hormones. The biological effects are realized through variable membrane or nuclear receptors. Hepatic synthesis, intestinal modifications, intestinal peristalsis and permeability, and receptor activity can affect the quantitative and qualitative bile acids composition significantly leading to extrahepatic pathologies. The complexity of bile acids receptors and the effects of cross-activations makes interpretation of the results of the studies rather difficult. In spite, this is a very perspective direction for pharmacology.
Collapse
Affiliation(s)
- Yulia Shulpekova
- Sechenov First Moscow State Medical University (Sechenov University), 119435 Moscow, Russia; (Y.S.); (M.Z.); (P.T.); (I.T.); (N.L.); (V.N.); (S.K.); (A.O.); (V.I.)
| | - Maria Zharkova
- Sechenov First Moscow State Medical University (Sechenov University), 119435 Moscow, Russia; (Y.S.); (M.Z.); (P.T.); (I.T.); (N.L.); (V.N.); (S.K.); (A.O.); (V.I.)
| | - Pyotr Tkachenko
- Sechenov First Moscow State Medical University (Sechenov University), 119435 Moscow, Russia; (Y.S.); (M.Z.); (P.T.); (I.T.); (N.L.); (V.N.); (S.K.); (A.O.); (V.I.)
| | - Igor Tikhonov
- Sechenov First Moscow State Medical University (Sechenov University), 119435 Moscow, Russia; (Y.S.); (M.Z.); (P.T.); (I.T.); (N.L.); (V.N.); (S.K.); (A.O.); (V.I.)
| | - Alexander Stepanov
- Biobanking Group, Branch of Institute of Biomedical Chemistry “Scientific and Education Center”, 119435 Moscow, Russia; (A.S.); (A.I.); (T.B.)
| | - Alexandra Synitsyna
- Biobanking Group, Branch of Institute of Biomedical Chemistry “Scientific and Education Center”, 119435 Moscow, Russia; (A.S.); (A.I.); (T.B.)
- Correspondence: ; Tel.: +7-499-764-98-78
| | - Alexander Izotov
- Biobanking Group, Branch of Institute of Biomedical Chemistry “Scientific and Education Center”, 119435 Moscow, Russia; (A.S.); (A.I.); (T.B.)
| | - Tatyana Butkova
- Biobanking Group, Branch of Institute of Biomedical Chemistry “Scientific and Education Center”, 119435 Moscow, Russia; (A.S.); (A.I.); (T.B.)
| | | | - Natalia Lapina
- Sechenov First Moscow State Medical University (Sechenov University), 119435 Moscow, Russia; (Y.S.); (M.Z.); (P.T.); (I.T.); (N.L.); (V.N.); (S.K.); (A.O.); (V.I.)
| | - Vladimir Nechaev
- Sechenov First Moscow State Medical University (Sechenov University), 119435 Moscow, Russia; (Y.S.); (M.Z.); (P.T.); (I.T.); (N.L.); (V.N.); (S.K.); (A.O.); (V.I.)
| | - Svetlana Kardasheva
- Sechenov First Moscow State Medical University (Sechenov University), 119435 Moscow, Russia; (Y.S.); (M.Z.); (P.T.); (I.T.); (N.L.); (V.N.); (S.K.); (A.O.); (V.I.)
| | - Alexey Okhlobystin
- Sechenov First Moscow State Medical University (Sechenov University), 119435 Moscow, Russia; (Y.S.); (M.Z.); (P.T.); (I.T.); (N.L.); (V.N.); (S.K.); (A.O.); (V.I.)
| | - Vladimir Ivashkin
- Sechenov First Moscow State Medical University (Sechenov University), 119435 Moscow, Russia; (Y.S.); (M.Z.); (P.T.); (I.T.); (N.L.); (V.N.); (S.K.); (A.O.); (V.I.)
| |
Collapse
|
17
|
Sharma R, Kumar K, Tanvi K. Dealkenylation of neoandrographolide, a phytochemical from Andrographis paniculata stimulates FXR (Farnesoid X Receptor) and enhances gallstone dissolution. J Biomol Struct Dyn 2022; 41:3339-3348. [PMID: 35253613 DOI: 10.1080/07391102.2022.2048078] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
FXR (Farnesoid X Receptor) is one of the nuclear receptors expressed in the liver performing a significant role in the maintenance of bile acid concentration. An imbalance of cholesterol and bile acid ratio due to any undefined reason could cause gallstone formation. Hence, this paper aims to screen phytochemicals that could maintain a requisite balance of cholesterol and bile acid by targeting FXR and thereby contributing to the dissolution of gallstone. Nineteen phytochemicals were selected and queried for Pa and Pi in the way2drug online server for hepatoprotective property, cholesterol synthesis and absorption inhibition property, and β-glucuronidase inhibiting activity. Cianidanol, neoandrographolide, cynarine, saponins, and tanins with satisfying stated properties were docked with the screened FXR (PDB ID- 1OSH) using HADDOCK server, followed by pharmacokinetics study utilizing SwissADME tool. Neoandrographolide fits best among the other selected literature-based phytochemicals with minor violation of 'Brenk's rule'. The violation was corrected with the removal of an alkene group in the provided ChemDraw space of SwissADME. This Dealkenylated compound was further docked with FXR. The promising response under the static condition of the Dealkenylated compound was analyzed for molecular dynamic simulation at physiological conditions for 100 ns. Dealkenylated Neoandrographolide (DN) exhibited hepatoprotective, cholesterol synthesis and absorption inhibition property, and β-glucuronidase inhibition activity with a superior binding score of -42.6+/-1.5 with FXR. The interaction of the FXR receptor and the DN showed exceptional stability at physiological conditions during MD simulation and fit for the ADME properties, therefore it could be a potent candidate to dissolve gallstones.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Rajani Sharma
- Amity Institute of Biotechnology, Amity University Jharkhand, Ranchi, India
| | - Kunal Kumar
- Amity Institute of Biotechnology, Amity University Jharkhand, Ranchi, India
| | - Kumari Tanvi
- Amity Institute of Biotechnology, Amity University Jharkhand, Ranchi, India
| |
Collapse
|
18
|
Kovacevic B, Jones M, Ionescu C, Walker D, Wagle S, Chester J, Foster T, Brown D, Mikov M, Mooranian A, Al-Salami H. The emerging role of bile acids as critical components in nanotechnology and bioengineering: Pharmacology, formulation optimizers and hydrogel-biomaterial applications. Biomaterials 2022; 283:121459. [PMID: 35303546 DOI: 10.1016/j.biomaterials.2022.121459] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Revised: 02/27/2022] [Accepted: 03/04/2022] [Indexed: 12/16/2022]
|
19
|
Fiorucci S, Distrutti E. Linking liver metabolic and vascular disease via bile acid signaling. Trends Mol Med 2021; 28:51-66. [PMID: 34815180 DOI: 10.1016/j.molmed.2021.10.005] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2021] [Revised: 10/22/2021] [Accepted: 10/22/2021] [Indexed: 12/12/2022]
Abstract
Non-alcoholic fatty liver disease (NAFLD) is a metabolic disorder affecting over one quarter of the global population. Liver fat accumulation in NAFLD is promoted by increased de novo lipogenesis leading to the development of a proatherosclerotic lipid profile and atherosclerotic cardiovascular disease (CVD). The CVD component of NAFLD is the main determinant of patient outcome. The farnesoid X receptor (FXR) and the G protein bile acid-activated receptor 1 (GPBAR1) are bile acid-activated receptors that modulate inflammation and lipid and glucose metabolism in the liver and CV system, and are thus potential therapeutic targets. We review bile acid signaling in liver, metabolic tissues, and the CV system, and we propose the development of dual FXR/GPBAR1 ligands, intestine-restricted FXR ligands, or statin combinations to limit side effects and effectively manage the liver and CV components of NAFLD.
Collapse
Affiliation(s)
- Stefano Fiorucci
- Dipartimento di Medicina e Chirurgia, Università di Perugia, Perugia, Italy.
| | - Eleonora Distrutti
- Struttura Complessa di Gastroenterologia ed Epatologia, Azienda Ospedaliera di Perugia, Perugia, Italy
| |
Collapse
|
20
|
Biagioli M, Fiorucci S. Bile acid activated receptors: Integrating immune and metabolic regulation in non-alcoholic fatty liver disease. LIVER RESEARCH 2021; 5:119-141. [PMID: 39957845 PMCID: PMC11791866 DOI: 10.1016/j.livres.2021.08.003] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/08/2021] [Revised: 07/29/2021] [Accepted: 08/27/2021] [Indexed: 02/06/2023]
Abstract
Bile acids are a family of atypical steroids generated at the interface of liver-intestinal microbiota acting on a ubiquitously expressed family of membrane and nuclear receptors known as bile acid activated receptors. The two best characterized receptors of this family are the nuclear receptor, farnesoid X receptor (FXR) and the G protein-coupled receptor, G protein-coupled bile acid receptor 1 (GPBAR1). FXR and GPBAR1 regulate major aspects of lipid and glucose metabolism, energy balance, autophagy and immunity and have emerged as potential pharmaceutical targets for the treatment of metabolic and inflammatory disorders. Clinical trials in non-alcoholic fatty liver disease (NAFLD), however, have shown that selective FXR agonists cause side effects while their efficacy is partial. Because FXR and GPBAR1 exert additive effects, dual FXR/GPBAR1 ligands have been developed for the treatment of metabolic disorders and are currently advanced to clinical trials. Here, we will review the role of FXR and GPBAR1 agonism in NAFLD and how the two receptors could be exploited to target multiple components of the disease.
Collapse
Affiliation(s)
- Michele Biagioli
- Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Stefano Fiorucci
- Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| |
Collapse
|
21
|
Liang C, Zhou XH, Jiao YH, Guo MJ, Meng L, Gong PM, Lyu LZ, Niu HY, Wu YF, Chen SW, Han X, Zhang LW. Ligilactobacillus Salivarius LCK11 Prevents Obesity by Promoting PYY Secretion to Inhibit Appetite and Regulating Gut Microbiota in C57BL/6J Mice. Mol Nutr Food Res 2021; 65:e2100136. [PMID: 34272917 DOI: 10.1002/mnfr.202100136] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2021] [Revised: 05/26/2021] [Indexed: 12/17/2022]
Abstract
SCOPE Obesity is a common disease worldwide and there is an urgent need for strategies to preventing obesity. METHODS AND RESULTS The anti-obesity effect and mechanism of Ligilactobacillus salivarius LCK11 (LCK11) is studied using a C57BL/6J male mouse model in which obesity is induced by a high-fat diet (HFD). Results show that LCK11 can prevent HFD-induced obesity, reflected as inhibited body weight gain, abdominal and liver fat accumulation and dyslipidemia. Analysis of its mechanism shows that on the one hand, LCK11 can inhibit food intake through significantly improving the transcriptional and translational levels of peptide YY (PYY) in the rectum, in addition to the eventual serum PYY level; this is attributed to the activation of the toll-like receptor 2/nuclear factor-κB signaling pathway in enteroendocrine L cells by the peptidoglycan of LCK11. On the other hand, LCK11 supplementation effectively reduces the Firmicutes/Bacteroidetes ratio and shifts the overall structure of the HFD-disrupted gut microbiota toward that of mice fed on a low-fat diet; this also contributes to preventing obesity. CONCLUSION LCK11 shows the potential to be used as a novel probiotic for preventing obesity by both promoting PYY secretion to inhibit food intake and regulating gut microbiota.
Collapse
Affiliation(s)
- Cong Liang
- School of Chemistry and Chemical Engineering, Harbin Institute of Technology, Harbin, 150010, China
| | | | - Yue-Hua Jiao
- Drug safety evaluation center, Heilongjiang University of Chinese Medicine, Harbin, Heilongjiang, 150040, China
| | - Mei-Jie Guo
- Department of Adolescent Medical Clinic, Harbin Children's Hospital, Harbin, 150010, China
| | - Li Meng
- Engineering Research Center of Agricultural Microbiology Technology, Heilongjiang University, Harbin, 150500, China
| | - Pi-Min Gong
- College of Food Science and Engineering, Ocean University of China, Qingdao, 266003, China
| | - Lin-Zheng Lyu
- School of Chemistry and Chemical Engineering, Harbin Institute of Technology, Harbin, 150010, China
| | - Hai-Yue Niu
- School of Chemistry and Chemical Engineering, Harbin Institute of Technology, Harbin, 150010, China
| | - Yi-Fan Wu
- School of Chemistry and Chemical Engineering, Harbin Institute of Technology, Harbin, 150010, China
| | - Shi-Wei Chen
- School of Chemistry and Chemical Engineering, Harbin Institute of Technology, Harbin, 150010, China
| | - Xue Han
- School of Chemistry and Chemical Engineering, Harbin Institute of Technology, Harbin, 150010, China
| | - Lan-Wei Zhang
- College of Food Science and Engineering, Ocean University of China, Qingdao, 266003, China
| |
Collapse
|
22
|
Allaband C, Lingaraju A, Martino C, Russell B, Tripathi A, Poulsen O, Dantas Machado AC, Zhou D, Xue J, Elijah E, Malhotra A, Dorrestein PC, Knight R, Haddad GG, Zarrinpar A. Intermittent Hypoxia and Hypercapnia Alter Diurnal Rhythms of Luminal Gut Microbiome and Metabolome. mSystems 2021; 6:e0011621. [PMID: 34184915 PMCID: PMC8269208 DOI: 10.1128/msystems.00116-21] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2021] [Accepted: 05/07/2021] [Indexed: 12/12/2022] Open
Abstract
Obstructive sleep apnea (OSA), characterized by intermittent hypoxia and hypercapnia (IHC), affects the composition of the gut microbiome and metabolome. The gut microbiome has diurnal oscillations that play a crucial role in regulating circadian and overall metabolic homeostasis. Thus, we hypothesized that IHC adversely alters the gut luminal dynamics of key microbial families and metabolites. The objective of this study was to determine the diurnal dynamics of the fecal microbiome and metabolome of Apoe-/- mice after a week of IHC exposure. Individually housed, 10-week-old Apoe-/- mice on an atherogenic diet were split into two groups. One group was exposed to daily IHC conditions for 10 h (Zeitgeber time 2 [ZT2] to ZT12), while the other was maintained in room air. Six days after the initiation of the IHC conditions, fecal samples were collected every 4 h for 24 h (6 time points). We performed 16S rRNA gene amplicon sequencing and untargeted liquid chromatography-mass spectrometry (LC-MS) to assess changes in the microbiome and metabolome. IHC induced global changes in the cyclical dynamics of the gut microbiome and metabolome. Ruminococcaceae, Lachnospiraceae, S24-7, and Verrucomicrobiaceae had the greatest shifts in their diurnal oscillations. In the metabolome, bile acids, glycerolipids (phosphocholines and phosphoethanolamines), and acylcarnitines were greatly affected. Multi-omic analysis of these results demonstrated that Ruminococcaceae and tauro-β-muricholic acid (TβMCA) cooccur and are associated with IHC conditions and that Coriobacteriaceae and chenodeoxycholic acid (CDCA) cooccur and are associated with control conditions. IHC significantly change the diurnal dynamics of the fecal microbiome and metabolome, increasing members and metabolites that are proinflammatory and proatherogenic while decreasing protective ones. IMPORTANCE People with obstructive sleep apnea are at a higher risk of high blood pressure, type 2 diabetes, cardiac arrhythmias, stroke, and sudden cardiac death. We wanted to understand whether the gut microbiome changes induced by obstructive sleep apnea could potentially explain some of these medical problems. By collecting stool from a mouse model of this disease at multiple time points during the day, we studied how obstructive sleep apnea changed the day-night patterns of microbes and metabolites of the gut. Since the oscillations of the gut microbiome play a crucial role in regulating metabolism, changes in these oscillations can explain why these patients can develop so many metabolic problems. We found changes in microbial families and metabolites that regulate many metabolic pathways contributing to the increased risk for heart disease seen in patients with obstructive sleep apnea.
Collapse
Affiliation(s)
- Celeste Allaband
- Division of Gastroenterology, University of California, San Diego, La Jolla, California, USA
- Biomedical Sciences Graduate Program, University of California, San Diego, La Jolla, California, USA
- Department of Pediatrics, University of California, San Diego, La Jolla, California, USA
| | - Amulya Lingaraju
- Division of Gastroenterology, University of California, San Diego, La Jolla, California, USA
| | - Cameron Martino
- Department of Pediatrics, University of California, San Diego, La Jolla, California, USA
- Bioinformatics and Systems Biology Program, University of California, San Diego, La Jolla, California, USA
- Center for Microbiome Innovation, University of California, San Diego, La Jolla, California, USA
| | - Baylee Russell
- Division of Gastroenterology, University of California, San Diego, La Jolla, California, USA
| | - Anupriya Tripathi
- Department of Pediatrics, University of California, San Diego, La Jolla, California, USA
- Collaborative Mass Spectrometry Innovation Center, Skaggs School of Pharmacy, University of California, San Diego, La Jolla, California, USA
| | - Orit Poulsen
- Department of Pediatrics, University of California, San Diego, La Jolla, California, USA
| | | | - Dan Zhou
- Department of Pediatrics, University of California, San Diego, La Jolla, California, USA
| | - Jin Xue
- Department of Pediatrics, University of California, San Diego, La Jolla, California, USA
| | - Emmanuel Elijah
- Collaborative Mass Spectrometry Innovation Center, Skaggs School of Pharmacy, University of California, San Diego, La Jolla, California, USA
| | - Atul Malhotra
- Center for Circadian Biology, University of California, San Diego, La Jolla, California, USA
| | - Pieter C. Dorrestein
- Biomedical Sciences Graduate Program, University of California, San Diego, La Jolla, California, USA
- Collaborative Mass Spectrometry Innovation Center, Skaggs School of Pharmacy, University of California, San Diego, La Jolla, California, USA
- Center for Microbiome Innovation, University of California, San Diego, La Jolla, California, USA
| | - Rob Knight
- Biomedical Sciences Graduate Program, University of California, San Diego, La Jolla, California, USA
- Department of Pediatrics, University of California, San Diego, La Jolla, California, USA
- Center for Microbiome Innovation, University of California, San Diego, La Jolla, California, USA
- Department of Computer Science and Engineering, University of California, San Diego, La Jolla, California, USA
| | - Gabriel G. Haddad
- Biomedical Sciences Graduate Program, University of California, San Diego, La Jolla, California, USA
- Department of Pediatrics, University of California, San Diego, La Jolla, California, USA
- Department of Neuroscience, University of California, San Diego, La Jolla, California, USA
- Center for Microbiome Innovation, University of California, San Diego, La Jolla, California, USA
| | - Amir Zarrinpar
- Division of Gastroenterology, University of California, San Diego, La Jolla, California, USA
- Biomedical Sciences Graduate Program, University of California, San Diego, La Jolla, California, USA
- Center for Microbiome Innovation, University of California, San Diego, La Jolla, California, USA
- Institute of Diabetes and Metabolic Health, University of California, San Diego, La Jolla, California, USA
- Center for Circadian Biology, University of California, San Diego, La Jolla, California, USA
- VA Health Sciences San Diego, La Jolla, California, USA
| |
Collapse
|
23
|
Fiorucci S, Biagioli M, Baldoni M, Ricci P, Sepe V, Zampella A, Distrutti E. The identification of farnesoid X receptor modulators as treatment options for nonalcoholic fatty liver disease. Expert Opin Drug Discov 2021; 16:1193-1208. [PMID: 33849361 DOI: 10.1080/17460441.2021.1916465] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
INTRODUCTION The farnesoid-x-receptor (FXR) is a ubiquitously expressed nuclear receptor selectively activated by primary bile acids. AREA COVERED FXR is a validated pharmacological target. Herein, the authors review preclinical and clinical data supporting the development of FXR agonists in the treatment of nonalcoholic fatty liver disease. EXPERT OPINION Development of systemic FXR agonists to treat the metabolic liver disease has been proven challenging because the side effects associated with these agents including increased levels of cholesterol and LDL-c and reduced HDL-c raising concerns over their long-term cardiovascular safety. Additionally, pruritus has emerged as a common, although poorly explained, dose-related side effect with all FXR ligands, but is especially common with OCA. FXR agonists that are currently undergoing phase 2/3 trials are cilofexor, tropifexor, nidufexor and MET409. Some of these agents are currently being developed as combination therapies with other agents including cenicriviroc, a CCR2/CCR5 inhibitor, or firsocostat an acetyl CoA carboxylase inhibitor. Additional investigations are needed to evaluate the beneficial effects of combination of these agents with statins. It is expected that in the coming years, FXR agonists will be developed as a combination therapy to minimize side effects and increase likelihood of success by targeting different metabolic pathways.
Collapse
Affiliation(s)
- Stefano Fiorucci
- Dipartimento Di Medicina E Chirurgia, Università Di Perugia, Perugia, Italy
| | - Michele Biagioli
- Dipartimento Di Medicina E Chirurgia, Università Di Perugia, Perugia, Italy
| | - Monia Baldoni
- Dipartimento Di Medicina E Chirurgia, Università Di Perugia, Perugia, Italy
| | - Patrizia Ricci
- Dipartimento Di Medicina E Chirurgia, Università Di Perugia, Perugia, Italy
| | - Valentina Sepe
- Department of Pharmacy University of Napoli, Federico II, Napoli, Italy
| | - Angela Zampella
- Department of Pharmacy University of Napoli, Federico II, Napoli, Italy
| | - Eleonora Distrutti
- SC Di Gastroenterologia Ed Epatologia, Azienda Ospedaliera Di Perugia, Perugia, Italy
| |
Collapse
|
24
|
Mannino G, Iovino P, Lauria A, Genova T, Asteggiano A, Notarbartolo M, Porcu A, Serio G, Chinigò G, Occhipinti A, Capuzzo A, Medana C, Munaron L, Gentile C. Bioactive Triterpenes of Protium heptaphyllum Gum Resin Extract Display Cholesterol-Lowering Potential. Int J Mol Sci 2021; 22:ijms22052664. [PMID: 33800828 PMCID: PMC7961947 DOI: 10.3390/ijms22052664] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Revised: 02/25/2021] [Accepted: 03/02/2021] [Indexed: 12/21/2022] Open
Abstract
Hypercholesterolemia is one of the major causes of cardiovascular disease, the risk of which is further increased if other forms of dyslipidemia occur. Current therapeutic strategies include changes in lifestyle coupled with drug administration. Statins represent the most common therapeutic approach, but they may be insufficient due to the onset of resistance mechanisms and side effects. Consequently, patients with mild hypercholesterolemia prefer the use of food supplements since these are perceived to be safer. Here, we investigate the phytochemical profile and cholesterol-lowering potential of Protium heptaphyllum gum resin extract (PHE). Chemical characterization via HPLC-APCI-HRMS2 and GC-FID/MS identified 13 compounds mainly belonging to ursane, oleanane, and tirucallane groups. Studies on human hepatocytes have revealed how PHE is able to reduce cholesterol production and regulate the expression of proteins involved in its metabolism. (HMGCR, PCSK9, LDLR, FXR, IDOL, and PPAR). Moreover, measuring the inhibitory activity of PHE against HMGR, moderate inhibition was recorded. Finally, molecular docking studies identified acidic tetra- and pentacyclic triterpenoids as the main compounds responsible for this action. In conclusion, our study demonstrates how PHE may be a useful alternative to contrast hypercholesterolemia, highlighting its potential as a sustainable multitarget natural extract for the nutraceutical industry that is rapidly gaining acceptance as a source of health-promoting compounds.
Collapse
Affiliation(s)
- Giuseppe Mannino
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies (STEBICEF), University of Palermo, 90128 Palermo, Italy; (G.M.); (A.L.); (M.N.); (G.S.)
| | - Piera Iovino
- Biosfered S.R.L., 10148 Turin, Italy; (P.I.); (A.A.)
| | - Antonino Lauria
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies (STEBICEF), University of Palermo, 90128 Palermo, Italy; (G.M.); (A.L.); (M.N.); (G.S.)
| | - Tullio Genova
- Department of Life Sciences and Systems Biology, University of Turin, 10123 Turin, Italy; (T.G.); (G.C.); (L.M.)
| | - Alberto Asteggiano
- Biosfered S.R.L., 10148 Turin, Italy; (P.I.); (A.A.)
- Department of Molecular Biotechnology and Health Sciences, University of Torino, 10125 Torino, Italy (C.M.)
| | - Monica Notarbartolo
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies (STEBICEF), University of Palermo, 90128 Palermo, Italy; (G.M.); (A.L.); (M.N.); (G.S.)
| | - Alessandra Porcu
- Abel Nutraceuticals S.R.L., 10148 Turin, Italy; (A.P.); (A.O.); (A.C.)
| | - Graziella Serio
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies (STEBICEF), University of Palermo, 90128 Palermo, Italy; (G.M.); (A.L.); (M.N.); (G.S.)
| | - Giorgia Chinigò
- Department of Life Sciences and Systems Biology, University of Turin, 10123 Turin, Italy; (T.G.); (G.C.); (L.M.)
| | - Andrea Occhipinti
- Abel Nutraceuticals S.R.L., 10148 Turin, Italy; (A.P.); (A.O.); (A.C.)
| | - Andrea Capuzzo
- Abel Nutraceuticals S.R.L., 10148 Turin, Italy; (A.P.); (A.O.); (A.C.)
| | - Claudio Medana
- Department of Molecular Biotechnology and Health Sciences, University of Torino, 10125 Torino, Italy (C.M.)
| | - Luca Munaron
- Department of Life Sciences and Systems Biology, University of Turin, 10123 Turin, Italy; (T.G.); (G.C.); (L.M.)
| | - Carla Gentile
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies (STEBICEF), University of Palermo, 90128 Palermo, Italy; (G.M.); (A.L.); (M.N.); (G.S.)
- Correspondence: ; Tel.: +39-091-2388-6472
| |
Collapse
|
25
|
Fiorucci S, Distrutti E, Carino A, Zampella A, Biagioli M. Bile acids and their receptors in metabolic disorders. Prog Lipid Res 2021; 82:101094. [PMID: 33636214 DOI: 10.1016/j.plipres.2021.101094] [Citation(s) in RCA: 153] [Impact Index Per Article: 38.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2020] [Revised: 02/03/2021] [Accepted: 02/12/2021] [Indexed: 02/08/2023]
Abstract
Bile acids are a large family of atypical steroids which exert their functions by binding to a family of ubiquitous cell membrane and nuclear receptors. There are two main bile acid activated receptors, FXR and GPBAR1, that are exclusively activated by bile acids, while other receptors CAR, LXRs, PXR, RORγT, S1PR2and VDR are activated by bile acids in addition to other more selective endogenous ligands. In the intestine, activation of FXR and GPBAR1 promotes the release of FGF15/19 and GLP1 which integrate their signaling with direct effects exerted by theother receptors in target tissues. This network is tuned in a time ordered manner by circadian rhythm and is critical for the regulation of metabolic process including autophagy, fast-to-feed transition, lipid and glucose metabolism, energy balance and immune responses. In the last decade FXR ligands have entered clinical trials but development of systemic FXR agonists has been proven challenging because their side effects including increased levels of cholesterol and Low Density Lipoproteins cholesterol (LDL-c) and reduced High-Density Lipoprotein cholesterol (HDL-c). In addition, pruritus has emerged as a common, dose related, side effect of FXR ligands. Intestinal-restricted FXR and GPBAR1 agonists and dual FXR/GPBAR1 agonists have been developed. Here we review the last decade in bile acids physiology and pharmacology.
Collapse
Affiliation(s)
- Stefano Fiorucci
- Dipartimento di Medicina e Chirurgia, Università di Perugia, Perugia, Italy.
| | - Eleonora Distrutti
- SC di Gastroenterologia ed Epatologia, Azienda Ospedaliera di Perugia, Perugia, Italy
| | - Adriana Carino
- Dipartimento di Medicina e Chirurgia, Università di Perugia, Perugia, Italy
| | - Angela Zampella
- Department of Pharmacy, University of Napoli, Federico II, Napoli, Italy
| | - Michele Biagioli
- Dipartimento di Medicina e Chirurgia, Università di Perugia, Perugia, Italy
| |
Collapse
|
26
|
Zhang R, Ma WQ, Fu MJ, Li J, Hu CH, Chen Y, Zhou MM, Gao ZJ, He YL. Overview of bile acid signaling in the cardiovascular system. World J Clin Cases 2021; 9:308-320. [PMID: 33521099 PMCID: PMC7812903 DOI: 10.12998/wjcc.v9.i2.308] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/06/2020] [Revised: 09/28/2020] [Accepted: 10/20/2020] [Indexed: 02/06/2023] Open
Abstract
Bile acids (BAs) are classically known to play a vital role in the metabolism of lipids and in absorption. It is now well established that BAs act as signaling molecules, activating different receptors (such as farnesoid X receptor, vitamin D receptor, Takeda G-protein-coupled receptor 5, sphingosine-1-phosphate, muscarinic receptors, and big potassium channels) and participating in the regulation of energy homeostasis and lipid and glucose metabolism. In addition, increased BAs can impair cardiovascular function in liver cirrhosis. Approximately 50% of patients with cirrhosis develop cirrhotic cardiomyopathy. Exposure to high concentrations of hydrophobic BAs has been shown to be related to adverse effects with respect to vascular tension, endothelial function, arrhythmias, coronary atherosclerotic heart disease, and heart failure. The BAs in the serum BA pool have relevant through their hydrophobicity, and the lipophilic BAs are more harmful to the heart. Interestingly, ursodeoxycholic acid is a hydrophilic BA, and it is used as a therapeutic drug to reverse and protect the harmful cardiac effects caused by hydrophobic elevated BAs. In order to elucidate the mechanism of BAs and cardiovascular function, abundant experiments have been conducted in vitro and in vivo. The aim of this review was to explore the mechanism of BAs in the cardiovascular system.
Collapse
Affiliation(s)
- Rou Zhang
- Department of Infectious Diseases, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an 710061, Shaanxi Province, China
| | - Wen-Qi Ma
- Department of Infectious Diseases, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an 710061, Shaanxi Province, China
| | - Meng-Jun Fu
- Department of Infectious Diseases, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an 710061, Shaanxi Province, China
| | - Juan Li
- Department of Infectious Diseases, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an 710061, Shaanxi Province, China
| | - Chun-Hua Hu
- Department of Infectious Diseases, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an 710061, Shaanxi Province, China
| | - Yi Chen
- Department of Infectious Diseases, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an 710061, Shaanxi Province, China
| | - Mi-Mi Zhou
- Department of Infectious Diseases, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an 710061, Shaanxi Province, China
| | - Zhi-Jie Gao
- Department of Infectious Diseases, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an 710061, Shaanxi Province, China
| | - Ying-Li He
- Department of Infectious Diseases, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an 710061, Shaanxi Province, China
| |
Collapse
|
27
|
Abstract
Nonalcoholic fatty liver disease (NAFLD) is considered the hepatic manifestation of the metabolic syndrome (MetS) and comprises one of the largest health threats of the twenty-first century. In this chapter, we review the current state of knowledge of NAFLD and underline the striking similarities with atherosclerosis. We first describe current epidemiological data showing the staggering increase of NAFLD numbers and its related clinical and economic costs. We then provide an overview of pathophysiological hepatic processes in NAFLD and highlight the systemic aspects of NAFLD that point toward metabolic crosstalk between organs as an important cause of metabolic disease. Finally, we end by highlighting the currently investigated therapeutic approaches for NAFLD, which also show strong similarities with a range of treatment options for atherosclerosis.
Collapse
|
28
|
Zhang BC, Chen JH, Xiang CH, Su MY, Zhang XS, Ma YF. Increased serum bile acid level is associated with high-risk coronary artery plaques in an asymptomatic population detected by coronary computed tomography angiography. J Thorac Dis 2019; 11:5063-5070. [PMID: 32030222 DOI: 10.21037/jtd.2019.12.16] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Background There are limited data on the association between serum total bile acid level and coronary plaque characteristics. This study investigated the relationship between serum total bile acid level and the severity of coronary stenosis and coronary plaque features in an asymptomatic population using coronary computed tomography angiography (CTA). Methods A total of 1,137 consecutive participants with no known coronary artery disease (CAD) undergoing CTA as part of a general routine health evaluation were recruited. Serum total bile acid level and clinical parameters were assayed. Coronary stenosis and high-risk plaques features (napkin-ring sign, low-attenuation plaque, spotty calcification, positive remodelling) were evaluated. Associations between serum total bile acid concentration and high-risk coronary plaques was tested through univariate and multivariate analyses. Results A total of 101 high-risk coronary plaques subjects and 93 controls were eligible for study inclusion. The severity of coronary artery stenosis and high-risk coronary plaques increased with serum total bile acid level quartiles (all P<0.001). The independent predictor of high-risk coronary plaques in multivariate analysis was serum total bile acid level (P<0.001). Receiver operating characteristic (ROC) confirmed that serum total bile acid concentration significantly differentiated high-risk coronary plaques [the area under the curve (AUC) =0.876; P<0.001, with a sensitivity of 87.13% and a specificity of 86.02%]. Conclusions Higher serum total bile acid level was associated with the severity of coronary artery stenosis and high-risk coronary artery plaques detected by CTA in asymptomatic populations.
Collapse
Affiliation(s)
- Bu-Chun Zhang
- Department of Cardiology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou 221000, China
| | - Jun-Hong Chen
- Department of Cardiology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou 221000, China
| | - Chu-Han Xiang
- Department of Cardiology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou 221000, China
| | - Ming-Yu Su
- Department of Cardiology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou 221000, China
| | - Xue-Shan Zhang
- Department of Cardiology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou 221000, China
| | - Yan-Feng Ma
- Department of Cardiology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou 221000, China
| |
Collapse
|
29
|
Deng JN, Li Q, Sun K, Pan CS, Li H, Fan JY, Li G, Hu BH, Chang X, Han JY. Cardiotonic Pills Plus Recombinant Human Prourokinase Ameliorates Atherosclerotic Lesions in LDLR -/- Mice. Front Physiol 2019; 10:1128. [PMID: 31551808 PMCID: PMC6747059 DOI: 10.3389/fphys.2019.01128] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2018] [Accepted: 08/15/2019] [Indexed: 11/24/2022] Open
Abstract
Aim This study was to explore the protective effects of cardiotonic pills (CP) or/and recombinant human prourokinase (proUK)on the atherosclerosis and the potential underlying mechanism. Methods and Results Atherosclerosis was induced in LDLR–/– mice by high fat diet contained 20% lard and 0.5% cholesterol. Daily oral administration of CP (130 mg/kg) or/and intravenous injection of proUK (2.5 mg/kg, twice a week) began at 8 weeks after feeding with high fat diet and continued for 4 weeks. CP alone treatment markedly decreased plasma triglyceride, but did not ameliorate atherosclerosis plaque. No effect was observed for proUK alone on any endpoints tested. CP plus proUK induced a significantly reduction in the atherosclerotic lesions, along with decreased levels of total cholesterol, triglyceride in the plasma. CP plus proUK inhibited the elevated hepatic total cholesterol and triglyceride in high fat diet-fed LDLR–/– mice, up-regulating the expressions of ATP-binding cassette gene 5 and 8, and adipose triglyceride lipase. In the aorta, CP plus proUK inhibited the expression of scavenger receptor A and CD36 in LDLR–/– mice. In addition, we observed that systemic inflammation was inhibited, manifested downregulation of plasma macrophage inflammatory protein-1α and intercellular cell adhesion molecule-1. Conclusion CP plus proUK effectively attenuated atherosclerosis plaque in LDLR–/– mice, which is associated with normalizing the lipid metabolism in the liver and aorta, reducing phagocytosis of receptor-mediated modified-LDL uptake and inhibiting systemic inflammation.
Collapse
Affiliation(s)
- Jing-Na Deng
- Department of Integration of Chinese and Western Medicine, School of Basic Medical Sciences, Peking University, Beijing, China.,Tasly Microcirculation Research Center, Peking University Health Science Center, Beijing, China.,State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.,Key Laboratory of Microcirculation, State Administration of Traditional Chinese Medicine of the People's Republic of China, Beijing, China.,Key Laboratory of Stasis and Phlegm, State Administration of Traditional Chinese Medicine of the People's Republic of China, Beijing, China.,State Key Laboratory of Core Technology in Innovative Chinese Medicine, Tianjin, China
| | - Quan Li
- Tasly Microcirculation Research Center, Peking University Health Science Center, Beijing, China.,State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.,Key Laboratory of Microcirculation, State Administration of Traditional Chinese Medicine of the People's Republic of China, Beijing, China.,Key Laboratory of Stasis and Phlegm, State Administration of Traditional Chinese Medicine of the People's Republic of China, Beijing, China.,State Key Laboratory of Core Technology in Innovative Chinese Medicine, Tianjin, China
| | - Kai Sun
- Tasly Microcirculation Research Center, Peking University Health Science Center, Beijing, China.,State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.,Key Laboratory of Microcirculation, State Administration of Traditional Chinese Medicine of the People's Republic of China, Beijing, China.,Key Laboratory of Stasis and Phlegm, State Administration of Traditional Chinese Medicine of the People's Republic of China, Beijing, China.,State Key Laboratory of Core Technology in Innovative Chinese Medicine, Tianjin, China
| | - Chun-Shui Pan
- Tasly Microcirculation Research Center, Peking University Health Science Center, Beijing, China.,State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.,Key Laboratory of Microcirculation, State Administration of Traditional Chinese Medicine of the People's Republic of China, Beijing, China.,Key Laboratory of Stasis and Phlegm, State Administration of Traditional Chinese Medicine of the People's Republic of China, Beijing, China.,State Key Laboratory of Core Technology in Innovative Chinese Medicine, Tianjin, China
| | - Huan Li
- Department of Integration of Chinese and Western Medicine, School of Basic Medical Sciences, Peking University, Beijing, China.,Tasly Microcirculation Research Center, Peking University Health Science Center, Beijing, China.,State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.,Key Laboratory of Microcirculation, State Administration of Traditional Chinese Medicine of the People's Republic of China, Beijing, China.,Key Laboratory of Stasis and Phlegm, State Administration of Traditional Chinese Medicine of the People's Republic of China, Beijing, China.,State Key Laboratory of Core Technology in Innovative Chinese Medicine, Tianjin, China
| | - Jing-Yu Fan
- Tasly Microcirculation Research Center, Peking University Health Science Center, Beijing, China.,State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.,Key Laboratory of Microcirculation, State Administration of Traditional Chinese Medicine of the People's Republic of China, Beijing, China.,Key Laboratory of Stasis and Phlegm, State Administration of Traditional Chinese Medicine of the People's Republic of China, Beijing, China.,State Key Laboratory of Core Technology in Innovative Chinese Medicine, Tianjin, China
| | - Gao Li
- Department of Oncology, Guizhou University of Chinese Medicine, Guiyang, China
| | - Bai-He Hu
- Tasly Microcirculation Research Center, Peking University Health Science Center, Beijing, China.,State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.,Key Laboratory of Microcirculation, State Administration of Traditional Chinese Medicine of the People's Republic of China, Beijing, China.,Key Laboratory of Stasis and Phlegm, State Administration of Traditional Chinese Medicine of the People's Republic of China, Beijing, China.,State Key Laboratory of Core Technology in Innovative Chinese Medicine, Tianjin, China
| | - Xin Chang
- Tasly Microcirculation Research Center, Peking University Health Science Center, Beijing, China.,State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.,Key Laboratory of Microcirculation, State Administration of Traditional Chinese Medicine of the People's Republic of China, Beijing, China.,Key Laboratory of Stasis and Phlegm, State Administration of Traditional Chinese Medicine of the People's Republic of China, Beijing, China.,State Key Laboratory of Core Technology in Innovative Chinese Medicine, Tianjin, China
| | - Jing-Yan Han
- Department of Integration of Chinese and Western Medicine, School of Basic Medical Sciences, Peking University, Beijing, China.,Tasly Microcirculation Research Center, Peking University Health Science Center, Beijing, China.,State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.,Key Laboratory of Microcirculation, State Administration of Traditional Chinese Medicine of the People's Republic of China, Beijing, China.,Key Laboratory of Stasis and Phlegm, State Administration of Traditional Chinese Medicine of the People's Republic of China, Beijing, China.,State Key Laboratory of Core Technology in Innovative Chinese Medicine, Tianjin, China
| |
Collapse
|
30
|
Xia Y, Zhang F, Zhao S, Li Y, Chen X, Gao E, Xu X, Xiong Z, Zhang X, Zhang J, Zhao H, Wang W, Wang H, Guo Y, Liu Y, Li C, Wang S, Zhang L, Yan W, Tao L. Adiponectin determines farnesoid X receptor agonism-mediated cardioprotection against post-infarction remodelling and dysfunction. Cardiovasc Res 2019; 114:1335-1349. [PMID: 29668847 DOI: 10.1093/cvr/cvy093] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/12/2017] [Accepted: 04/12/2018] [Indexed: 12/16/2022] Open
Abstract
Aims The farnesoid X receptor (FXR) is a member of the metabolic nuclear receptor superfamily that plays a critical regulatory role in cardiovascular physiology/pathology. However, the role of systemic FXR activation in the chronic phase in myocardial infarction (MI)-induced cardiac remodelling and dysfunction remains unclear. In this study, we aimed to elucidate the role of long-term FXR activation on post-MI cardiac remodelling and dysfunction. Methods and results Mice underwent either MI surgery or sham operation. At 1 week after MI, both sham and MI mice were gavaged with 25 mg/kg/d of a synthetic FXR agonist (GW4064) or a vehicle control for 7 weeks, and cardiac performance was assessed by consecutive echocardiography studies. Administration of GW4064 significantly increased left ventricular ejection fraction at 4 weeks and 8 weeks after MI (both P < 0.01). Moreover, GW4064 treatment increased angiogenesis and mitochondrial biogenesis, reduced cardiomyocyte loss and inflammation, and ameliorated cardiac remodelling as evidenced by heart weight, lung weight, atrial natriuretic peptide/brain natriuretic peptide levels, and myocardial fibrosis at 8 weeks post-MI. At the molecular level, GW4064 significantly increased FXR mRNA expression and transcriptional activity in heart tissue. Moreover, over-expression of myocardial FXR failed to exert significant cardioprotection in vivo, indicating that GW4064 improved post-MI heart remodelling and function independent of myocardial FXR expression/activity. Among the four down-stream soluble molecules of FXR, plasma adiponectin was most significantly increased by GW4064. In cultured adipocytes, GW4064 increased mRNA levels and protein expression of adiponectin. Conditioned medium of GW4064-treated adipocytes activated AMPK-PGC-1α signalling and reduced hypoxia-induced cardiomyocyte apoptosis, all of which were attenuated by an adiponectin neutralizing anti-body. More importantly, when knocking-out adiponectin in mice, the cardioprotective effects of GW4064 were attenuated. Conclusions We are the first to show that FXR agonism ameliorated post-MI cardiac dysfunction and remodelling by stimulating adiponectin secretion. Thus, we demonstrated that FXR agonism is a potential therapeutic strategy in post-MI heart failure.
Collapse
Affiliation(s)
- Yunlong Xia
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, 127 West Changle Road, Xi'an 710032, China
| | - Fuyang Zhang
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, 127 West Changle Road, Xi'an 710032, China
| | - Shihao Zhao
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, 127 West Changle Road, Xi'an 710032, China.,Department of Cardiology, Hainan Branch of PLA General Hospital, Sanya 572013, China
| | - Yueyang Li
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, 127 West Changle Road, Xi'an 710032, China
| | - Xiyao Chen
- Department of Geriatrics, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, China
| | - Erhe Gao
- Center for Translational Medicine, Temple University, Philadelphia, PA 19140, USA
| | - Xinyue Xu
- State Key Laboratory of Military Stomatology and National Clinical Research Center for Oral Diseases, Department of Periodontology, School of Stomatology, Fourth Military Medical University, Xi'an 710032, China
| | - Zhenyu Xiong
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, 127 West Changle Road, Xi'an 710032, China
| | - Xiaomeng Zhang
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, 127 West Changle Road, Xi'an 710032, China
| | - Jinglong Zhang
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, 127 West Changle Road, Xi'an 710032, China
| | - Huishou Zhao
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, 127 West Changle Road, Xi'an 710032, China
| | - Wei Wang
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, 127 West Changle Road, Xi'an 710032, China
| | - Helin Wang
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, 127 West Changle Road, Xi'an 710032, China
| | - Yanjie Guo
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, 127 West Changle Road, Xi'an 710032, China
| | - Yi Liu
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, 127 West Changle Road, Xi'an 710032, China
| | - Congye Li
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, 127 West Changle Road, Xi'an 710032, China
| | - Shan Wang
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, 127 West Changle Road, Xi'an 710032, China
| | - Ling Zhang
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, 127 West Changle Road, Xi'an 710032, China
| | - Wenjun Yan
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, 127 West Changle Road, Xi'an 710032, China
| | - Ling Tao
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, 127 West Changle Road, Xi'an 710032, China
| |
Collapse
|
31
|
Anlu W, Dongcheng C, He Z, Qiuyi L, Yan Z, Yu Q, Hao X, Keji C. Using herbal medicine to target the “microbiota-metabolism-immunity” axis as possible therapy for cardiovascular disease. Pharmacol Res 2019; 142:205-222. [DOI: 10.1016/j.phrs.2019.02.018] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/21/2018] [Revised: 02/18/2019] [Accepted: 02/18/2019] [Indexed: 02/08/2023]
|
32
|
Circulating primary bile acid is correlated with structural remodeling in atrial fibrillation. J Interv Card Electrophysiol 2019; 57:371-377. [DOI: 10.1007/s10840-019-00540-z] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/03/2018] [Accepted: 03/14/2019] [Indexed: 10/27/2022]
|
33
|
Di Ciaula A, Wang DQH, Portincasa P. Cholesterol cholelithiasis: part of a systemic metabolic disease, prone to primary prevention. Expert Rev Gastroenterol Hepatol 2019; 13:157-171. [PMID: 30791781 DOI: 10.1080/17474124.2019.1549988] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/26/2018] [Accepted: 11/15/2018] [Indexed: 02/07/2023]
Abstract
Cholesterol gallstone disease have relationships with various conditions linked with insulin resistance, but also with heart disease, atherosclerosis, and cancer. These associations derive from mechanisms active at a local (i.e. gallbladder, bile) and a systemic level and are involved in inflammation, hormones, nuclear receptors, signaling molecules, epigenetic modulation of gene expression, and gut microbiota. Despite advanced knowledge of these pathways, the available therapeutic options for symptomatic gallstone patients remain limited. Therapy includes oral litholysis by the bile acid ursodeoxycholic acid (UDCA) in a small subgroup of patients at high risk of postdissolution recurrence, or laparoscopic cholecystectomy, which is the therapeutic radical gold standard treatment. Cholecystectomy, however, may not be a neutral event, and potentially generates health problems, including the metabolic syndrome. Areas covered: Several studies on risk factors and pathogenesis of cholesterol gallstone disease, acting at a systemic level have been reviewed through a PubMed search. Authors have focused on primary prevention and novel potential therapeutic strategies. Expert commentary: The ultimate goal appears to target the manageable systemic mechanisms responsible for gallstone occurrence, pointing to primary prevention measures. Changes must target lifestyles, as well as experimenting innovative pharmacological tools in subgroups of patients at high risk of developing gallstones.
Collapse
Affiliation(s)
- Agostino Di Ciaula
- a Division of Internal Medicine , Hospital of Bisceglie , Bisceglie , Italy
| | - David Q-H Wang
- b Department of Medicine, Division of Gastroenterology and Liver Diseases , Marion Bessin Liver Research Center, Albert Einstein College of Medicine , Bronx , NY , USA
| | - Piero Portincasa
- c Department of Biomedical Sciences and Human Oncology, Clinica Medica "A. Murri" , University of Bari Medical School , Bari , Italy
| |
Collapse
|
34
|
McCarty MF, DiNicolantonio JJ. Minimizing Membrane Arachidonic Acid Content as a Strategy for Controlling Cancer: A Review. Nutr Cancer 2018; 70:840-850. [DOI: 10.1080/01635581.2018.1470657] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Affiliation(s)
| | - James J. DiNicolantonio
- Preventive Cardiology Department, St. Luke’s Mid America Heart Institute, Kansas City, Missouri, USA
| |
Collapse
|
35
|
Liu X, Xue R, Yang C, Gu J, Chen S, Zhang S. Cholestasis-induced bile acid elevates estrogen level via farnesoid X receptor-mediated suppression of the estrogen sulfotransferase SULT1E1. J Biol Chem 2018; 293:12759-12769. [PMID: 29929982 PMCID: PMC6102144 DOI: 10.1074/jbc.ra118.001789] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2018] [Revised: 06/15/2018] [Indexed: 12/12/2022] Open
Abstract
The liver is the main site of estrogen metabolism, and liver disease is usually associated with an abnormal estrogen status. However, little is known about the mechanism underlying this connection. Here, we investigated the effects of bile acid (BA)-activated farnesoid X receptor (FXR) on the metabolism of 17β-estradiol (E2) during blockage of bile flow (cholestasis). Correlations between BA levels and E2 concentrations were established in patients with cholestasis, and hepatic expression profiles of key genes involved in estrogen metabolism were investigated in both WT and FXR-/- mice. We found that the elevated E2 level positively correlated with BA concentrations in the patients with cholestasis. We further observed that bile duct ligation (BDL) increases E2 levels in mouse serum, and this elevation effect was alleviated by deleting the FXR gene. Of note, FXR down-regulated the expression of hepatic sulfotransferase SULT1E1, the primary enzyme responsible for metabolic estrogen inactivation. At the molecular level, we found that FXR competes with the protein acetylase CREB-binding protein (CBP) for binding to the transcription factor hepatocyte nuclear factor 4α (HNF4α). This competition decreased HNF4α acetylation and nuclear retention, which, in turn, repressed HNF4α-dependent SULT1E1 gene transcription. These findings suggest that cholestasis induces BA-activated FXR activity, leading to downstream inhibition of SULT1E1 and hence impeding hepatic degradation of estrogen.
Collapse
Affiliation(s)
- Xijun Liu
- Key Laboratory of Glycoconjugate Research Ministry of Public Health, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Shanghai 200032, China
| | - Ruyi Xue
- Department of Gastroenterology and Hepatology, Shanghai Institute of Liver Disease, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Caiting Yang
- Key Laboratory of Glycoconjugate Research Ministry of Public Health, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Shanghai 200032, China
| | - Jianxin Gu
- Key Laboratory of Glycoconjugate Research Ministry of Public Health, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Shanghai 200032, China
| | - She Chen
- Key Laboratory of Glycoconjugate Research Ministry of Public Health, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Shanghai 200032, China.
| | - Si Zhang
- Key Laboratory of Glycoconjugate Research Ministry of Public Health, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Shanghai 200032, China.
| |
Collapse
|
36
|
Comeglio P, Cellai I, Mello T, Filippi S, Maneschi E, Corcetto F, Corno C, Sarchielli E, Morelli A, Rapizzi E, Bani D, Guasti D, Vannelli GB, Galli A, Adorini L, Maggi M, Vignozzi L. INT-767 prevents NASH and promotes visceral fat brown adipogenesis and mitochondrial function. J Endocrinol 2018; 238:107-127. [PMID: 29945982 DOI: 10.1530/joe-17-0557] [Citation(s) in RCA: 52] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/16/2018] [Accepted: 05/29/2018] [Indexed: 12/12/2022]
Abstract
The bile acid receptors, farnesoid X receptor (FXR) and Takeda G-protein-coupled receptor 5 (TGR5), regulate multiple pathways, including glucose and lipid metabolism. In a rabbit model of high-fat diet (HFD)-induced metabolic syndrome, long-term treatment with the dual FXR/TGR5 agonist INT-767 reduces visceral adipose tissue accumulation, hypercholesterolemia and nonalcoholic steatohepatitis. INT-767 significantly improves the hallmarks of insulin resistance in visceral adipose tissue (VAT) and induces mitochondrial and brown fat-specific markers. VAT preadipocytes isolated from INT-767-treated rabbits, compared to preadipocytes from HFD, show increased mRNA expression of brown adipogenesis markers. In addition, INT-767 induces improved mitochondrial ultrastructure and dynamic, reduced superoxide production and improved insulin signaling and lipid handling in preadipocytes. Both in vivo and in vitro treatments with INT-767 counteract, in preadipocytes, the HFD-induced alterations by upregulating genes related to mitochondrial biogenesis and function. In preadipocytes, INT-767 behaves mainly as a TGR5 agonist, directly activating dose dependently the cAMP/PKA pathway. However, in vitro experiments also suggest that FXR activation by INT-767 contributes to the insulin signaling improvement. INT-767 treatment counteracts HFD-induced liver histological alterations and normalizes the increased pro-inflammatory genes. INT-767 also induces a significant reduction of fatty acid synthesis and fibrosis markers, while increasing lipid handling, insulin signaling and mitochondrial markers. In conclusion, INT-767 significantly counteracts HFD-induced liver and fat alterations, restoring insulin sensitivity and prompting preadipocytes differentiation toward a metabolically healthy phenotype.
Collapse
Affiliation(s)
- Paolo Comeglio
- Sexual Medicine and Andrology UnitDepartment of Experimental and Clinical Biomedical Sciences 'Mario Serio', University of Florence, Florence, Italy
| | - Ilaria Cellai
- Sexual Medicine and Andrology UnitDepartment of Experimental and Clinical Biomedical Sciences 'Mario Serio', University of Florence, Florence, Italy
| | - Tommaso Mello
- Gastroenterology UnitDepartment of Experimental and Clinical Biomedical Sciences 'Mario Serio', University of Florence, Florence, Italy
| | - Sandra Filippi
- Interdepartmental Laboratory of Functional and Cellular Pharmacology of ReproductionDepartment of NEUROFARBA, University of Florence, Florence, Italy
| | - Elena Maneschi
- Sexual Medicine and Andrology UnitDepartment of Experimental and Clinical Biomedical Sciences 'Mario Serio', University of Florence, Florence, Italy
| | - Francesca Corcetto
- Sexual Medicine and Andrology UnitDepartment of Experimental and Clinical Biomedical Sciences 'Mario Serio', University of Florence, Florence, Italy
| | - Chiara Corno
- Sexual Medicine and Andrology UnitDepartment of Experimental and Clinical Biomedical Sciences 'Mario Serio', University of Florence, Florence, Italy
| | - Erica Sarchielli
- Department of Experimental and Clinical MedicineUniversity of Florence, Florence, Italy
| | - Annamaria Morelli
- Department of Experimental and Clinical MedicineUniversity of Florence, Florence, Italy
| | - Elena Rapizzi
- Department of Experimental and Clinical Biomedical Sciences 'Mario Serio'University of Florence, Florence, Italy
| | - Daniele Bani
- Department of Experimental and Clinical MedicineUniversity of Florence, Florence, Italy
| | - Daniele Guasti
- Department of Experimental and Clinical MedicineUniversity of Florence, Florence, Italy
| | | | - Andrea Galli
- Gastroenterology UnitDepartment of Experimental and Clinical Biomedical Sciences 'Mario Serio', University of Florence, Florence, Italy
| | | | - Mario Maggi
- Sexual Medicine and Andrology UnitDepartment of Experimental and Clinical Biomedical Sciences 'Mario Serio', University of Florence, Florence, Italy
- I.N.B.B. - Istituto Nazionale Biostrutture e BiosistemiRome, Italy
| | - Linda Vignozzi
- Sexual Medicine and Andrology UnitDepartment of Experimental and Clinical Biomedical Sciences 'Mario Serio', University of Florence, Florence, Italy
- I.N.B.B. - Istituto Nazionale Biostrutture e BiosistemiRome, Italy
- Gynecologic Endocrinology Research UnitDepartment of Experimental and Clinical Biomedical Sciences 'Mario Serio', University of Florence, Florence, Italy
| |
Collapse
|
37
|
Abstract
Bile acids facilitate intestinal nutrient absorption and biliary cholesterol secretion to maintain bile acid homeostasis, which is essential for protecting liver and other tissues and cells from cholesterol and bile acid toxicity. Bile acid metabolism is tightly regulated by bile acid synthesis in the liver and bile acid biotransformation in the intestine. Bile acids are endogenous ligands that activate a complex network of nuclear receptor farnesoid X receptor and membrane G protein-coupled bile acid receptor-1 to regulate hepatic lipid and glucose metabolic homeostasis and energy metabolism. The gut-to-liver axis plays a critical role in the regulation of enterohepatic circulation of bile acids, bile acid pool size, and bile acid composition. Bile acids control gut bacteria overgrowth, and gut bacteria metabolize bile acids to regulate host metabolism. Alteration of bile acid metabolism by high-fat diets, sleep disruption, alcohol, and drugs reshapes gut microbiome and causes dysbiosis, obesity, and metabolic disorders. Gender differences in bile acid metabolism, FXR signaling, and gut microbiota have been linked to higher prevalence of fatty liver disease and hepatocellular carcinoma in males. Alteration of bile acid homeostasis contributes to cholestatic liver diseases, inflammatory diseases in the digestive system, obesity, and diabetes. Bile acid-activated receptors are potential therapeutic targets for developing drugs to treat metabolic disorders.
Collapse
Affiliation(s)
- John Y. L. Chiang
- Department of Integrative Medical Sciences, Northeast Ohio Medical University, Rootstown, OH, USA
| | - Jessica M. Ferrell
- Department of Integrative Medical Sciences, Northeast Ohio Medical University, Rootstown, OH, USA
| |
Collapse
|
38
|
Hameed B, Terrault NA, Gill RM, Loomba R, Chalasani N, Hoofnagle JH, Van Natta ML. Clinical and metabolic effects associated with weight changes and obeticholic acid in non-alcoholic steatohepatitis. Aliment Pharmacol Ther 2018; 47:645-656. [PMID: 29333665 PMCID: PMC5931362 DOI: 10.1111/apt.14492] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/30/2017] [Revised: 08/17/2017] [Accepted: 12/07/2017] [Indexed: 02/07/2023]
Abstract
BACKGROUND In a 72-week, randomised controlled trial of obeticholic acid (OCA) in non-alcoholic steatohepatitis (NASH), OCA was superior to placebo in improving serum ALT levels and liver histology. OCA therapy also reduced weight. AIMS Because weight loss by itself can improve histology, to perform a post hoc analysis of the effects of weight loss and OCA treatment in improving clinical and metabolic features of NASH. METHODS The analysis was limited to the 200 patients with baseline and end-of-treatment liver biopsies. Weight loss was defined as a relative decline from baseline of 2% or more at treatment end. RESULTS Weight loss occurred in 44% (45/102) of OCA and 32% (31/98) of placebo-treated patients (P = 0.08). The NAFLD Activity score (NAS) improved more in those with than without weight loss in both the OCA- (-2.4 vs -1.2, P<0.001) and placebo-treated patients (-1.2 vs -0.5, P = 0.03). ALT levels also improved in those with vs without weight loss in OCA- (-43 vs -34 U/L, P = 0.12) and placebo-treated patients (-29 vs -10 U/L, P = 0.02). However, among those who lost weight, OCA was associated with opposite effects from placebo on changes in alkaline phosphatase (+21 vs -12 U/L, P<0.001), total (+13 vs -14 mg/dL, P = 0.02) and LDL cholesterol (+18 vs -12 mg/dL, P = 0.01), and HbA1c (+0.1 vs -0.4%, P = 0.01). CONCLUSIONS OCA leads to weight loss in up to 44% of patients with NASH, and OCA therapy and weight loss have additive benefits on serum aminotransferases and histology. However, favourable effects of weight loss on alkaline phosphatase, lipids and blood glucose seen in placebo-treated patients were absent or reversed on OCA treatment. These findings stress the importance of assessing concomitant metabolic effects of new therapies of NASH. Clinical trial number: NCT01265498.
Collapse
Affiliation(s)
- B Hameed
- University of California San Francisco, San Francisco, CA, USA
| | - N A Terrault
- University of California San Francisco, San Francisco, CA, USA
| | - R M Gill
- University of California San Francisco, San Francisco, CA, USA
| | - R Loomba
- University of California San Diego, San Diego, CA, USA
| | - N Chalasani
- Indiana University School, Indianapolis, IN, USA
| | - J H Hoofnagle
- National Institute of Diabetes and Digestive and Kidney Diseases, Bethesda, MD, USA
| | | |
Collapse
|
39
|
Perito ER, Phelps A, Vase T, Feldstein VA, Lustig RH, Rosenthal P. Subclinical Atherosclerosis in Pediatric Liver Transplant Recipients: Carotid and Aorta Intima-Media Thickness and Their Predictors. J Pediatr 2018; 193:119-127.e1. [PMID: 29224938 PMCID: PMC5794603 DOI: 10.1016/j.jpeds.2017.10.006] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/12/2017] [Revised: 08/21/2017] [Accepted: 10/10/2017] [Indexed: 12/14/2022]
Abstract
OBJECTIVE To investigate prevalence and predictors of cardiovascular risk in pediatric liver transplant recipients using noninvasive markers of subclinical atherosclerosis: carotid intima-media thickness (cIMT) and aorta intima-media thickness (aIMT). STUDY DESIGN Cross-sectional study of 88 pediatric liver transplant recipients. The cIMT and aIMT were measured by ultrasound imaging using standardized protocol. RESULTS Participants were 15.4 ± 4.8 years of age, and 11.2 ± 5.6 years post-transplantation. The cIMT and aIMT were both higher in males than females. In analyses adjusted for sex, age, and height, the cIMT was higher in subjects transplanted for chronic/cirrhotic liver disease and lower in subjects on cyclosporine (n = 9) than tacrolimus (n = 71). The cIMT was not associated with rejection history or current corticosteroid use. The cIMT increased with increasing diastolic blood pressure and triglycerides. The aIMT (n = 83) also increased with age, and its rate of increase post-transplant varied by age at transplantation. In adjusted analyses, aIMT was higher in subjects with glucose intolerance. In analysis of patients ≤20 years of age for whom blood pressure percentiles could be calculated (n = 66), aIMT increased with increasing diastolic blood pressure percentile (0.010 mm per 5-percentile; 95% CI, 0.000-0.021; P = 0.05). Neither the cIMT nor the aIMT was associated with obesity, systolic hypertension, or other dyslipidemia at study visit. CONCLUSION Measures of long-term cardiovascular risk were associated with conditions that are more common in pediatric liver transplant recipients than nontransplanted peers, namely, diastolic hypertension and glucose intolerance. Larger, longitudinal studies are warranted to investigate whether cIMT could be useful for stratifying these patients' cardiovascular risk-and potential need for proactive intervention-during long-term follow-up.
Collapse
Affiliation(s)
- Emily R. Perito
- Department of Pediatrics, UCSF Benioff Children’s Hospital, University of California, San Francisco, San Francisco, CA,Department of Epidemiology and Biostatistics, UCSF Benioff Children’s Hospital, University of California, San Francisco, San Francisco, CA
| | - Andrew Phelps
- Department of Radiology and Biomedical Imaging, UCSF Benioff Children’s Hospital, University of California, San Francisco, San Francisco, CA
| | - Tabitha Vase
- School of Medicine, UCSF Benioff Children’s Hospital, University of California, San Francisco, San Francisco, CA
| | - Vickie A. Feldstein
- Department of Radiology and Biomedical Imaging, UCSF Benioff Children’s Hospital, University of California, San Francisco, San Francisco, CA
| | - Robert H. Lustig
- Department of Pediatrics, UCSF Benioff Children’s Hospital, University of California, San Francisco, San Francisco, CA
| | - Philip Rosenthal
- Department of Pediatrics, UCSF Benioff Children’s Hospital, University of California, San Francisco, San Francisco, CA,Department of Surgery, UCSF Benioff Children’s Hospital, University of California, San Francisco, San Francisco, CA
| |
Collapse
|
40
|
Vasavan T, Ferraro E, Ibrahim E, Dixon P, Gorelik J, Williamson C. Heart and bile acids - Clinical consequences of altered bile acid metabolism. Biochim Biophys Acta Mol Basis Dis 2018; 1864:1345-1355. [PMID: 29317337 DOI: 10.1016/j.bbadis.2017.12.039] [Citation(s) in RCA: 85] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2017] [Revised: 12/18/2017] [Accepted: 12/22/2017] [Indexed: 12/11/2022]
Abstract
Cardiac dysfunction has an increased prevalence in diseases complicated by liver cirrhosis such as primary biliary cholangitis and primary sclerosing cholangitis. This observation has led to research into the association between abnormalities in bile acid metabolism and cardiac pathology. Approximately 50% of liver cirrhosis cases develop cirrhotic cardiomyopathy. Bile acids are directly implicated in this, causing QT interval prolongation, cardiac hypertrophy, cardiomyocyte apoptosis and abnormal haemodynamics of the heart. Elevated maternal serum bile acids in intrahepatic cholestasis of pregnancy, a disorder which causes an impaired feto-maternal bile acid gradient, have been associated with fatal fetal arrhythmias. The hydrophobicity of individual bile acids in the serum bile acid pool is of relevance, with relatively lipophilic bile acids having a more harmful effect on the heart. Ursodeoxycholic acid can reverse or protect against these detrimental cardiac effects of elevated bile acids.
Collapse
Affiliation(s)
- Tharni Vasavan
- Department of Women and Children's Health, King's College London, Guy's Campus, Hodgkin Building, SE1 1UL London, United Kingdom
| | - Elisa Ferraro
- National Heart and Lung Institute, Imperial Centre for Translational and Experimental Medicine, Imperial College London, Du Cane Road, W12 0NN London, United Kingdom
| | - Effendi Ibrahim
- National Heart and Lung Institute, Imperial Centre for Translational and Experimental Medicine, Imperial College London, Du Cane Road, W12 0NN London, United Kingdom; Faculty of Medicine, MARA University of Technology, 40000 Sungai Buloh, Malaysia
| | - Peter Dixon
- Department of Women and Children's Health, King's College London, Guy's Campus, Hodgkin Building, SE1 1UL London, United Kingdom
| | - Julia Gorelik
- National Heart and Lung Institute, Imperial Centre for Translational and Experimental Medicine, Imperial College London, Du Cane Road, W12 0NN London, United Kingdom
| | - Catherine Williamson
- Department of Women and Children's Health, King's College London, Guy's Campus, Hodgkin Building, SE1 1UL London, United Kingdom.
| |
Collapse
|
41
|
Voiosu A, Wiese S, Voiosu T, Bendtsen F, Møller S. Bile acids and cardiovascular function in cirrhosis. Liver Int 2017; 37:1420-1430. [PMID: 28222247 DOI: 10.1111/liv.13394] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/02/2016] [Accepted: 02/12/2017] [Indexed: 02/13/2023]
Abstract
Cirrhotic cardiomyopathy and the hyperdynamic syndrome are clinically important complications of cirrhosis, but their exact pathogenesis is still partly unknown. Experimental models have proven the cardiotoxic effects of bile acids and recent studies of their varied receptor-mediated functions offer new insight into their involvement in cardiovascular dysfunction in cirrhosis. Bile acid receptors such as farnesoid X-activated receptor and TGR5 are currently under investigation as potential therapeutic targets in a variety of pathological conditions. These receptors have also recently been identified in cardiomyocytes, vascular endothelial cells and smooth muscle cells where they seem to play an important role in cellular metabolism. Chronic cholestasis leading to abnormal levels of circulating bile acids alters the normal signalling pathways and contributes to the development of profound cardiovascular disturbances. This review summarizes the evidence regarding the role of bile acids and their receptors in the generation of cardiovascular dysfunction in cirrhosis.
Collapse
Affiliation(s)
- Andrei Voiosu
- Department of Clinical Physiology and Nuclear Medicine, Center for Functional and Diagnostic Imaging and Research, Hvidovre Hospital, Hvidovre, Denmark.,Gastroenterology and Hepatology Department, Colentina Clinical Hospital, Bucharest, Romania.,"Carol Davila" University of Medicine and Pharmacy, Bucharest, Romania
| | - Signe Wiese
- Department of Clinical Physiology and Nuclear Medicine, Center for Functional and Diagnostic Imaging and Research, Hvidovre Hospital, Hvidovre, Denmark.,Faculty of Health Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Theodor Voiosu
- Gastroenterology and Hepatology Department, Colentina Clinical Hospital, Bucharest, Romania.,"Carol Davila" University of Medicine and Pharmacy, Bucharest, Romania
| | - Flemming Bendtsen
- Faculty of Health Sciences, University of Copenhagen, Copenhagen, Denmark.,Gastro Unit, Medical Division, Hvidovre Hospital, Hvidovre, Denmark
| | - Søren Møller
- Department of Clinical Physiology and Nuclear Medicine, Center for Functional and Diagnostic Imaging and Research, Hvidovre Hospital, Hvidovre, Denmark.,Faculty of Health Sciences, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
42
|
Gao J, Liu X, Wang B, Xu H, Xia Q, Lu T, Wang F. Farnesoid X receptor deletion improves cardiac function, structure and remodeling following myocardial infarction in mice. Mol Med Rep 2017; 16:673-679. [PMID: 28560412 PMCID: PMC5482148 DOI: 10.3892/mmr.2017.6643] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2016] [Accepted: 02/20/2017] [Indexed: 12/28/2022] Open
Abstract
The farnesoid X receptor (FXR) is implicated in cholesterol and bile acid homeostasis; however, its role following myocardial infarction (MI) has yet to be elucidated. The aim of the present study was to investigate the effects of FXR knockout on left ventricular (LV) remodeling following MI. Coronary arteries of wild type (WT) and FXR‑/‑ mice were permanently occluded to cause MI, and serial echocardiographic and histological tests were conducted. At 4 weeks post‑MI, FXR‑/‑ mice exhibited significantly smaller infarct sizes (34.20±2.58 vs. 44.20±3.19%), improved ejection fraction (47.31±1.08 vs. 37.64±0.75%) and reduced LV chamber dilation compared with WT mice. LV remodeling was significant as early as 7 days post‑MI in FXR‑/‑ compared with WT mice. Histological features associated with enhanced long‑term remodeling and improved functionality, such as increased angiogenesis via detection of CD31 and reduced fibrosis, were observed in the FXR‑/‑ group. Myocyte apoptosis within the infarcted zones appeared significantly reduced by day 7 in FXR‑/‑ mice. In conclusion, the results of the present study suggested that FXR knockout may participate in the preservation of post‑MI cardiac functionality, via reducing fibrosis and chronic apoptosis, and ameliorating ventricular function.
Collapse
Affiliation(s)
- Jianshu Gao
- Department of Cardiology, Shanghai General Hospital of Nanjing Medical University, Shanghai 200080, P.R. China
| | - Xiaoqiang Liu
- Department of Cardiology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, P.R. China
| | - Bingjian Wang
- Department of Cardiology, Huai'an First People's Hospital, Nanjing Medical University, Huai'an, Jiangsu 223300, P.R. China
| | - Haiyan Xu
- Department of Cardiology, Huai'an First People's Hospital, Nanjing Medical University, Huai'an, Jiangsu 223300, P.R. China
| | - Qiang Xia
- Department of Transplantation and Hepatic Surgery, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, P.R. China
| | - Tianfei Lu
- Department of Transplantation and Hepatic Surgery, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, P.R. China
| | - Fang Wang
- Department of Cardiology, Shanghai General Hospital of Nanjing Medical University, Shanghai 200080, P.R. China
| |
Collapse
|
43
|
Synthesis and biological evaluations of chalcones, flavones and chromenes as farnesoid x receptor (FXR) antagonists. Eur J Med Chem 2017; 129:303-309. [PMID: 28235703 DOI: 10.1016/j.ejmech.2017.02.037] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2016] [Revised: 01/13/2017] [Accepted: 02/14/2017] [Indexed: 01/06/2023]
Abstract
Farnesoid X receptor (FXR), a nuclear receptor mainly distributed in liver and intestine, has been regarded as a potential target for the treatment of various metabolic diseases, cancer and infectious diseases related to liver. Starting from two previously identified chalcone-based FXR antagonists, we tried to increase the activity through the design and synthesis of a library containing chalcones, flavones and chromenes, based on substitution manipulation and conformation (ring closure) restriction strategy. Many chalcones and four chromenes were identified as microM potent FXR antagonists, among which chromene 11c significantly decreased the plasma and hepatic triglyceride level in KKay mice.
Collapse
|
44
|
Unsworth AJ, Kriek N, Bye AP, Naran K, Sage T, Flora GD, Gibbins JM. PPARγ agonists negatively regulate αIIbβ3 integrin outside-in signaling and platelet function through up-regulation of protein kinase A activity. J Thromb Haemost 2017; 15:356-369. [PMID: 27896950 PMCID: PMC5396324 DOI: 10.1111/jth.13578] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2016] [Indexed: 12/31/2022]
Abstract
Essentials peroxisome proliferator-activated receptor γ (PPARγ) agonists inhibit platelet function. PPARγ agonists negatively regulate outside-in signaling via integrin αIIbβ3. PPARγ agonists disrupt the interaction of Gα13 with integrin β3. This is attributed to an upregulation of protein kinase A activity. SUMMARY Background Agonists for the peroxisome proliferator-activated receptor (PPARγ) have been shown to have inhibitory effects on platelet activity following stimulation by GPVI and GPCR agonists. Objectives Profound effects on thrombus formation led us to suspect a role for PPARγ agonists in the regulation of integrin αIIbβ3 mediated signaling. Both GPVI and GPCR signaling pathways lead to αIIbβ3 activation, and signaling through αIIbβ3 plays a critical role in platelet function and normal hemostasis. Methods The effects of PPARγ agonists on the regulation of αIIbβ3 outside-in signaling was determined by monitoring the ability of platelets to adhere and spread on fibrinogen and undergo clot retraction. Effects on signaling components downstream of αIIbβ3 activation were also determined following adhesion to fibrinogen by Western blotting. Results Treatment of platelets with PPARγ agonists inhibited platelet adhesion and spreading on fibrinogen and diminished clot retraction. A reduction in phosphorylation of several components of αIIbβ3 signaling, including the integrin β3 subunit, Syk, PLCγ2, focal adhesion kinase (FAK) and Akt, was also observed as a result of reduced interaction of the integrin β3 subunit with Gα13. Studies of VASP phosphorylation revealed that this was because of an increase in PKA activity following treatment with PPARγ receptor agonists. Conclusions This study provides further evidence for antiplatelet actions of PPARγ agonists, identifies a negative regulatory role for PPARγ agonists in the control of integrin αIIbβ3 outside-in signaling, and provides a molecular basis by which the PPARγ agonists negatively regulate platelet activation and thrombus formation.
Collapse
Affiliation(s)
- A. J. Unsworth
- Institute for Cardiovascular and Metabolic ResearchSchool of Biological SciencesUniversity of ReadingReadingUK
| | - N. Kriek
- Institute for Cardiovascular and Metabolic ResearchSchool of Biological SciencesUniversity of ReadingReadingUK
| | - A. P. Bye
- Institute for Cardiovascular and Metabolic ResearchSchool of Biological SciencesUniversity of ReadingReadingUK
| | - K. Naran
- Institute for Cardiovascular and Metabolic ResearchSchool of Biological SciencesUniversity of ReadingReadingUK
| | - T. Sage
- Institute for Cardiovascular and Metabolic ResearchSchool of Biological SciencesUniversity of ReadingReadingUK
| | - G. D. Flora
- Institute for Cardiovascular and Metabolic ResearchSchool of Biological SciencesUniversity of ReadingReadingUK
| | - J. M. Gibbins
- Institute for Cardiovascular and Metabolic ResearchSchool of Biological SciencesUniversity of ReadingReadingUK
| |
Collapse
|
45
|
Ponnusamy S, Tran QT, Thiyagarajan T, Miller DD, Bridges D, Narayanan R. An estrogen receptor β-selective agonist inhibits non-alcoholic steatohepatitis in preclinical models by regulating bile acid and xenobiotic receptors. Exp Biol Med (Maywood) 2017; 242:606-616. [PMID: 28092182 DOI: 10.1177/1535370216688569] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Non-alcoholic steatohepatitis (NASH) affects 8-10 million people in the US and up to 75% of obese individuals. Despite this, there are no approved oral therapeutics to treat NASH and therefore the need for novel approaches exists. The estrogen receptor β (ER-β)-selective agonist, β-LGND2, inhibits body weight and white adipose tissue, and increases metabolism, resulting in higher energy expenditure and thermogenesis. Due to favorable effects of β-LGND2 on obesity, we hypothesized that β-LGND2 will prevent NASH directly by reducing lipid accumulation in the liver or indirectly by favorably changing body composition. Male C57BL/6 mice fed with high fat diet (HFD) for 10 weeks or methionine choline-deficient diet for four weeks and treated with vehicle exhibited altered liver weights by twofold and increased serum transaminases by 2-6-folds. These changes were not observed in β-LGND2-treated animals. Infiltration of inflammatory cells and collagen deposits, an indication of fibrosis, were observed in the liver of mice fed with HFD for 10 weeks, which were effectively blocked by β-LGND2. Gene expression studies in the liver indicate that pregnane X receptor target genes were significantly increased by HFD, and the increase was inhibited by β-LGND2. On the other hand, metabolomics indicate that bile acid metabolites were significantly increased by β-LGND2. These studies demonstrate that an ER-β agonist might provide therapeutic benefits in NASH by directly modulating the function of xenobiotic and bile acid receptors in the liver, which have important functions in the liver, and indirectly, as demonstrated before, by inhibiting adiposity. Impact statement Over 75-90% of those classified as clinically obese suffer from co-morbidities, the most common of which is non-alcoholic steatohepatitis (NASH). While there are currently no effective treatment approaches for NASH, data presented here provide preliminary evidence that an estrogen receptor β-selective ligand could have the potential to reduce lipid accumulation and inflammation, and protect liver from NASH.
Collapse
Affiliation(s)
- Suriyan Ponnusamy
- 1 Department of Medicine, University of Tennessee Health Science Center, Memphis, TN 38103, USA
| | - Quynh T Tran
- 2 Department of Preventive Medicine, University of Tennessee Health Science Center, Memphis, TN 38103, USA
| | - Thirumagal Thiyagarajan
- 1 Department of Medicine, University of Tennessee Health Science Center, Memphis, TN 38103, USA
| | - Duane D Miller
- 3 Department of Pharmaceutical Sciences, University of Tennessee Health Science Center, Memphis, TN 38103, USA
| | - Dave Bridges
- 4 Department of Physiology, University of Tennessee Health Science Center, Memphis, TN 38103, USA.,5 Department of Pediatrics, University of Tennessee Health Science Center, Memphis, TN 38103, USA.,6 Department of Nutritional Sciences, University of Michigan School of Public Health, Ann Arbor, MI 48104, USA (present address)
| | - Ramesh Narayanan
- 1 Department of Medicine, University of Tennessee Health Science Center, Memphis, TN 38103, USA.,7 West Cancer Center, Memphis, TN 38103, USA
| |
Collapse
|
46
|
Zhang R, Ran H, Peng L, Zhang Y, Shen W, Sun T, Cao F, Chen Y. Farnesoid X receptor regulates vasoreactivity via Angiotensin II type 2 receptor and the kallikrein-kinin system in vascular endothelial cells. Clin Exp Pharmacol Physiol 2016; 43:327-34. [PMID: 26710942 DOI: 10.1111/1440-1681.12535] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2014] [Revised: 12/21/2015] [Accepted: 12/23/2015] [Indexed: 01/02/2023]
Abstract
Vascular farnesoid X receptor (FXR) ligands have been shown previously to regulate vascular tension. This study investigated whether FXR activation regulates vasoreactivity via the angiotensin II (Ang II) type 2 receptor (AT2 R) and the kallikrein-kinin system in rat aortic vascular endothelial cells (RAECs). Protein abundances of Ang II type 1 receptor (AT1 R), AT2 R, bradykinin type 1/2 receptor (B1 R, B2 R), small heterodimer partner-1 (SHP-1) and the endothelial and inducible NO synthases (eNOS/iNOS) were analysed by Western blotting. Real-time quantitative polymerase chain reaction was performed to analyse expression of eNOS and iNOS mRNA. Kallikrein activity and bradykinin content were assayed using spectrophotometry and a bradykinin assay kit, respectively. Aortic vasoconstriction and vasodilation were also investigated following FXR activation in the presence or absence of AT2 R and B2 R blockade. It was found that the FXR agonists GW4064 and INT-747, in a dose-dependent manner, increased the protein abundance of AT2 R, B2 R and SHP-1 and decreased that of AT1 R. AT2 R blockade with PD123319 reversed effects of FXR agonists on kallikrein activity and levels of SHP-1, B2 R and bradykinin. Moreover, it was found that GW4064 and INT-747 upregulated expression of eNOS and enhanced NOS activity, which attenuated vasoconstriction and induced vasodilation, respectively. These effects were partially reversed by PD123319 and by B2 R blockade with HOE140. The current work suggests that FXR regulates vascular tension by controlling the eNOS-NO system via activation of a pathway mediated by AT2 R-B2 R pathway in RAECs.
Collapse
Affiliation(s)
- Ran Zhang
- Department of Cardiology, Chinese PLA General Hospital, Beijing, China
| | - Haihong Ran
- Department of Geriatric Haematology, Chinese PLA General Hospital, Beijing, China
| | - Liang Peng
- Department of Cardiology, Chinese PLA General Hospital, Beijing, China
| | - Ying Zhang
- Department of Cardiology, Chinese PLA General Hospital, Beijing, China
| | - Wenbin Shen
- Department of Cardiology, Chinese PLA General Hospital, Beijing, China
| | - Ting Sun
- Department of Cardiology, Chinese PLA General Hospital, Beijing, China
| | - Feng Cao
- Department of Cardiology, Chinese PLA General Hospital, Beijing, China
| | - Yundai Chen
- Department of Cardiology, Chinese PLA General Hospital, Beijing, China
| |
Collapse
|
47
|
Obeticholic acid, a synthetic bile acid agonist of the farnesoid X receptor, attenuates experimental autoimmune encephalomyelitis. Proc Natl Acad Sci U S A 2016; 113:1600-5. [PMID: 26811456 DOI: 10.1073/pnas.1524890113] [Citation(s) in RCA: 64] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Bile acids are ligands for the nuclear hormone receptor, farnesoid X receptor (FXR). The bile acid-FXR interaction regulates bile acid synthesis, transport, and cholesterol metabolism. Recently, bile acid-FXR regulation has been reported to play an integral role in both hepatic and intestinal inflammation, and in atherosclerosis. In this study, we found that FXR knockout mice had more disease severity in experimental autoimmune encephalomyelitis (EAE), an animal model of multiple sclerosis (MS). Obeticholic acid (6α-ethyl-chenodeoxycholic acid, 6-ECDCA), a synthetic FXR agonist, is an orally available drug that is currently in clinical trials for the treatment of inflammatory diseases such as alcoholic hepatitis, nonalcoholic steatohepatitis, and primary biliary cirrhosis. When we treated mice exhibiting established EAE with 6-ECDCA, or the natural FXR ligand chenodeoxycholic acid (CDCA), clinical disease was ameliorated by (i) suppressing lymphocyte activation and proinflammatory cytokine production; (ii) reducing CD4(+) T cells and CD19(+) B cell populations and their expression of negative checkpoint regulators programmed cell death protein 1 (PD1), programmed death-ligand 1 (PD-L1), and B and T lymphocyte attenuator (BTLA); (iii) increasing CD8(+) T cells and PD1, PDl-1, and BTLA expression; and (iv) reducing VLA-4 expression in both the T- and B-cell populations. Moreover, adoptive transfer of 6-ECDCA- or CDCA-treated donor cells failed to transfer disease in naive recipients. Thus, we show that FXR functions as a negative regulator in neuroinflammation and we highlight that FXR agonists represent a potential previously unidentified therapy for MS.
Collapse
|
48
|
Kumar S, Lau R, Hall C, Palaia T, Brathwaite CE, Ragolia L. Bile acid elevation after Roux-en-Y gastric bypass is associated with cardio-protective effect in Zucker Diabetic Fatty rats. Int J Surg 2015; 24:70-74. [PMID: 26563489 DOI: 10.1016/j.ijsu.2015.11.010] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2015] [Revised: 10/26/2015] [Accepted: 11/05/2015] [Indexed: 01/06/2023]
Abstract
BACKGROUND Roux-en-Y gastric bypass (RYGB) may improve cardiometabolic risk through alteration of bile acids and L-PGDS levels. OBJECTIVE The objective of this study was to investigate the effect of RYGB on aortic wall thickness, in relation to bile acid and L-PGDS metabolism. METHODS Zucker diabetic fatty (ZDF) rats were divided into two groups, ad lib (n = 4), and RYGB (n = 6). Bile acid and L-PGDS were measured presurgery and fourteen weeks post-surgery. RESULTS Elevation of bile acid levels following RYGB in Zucker Diabetic Fatty (ZDF) rodents was observed, as compared to ad lib. RYGB in ZDF rodents led to a significantly decreased aortic wall thickness (25%) as compared to ad lib control. Although bile acid metabolism is implicated in these alterations, other mediators are likely involved. Our laboratory has demonstrated lipocalin prostaglandin D2 synthase (L-PGDS) is a kno n cardiometabolic modulator that also functions as a bile acid binding protein. Therefore, L-PGDS levels were measured and a significant elevation was observed with RYGB compared to ad lib control. CONCLUSION Based on these findings, RYGB showed beneficial effect on aortic wall thickness, possibly through bile acids and L-PGDS elevation in a severely obese and diabetic rodent model.
Collapse
Affiliation(s)
- Sunil Kumar
- Department of Biomedical Research, Winthrop University Hospital, Mineola, NY 11501, USA
| | - Raymond Lau
- Department of Surgery, Winthrop University Hospital, Mineola, NY 11501, USA; Department of Endocrinology, Winthrop University Hospital, Mineola, NY 11501, USA
| | - Christopher Hall
- Department of Biomedical Research, Winthrop University Hospital, Mineola, NY 11501, USA
| | - Thomas Palaia
- Department of Biomedical Research, Winthrop University Hospital, Mineola, NY 11501, USA
| | - Collin E Brathwaite
- Department of Surgery, Winthrop University Hospital, Mineola, NY 11501, USA; Department of Medicine, Stony Brook University School of Medicine, Stony Brook, NY 11794, USA
| | - Louis Ragolia
- Department of Biomedical Research, Winthrop University Hospital, Mineola, NY 11501, USA; Department of Medicine, Stony Brook University School of Medicine, Stony Brook, NY 11794, USA.
| |
Collapse
|
49
|
TGR5 and Immunometabolism: Insights from Physiology and Pharmacology. Trends Pharmacol Sci 2015; 36:847-857. [PMID: 26541439 DOI: 10.1016/j.tips.2015.08.002] [Citation(s) in RCA: 112] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2015] [Revised: 08/02/2015] [Accepted: 08/04/2015] [Indexed: 12/15/2022]
Abstract
In the past decade substantial progress has been made in understanding how the insurgence of chronic low-grade inflammation influences the physiology of several metabolic diseases. Tissue-resident immune cells have been identified as central players in these processes, linking inflammation to metabolism. The bile acid-responsive G-protein-coupled receptor TGR5 is expressed in monocytes and macrophages, and its activation mediates potent anti-inflammatory effects. Herein, we summarize recent advances in TGR5 research, focusing on the downstream effector pathways that are modulated by TGR5 activators, and on its therapeutic potential in inflammatory and metabolic diseases.
Collapse
|
50
|
Ryan PM, Ross RP, Fitzgerald GF, Caplice NM, Stanton C. Functional food addressing heart health: do we have to target the gut microbiota? Curr Opin Clin Nutr Metab Care 2015; 18:566-71. [PMID: 26406391 DOI: 10.1097/mco.0000000000000224] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
PURPOSE OF REVIEW Health promoting functional food ingredients for cardiovascular health are generally aimed at modulating lipid metabolism in consumers. However, significant advances have furthered our understanding of the mechanisms involved in development, progression, and treatment of cardiovascular disease. In parallel, a central role of the gut microbiota, both in accelerating and attenuating cardiovascular disease, has emerged. RECENT FINDINGS Modulation of the gut microbiota, by use of prebiotics and probiotics, has recently shown promise in cardiovascular disease prevention. Certain prebiotics can promote a short chain fatty acid profile that alters hormone secretion and attenuates cholesterol synthesis, whereas bile salt hydrolase and exopolysaccharide-producing probiotics have been shown to actively correct hypercholesterolemia. Furthermore, specific microbial genera have been identified as potential cardiovascular disease risk factors. This effect is attributed to the ability of certain members of the gut microbiota to convert dietary quaternary amines to trimethylamine, the primary substrate of the putatively atherosclerosis-promoting compound trimethylamine-N-oxide. In this respect, current research is indicating trimethylamine-depleting Achaea - termed Archeabiotics as a potential novel dietary strategy for promoting heart health. SUMMARY The microbiota offers a modifiable target, which has the potential to progress or prevent cardiovascular disease development. Whereas host-targeted interventions remain the standard, current research implicates microbiota-mediated therapies as an effective means of modulating cardiovascular health.
Collapse
Affiliation(s)
- Paul M Ryan
- aFood Biosciences Department, Teagasc Food Research Centre, Moorepark, Fermoy bSchool of Microbiology cAPC Microbiome Institute, Biosciences Institute dCollege of Science, Engineering and Food Science eCentre for Research in Vascular Biology, University College Cork, Cork, Ireland
| | | | | | | | | |
Collapse
|