1
|
Chen Y, Chen R, Li H, Shuai Z. Clinical management of autoimmune liver diseases: juncture, opportunities, and challenges ahead. Immunol Res 2025; 73:67. [PMID: 40195209 PMCID: PMC11976385 DOI: 10.1007/s12026-025-09622-9] [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: 11/27/2024] [Accepted: 03/14/2025] [Indexed: 04/09/2025]
Abstract
The three major autoimmune liver diseases are autoimmune hepatitis (AIH), primary biliary cholangitis (PBC), and primary sclerosing cholangitis (PSC).These conditions are assumed to result from a breakdown in immunological tolerance, which leads to an inflammatory process that causes liver damage.The self-attack is started by T-helper cell-mediated identification of liver autoantigens and B-cell production of autoantibodies,and it is maintained by a reduction in the number and activity of regulatory T-cells.Infections and environmental factors have been explored as triggering factors for these conditions, in addition to a genetic predisposition.Allelic mutations in the HLA locus have been linked to vulnerability, as have relationships with single nucleotide polymorphisms in non-HLA genes.Despite the advances in the management of these diseases, there is no curative treatment for these disorders, and a significant number of patients eventually progress to an end-stage liver disease requiring liver transplantation.In this line, tailored immune-therapeutics have emerged as possible treatments to control the disease.In addition, early diagnosis and treatment are pivotal for reducing the long-lasting effects of these conditions and their burden on quality of life.Herein we present a review of the etiology, clinical presentation, diagnosis, and challenges on ALDs and the feasible solutions for these complex diseases.
Collapse
MESH Headings
- Humans
- Hepatitis, Autoimmune/therapy
- Hepatitis, Autoimmune/diagnosis
- Hepatitis, Autoimmune/immunology
- Hepatitis, Autoimmune/etiology
- Cholangitis, Sclerosing/therapy
- Cholangitis, Sclerosing/diagnosis
- Cholangitis, Sclerosing/immunology
- Liver Cirrhosis, Biliary/therapy
- Liver Cirrhosis, Biliary/diagnosis
- Liver Cirrhosis, Biliary/immunology
- Animals
- Immunotherapy/methods
- Autoimmune Diseases/therapy
- Autoimmune Diseases/diagnosis
- Disease Management
- Genetic Predisposition to Disease
Collapse
Affiliation(s)
- Yangfan Chen
- Department of Rheumatology and Immunology, The First Affiliated Hospital of Anhui Medical University, Hefei, 230022, China
| | - Ruofei Chen
- Department of Rheumatology and Immunology, The First Affiliated Hospital of Anhui Medical University, Hefei, 230022, China
| | - Haiyan Li
- Department of Rheumatology and Immunology, The First Affiliated Hospital of Anhui Medical University, Hefei, 230022, China
| | - Zongwen Shuai
- Department of Rheumatology and Immunology, The First Affiliated Hospital of Anhui Medical University, Hefei, 230022, China.
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Hefei, 230032, China.
| |
Collapse
|
2
|
Zhang C, Ma H, DeRoche D, Gale EM, Pantazopoulos P, Rotile NJ, Diyabalanage H, Humblet V, Caravan P, Zhou IY. Manganese-based type I collagen-targeting MRI probe for in vivo imaging of liver fibrosis. RESEARCH SQUARE 2024:rs.3.rs-5349052. [PMID: 39606447 PMCID: PMC11601876 DOI: 10.21203/rs.3.rs-5349052/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/29/2024]
Abstract
Liver fibrosis is a common pathway shared by all forms of progressive chronic liver disease. There is an unmet clinical need for noninvasive imaging tools to diagnose and stage fibrosis, which presently relies heavily on percutaneous liver biopsy. Here we explored the feasibility of using a novel type I collagen-targeted manganese (Mn)-based MRI probe, Mn-CBP20, for liver fibrosis imaging. In vitro characterization of Mn-CBP20 demonstrated its high binding affinity for human collagen (K d = 9.6 μM), high T1-relaxivity (48.9 mM-1s-1 at 1.4T and 27°C), and kinetic inertness to Mn release under forcing conditions. We demonstrated MRI using Mn-CBP20 performs comparably to previously reported gadolinium-based type I collagen-targeted probe EP-3533 in a mouse model of carbon tetrachloride-induced liver fibrosis, and further demonstrate efficacy to detect fibrosis in a diet-induced mouse model of metabolically-associated steatohepatitis. Biodistribution studies using the Mn-CBP20 radio-labeled with the positron-emitting 52Mn isotope demonstrate efficient clearance of Mn-CBP20 primarily via renal excretion. Mn-CBP20 represents a promising candidate that merits further evaluation and development for molecular imaging of liver fibrosis.
Collapse
Affiliation(s)
- Chunxiang Zhang
- Athinoula A. Martinos Center for Biomedical Imaging, Institute for Innovation in Imaging (i), Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, USA
| | - Hua Ma
- Athinoula A. Martinos Center for Biomedical Imaging, Institute for Innovation in Imaging (i), Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, USA
| | - Daniel DeRoche
- Athinoula A. Martinos Center for Biomedical Imaging, Institute for Innovation in Imaging (i), Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, USA
| | - Eric M. Gale
- Athinoula A. Martinos Center for Biomedical Imaging, Institute for Innovation in Imaging (i), Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, USA
| | - Pamela Pantazopoulos
- Athinoula A. Martinos Center for Biomedical Imaging, Institute for Innovation in Imaging (i), Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, USA
| | - Nicholas J. Rotile
- Athinoula A. Martinos Center for Biomedical Imaging, Institute for Innovation in Imaging (i), Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, USA
| | | | | | - Peter Caravan
- Athinoula A. Martinos Center for Biomedical Imaging, Institute for Innovation in Imaging (i), Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, USA
| | - Iris Y. Zhou
- Athinoula A. Martinos Center for Biomedical Imaging, Institute for Innovation in Imaging (i), Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, USA
| |
Collapse
|
3
|
Chen S, Zhuang D, Jia Q, Guo B, Hu G. Advances in Noninvasive Molecular Imaging Probes for Liver Fibrosis Diagnosis. Biomater Res 2024; 28:0042. [PMID: 38952717 PMCID: PMC11214848 DOI: 10.34133/bmr.0042] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2024] [Accepted: 05/08/2024] [Indexed: 07/03/2024] Open
Abstract
Liver fibrosis is a wound-healing response to chronic liver injury, which may lead to cirrhosis and cancer. Early-stage fibrosis is reversible, and it is difficult to precisely diagnose with conventional imaging modalities such as magnetic resonance imaging, positron emission tomography, single-photon emission computed tomography, and ultrasound imaging. In contrast, probe-assisted molecular imaging offers a promising noninvasive approach to visualize early fibrosis changes in vivo, thus facilitating early diagnosis and staging liver fibrosis, and even monitoring of the treatment response. Here, the most recent progress in molecular imaging technologies for liver fibrosis is updated. We start by illustrating pathogenesis for liver fibrosis, which includes capillarization of liver sinusoidal endothelial cells, cellular and molecular processes involved in inflammation and fibrogenesis, as well as processes of collagen synthesis, oxidation, and cross-linking. Furthermore, the biological targets used in molecular imaging of liver fibrosis are summarized, which are composed of receptors on hepatic stellate cells, macrophages, and even liver collagen. Notably, the focus is on insights into the advances in imaging modalities developed for liver fibrosis diagnosis and the update in the corresponding contrast agents. In addition, challenges and opportunities for future research and clinical translation of the molecular imaging modalities and the contrast agents are pointed out. We hope that this review would serve as a guide for scientists and students who are interested in liver fibrosis imaging and treatment, and as well expedite the translation of molecular imaging technologies from bench to bedside.
Collapse
Affiliation(s)
- Shaofang Chen
- Department of Radiology, Shenzhen People’s Hospital (The Second Clinical Medical College,
Jinan University; The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen 518020, Guangdong, China
| | - Danping Zhuang
- Department of Radiology, Shenzhen People’s Hospital (The Second Clinical Medical College,
Jinan University; The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen 518020, Guangdong, China
| | - Qingyun Jia
- Department of Radiology, Shenzhen People’s Hospital (The Second Clinical Medical College,
Jinan University; The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen 518020, Guangdong, China
| | - Bing Guo
- School of Science, Shenzhen Key Laboratory of Flexible Printed Electronics Technology, Shenzhen Key Laboratory of Advanced Functional Carbon Materials Research and Comprehensive Application,
Harbin Institute of Technology, Shenzhen 518055, China
| | - Genwen Hu
- Department of Radiology, Shenzhen People’s Hospital (The Second Clinical Medical College,
Jinan University; The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen 518020, Guangdong, China
| |
Collapse
|
4
|
Adorini L, Trauner M. FXR agonists in NASH treatment. J Hepatol 2023; 79:1317-1331. [PMID: 37562746 DOI: 10.1016/j.jhep.2023.07.034] [Citation(s) in RCA: 42] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/14/2023] [Revised: 06/19/2023] [Accepted: 07/16/2023] [Indexed: 08/12/2023]
Abstract
The farnesoid X receptor (FXR), a bile acid (BA)-activated nuclear receptor highly expressed in the liver and intestine, regulates the expression of genes involved in cholesterol and bile acid homeostasis, hepatic gluconeogenesis, lipogenesis, inflammation and fibrosis, in addition to controlling intestinal barrier integrity, preventing bacterial translocation and maintaining gut microbiota eubiosis. Non-alcoholic steatohepatitis (NASH), an advanced stage of non-alcoholic fatty liver disease, is characterized by hepatic steatosis, hepatocyte damage (ballooning) and inflammation, leading to fibrosis, cirrhosis and hepatocellular carcinoma. NASH represents a major unmet medical need, but no pharmacological treatments have yet been approved. The pleiotropic mechanisms involved in NASH development offer a range of therapeutic opportunities and among them FXR activation has emerged as an established pharmacological target. Various FXR agonists with different physicochemical properties, which can be broadly classified as BA derivatives, non-BA-derived steroidal FXR agonists, non-steroidal FXR agonists, and partial FXR agonists, are in advanced clinical development. In this review we will summarize key preclinical and clinical features of the most advanced FXR agonists and critically evaluate their potential in NASH treatment.
Collapse
Affiliation(s)
- Luciano Adorini
- Intercept Pharmaceuticals Inc., 305 Madison Ave., Morristown, NJ 07960, USA.
| | - Michael Trauner
- Division of Gastroenterology and Hepatology, Department of Internal Medicine III, Medical University of Vienna, Waehringer Guertel 18-20, A-1090 Vienna, Austria.
| |
Collapse
|
5
|
Mik P, Barannikava K, Surkova P. Biased Quantification of Rat Liver Fibrosis-Meta-Analysis with Practical Recommendations and Clinical Implications. J Clin Med 2023; 12:5072. [PMID: 37568474 PMCID: PMC10420125 DOI: 10.3390/jcm12155072] [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: 06/30/2023] [Revised: 07/21/2023] [Accepted: 07/29/2023] [Indexed: 08/13/2023] Open
Abstract
For liver fibrosis assessment, the liver biopsy is usually stained with Masson's trichrome (MT) or picrosirius red (PSR) to quantify liver connective tissue (LCT) for fibrosis scoring. However, several concerns of such semiquantitative assessments have been raised, and when searching for data on the amount of LCT in healthy rats, the results vastly differ. Regarding the ongoing reproducibility crisis in science, it is necessary to inspect the results and methods, and to design an unbiased and reproducible method of LCT assessment. We searched the Medline database using search terms related to liver fibrosis, LCT and collagen, rat strains, and staining methods. Our search identified 74 eligible rat groups in 57 studies. We found up to 170-fold differences in the amount of LCT among healthy Wistar and Sprague-Dawley rats, with significant differences even within individual studies. Biased sampling and quantification probably caused the observed differences. In addition, we also found incorrect handling of liver fibrosis scoring. Assessment of LCT using stereological sampling methods (such as systematic uniform sampling) would provide us with unbiased data. Such data could eventually be used not only for the objective assessment of liver fibrosis but also for validation of noninvasive methods of the assessment of early stages of liver fibrosis.
Collapse
Affiliation(s)
- Patrik Mik
- Department of Anatomy, Faculty of Medicine in Pilsen, Charles University, alej Svobody 76, 323 00 Pilsen, Czech Republic
- Biomedical Center and Department of Histology, Faculty of Medicine in Pilsen, Charles University, alej Svobody 76, 323 00 Pilsen, Czech Republic
| | - Katsiaryna Barannikava
- Department of Anatomy, Faculty of Medicine in Pilsen, Charles University, alej Svobody 76, 323 00 Pilsen, Czech Republic
| | - Polina Surkova
- Department of Anatomy, Faculty of Medicine in Pilsen, Charles University, alej Svobody 76, 323 00 Pilsen, Czech Republic
| |
Collapse
|
6
|
Xu X, Qiu J, Li X, Chen J, Li Y, Huang X, Zang S, Ma X, Liu J. Perilipin5 protects against non-alcoholic steatohepatitis by increasing 11-Dodecenoic acid and inhibiting the occurrence of ferroptosis. Nutr Metab (Lond) 2023; 20:29. [PMID: 37349836 DOI: 10.1186/s12986-023-00751-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2023] [Accepted: 05/31/2023] [Indexed: 06/24/2023] Open
Abstract
BACKGROUND Non-alcoholic steatohepatitis (NASH) is a major contributor to liver cirrhosis and hepatocellular carcinoma. There remains no effective pharmacological therapy. The hepatic lipid metabolism and fatty acid β-oxidation are regulated by Perilipin5 (Plin5). However, it is yet unknown how Plin5 affects NASH and the molecular process. METHODS High-fat, high-cholesterol and high-fructose (HFHC) diets were used to mimic the progression of NASH in wild type (WT) mice and Plin5 knockout (Plin5 KO) mice. The degree of ferroptosis was measured by detecting the expression of key genes of ferroptosis and the level of lipid peroxide. The degree of NASH was judged by observing the morphology of the liver, detecting the expression of inflammation and fibrosis related genes of liver damage. Plin5 was overexpressed in the liver of mice by tail vein injection of adenovirus, and the process of NASH was simulated by methionine choline deficiency (MCD) diet. The occurrence of ferroptosis and NASH was detected by the same detection method. Targeted lipidomics sequencing was used to detect the difference in free fatty acid expression in the WT Plin5 KO group. Finally, it was verified in cell experiments to further study the effect of free fatty acids on ferroptosis of hepatocytes. RESULTS In various NASH models, hepatic Plin5 was dramatically reduced. Plin5 knockout (KO) worsened NASH-associated characteristics in mice given a high-fat/high-cholesterol (HFHC) diet, such as lipid accumulation, inflammation and hepatic fibrosis. It has been shown that ferroptosis is involved in NASH progression. We revealed that Plin5 KO in mice aggravated the degree of ferroptosis in NASH models. Conversely, overexpression of Plin5 significantly alleviated ferroptosis and further ameliorated progression of MCD-induced NASH. Analysis of livers obtained from HFHC diet-fed mice by targeted lipidomics revealed that 11-Dodecenoic acid was significantly decreased in Plin5 KO mice. Addition of 11-Dodecenoia acid to Plin5 knockdown hepatocytes effectively prevented ferroptosis. CONCLUSION Our study demonstrates that Plin5 protects against NASH progression by increasing 11-Dodecenoic acid level and further inhibiting ferroptosis, suggesting that Plin5 has therapeutic potential as a target for the management of NASH.
Collapse
Affiliation(s)
- Xinming Xu
- Department of Endocrinology, Shanghai Fifth People's Hospital, Fudan University, 801 Heqing Road, Shanghai, 200240, China
| | - Jin Qiu
- Shanghai Key Laboratory of Regulatory Biological, Institute of Biomedical Science, School of Life Science, East China Normal University, Shanghai, 200241, China
| | - Xiaoya Li
- Department of Endocrinology, Shanghai Fifth People's Hospital, Fudan University, 801 Heqing Road, Shanghai, 200240, China
| | - Juntong Chen
- Shanghai Key Laboratory of Regulatory Biological, Institute of Biomedical Science, School of Life Science, East China Normal University, Shanghai, 200241, China
| | - Yue Li
- Department of Endocrinology, Shanghai Fifth People's Hospital, Fudan University, 801 Heqing Road, Shanghai, 200240, China
| | - Xinmei Huang
- Department of Endocrinology, Shanghai Fifth People's Hospital, Fudan University, 801 Heqing Road, Shanghai, 200240, China
| | - Shufei Zang
- Department of Endocrinology, Shanghai Fifth People's Hospital, Fudan University, 801 Heqing Road, Shanghai, 200240, China.
| | - Xinran Ma
- Shanghai Key Laboratory of Regulatory Biological, Institute of Biomedical Science, School of Life Science, East China Normal University, Shanghai, 200241, China.
| | - Jun Liu
- Department of Endocrinology, Shanghai Fifth People's Hospital, Fudan University, 801 Heqing Road, Shanghai, 200240, China.
| |
Collapse
|
7
|
Zeng J, Fan J, Zhou H. Bile acid-mediated signaling in cholestatic liver diseases. Cell Biosci 2023; 13:77. [PMID: 37120573 PMCID: PMC10149012 DOI: 10.1186/s13578-023-01035-1] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2023] [Accepted: 04/18/2023] [Indexed: 05/01/2023] Open
Abstract
Chronic cholestatic liver diseases, such as primary biliary cholangitis (PBC) and primary sclerosing cholangitis (PSC), are associated with bile stasis and gradually progress to fibrosis, cirrhosis, and liver failure, which requires liver transplantation. Although ursodeoxycholic acid is effective in slowing the disease progression of PBC, it has limited efficacy in PSC patients. It is challenging to develop effective therapeutic agents due to the limited understanding of disease pathogenesis. During the last decade, numerous studies have demonstrated that disruption of bile acid (BA) metabolism and intrahepatic circulation promotes the progression of cholestatic liver diseases. BAs not only play an essential role in nutrition absorption as detergents but also play an important role in regulating hepatic metabolism and modulating immune responses as key signaling molecules. Several excellent papers have recently reviewed the role of BAs in metabolic liver diseases. This review focuses on BA-mediated signaling in cholestatic liver disease.
Collapse
Affiliation(s)
- Jing Zeng
- Department of Microbiology and Immunology, Medical College of Virginia and Richmond VA Medical Center, Central Virginia Veterans Healthcare System, Virginia Commonwealth University, 1220 East Broad Street, MMRB-5044, Richmond, VA, 23298-0678, USA
- Department of Gastroenterology, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China
| | - Jiangao Fan
- Department of Gastroenterology, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China
| | - Huiping Zhou
- Department of Microbiology and Immunology, Medical College of Virginia and Richmond VA Medical Center, Central Virginia Veterans Healthcare System, Virginia Commonwealth University, 1220 East Broad Street, MMRB-5044, Richmond, VA, 23298-0678, USA.
| |
Collapse
|
8
|
Finamore C, Festa C, Fiorillo B, Leva FSD, Roselli R, Marchianò S, Biagioli M, Spinelli L, Fiorucci S, Limongelli V, Zampella A, De Marino S. Expanding the Library of 1,2,4-Oxadiazole Derivatives: Discovery of New Farnesoid X Receptor (FXR) Antagonists/Pregnane X Receptor (PXR) Agonists. Molecules 2023; 28:molecules28062840. [PMID: 36985811 PMCID: PMC10057480 DOI: 10.3390/molecules28062840] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2023] [Revised: 03/17/2023] [Accepted: 03/18/2023] [Indexed: 03/30/2023] Open
Abstract
Compounds featuring a 1,2,4-oxadiazole core have been recently identified as a new chemotype of farnesoid X receptor (FXR) antagonists. With the aim to expand this class of compounds and to understand the building blocks necessary to maintain the antagonistic activity, we describe herein the synthesis, the pharmacological evaluation, and the in vitro pharmacokinetic properties of a novel series of 1,2,4-oxadiazole derivatives decorated on the nitrogen of the piperidine ring with different N-alkyl and N-aryl side chains. In vitro pharmacological evaluation showed compounds 5 and 11 as the first examples of nonsteroidal dual FXR/Pregnane X receptor (PXR) modulators. In HepG2 cells, these compounds modulated PXR- and FXR-regulated genes, resulting in interesting leads in the treatment of inflammatory disorders. Moreover, molecular docking studies supported the experimental results, disclosing the ligand binding mode and allowing rationalization of the activities of compounds 5 and 11.
Collapse
Affiliation(s)
- Claudia Finamore
- Department of Pharmacy, University of Naples "Federico II", Via D. Montesano 49, 80131 Naples, Italy
| | - Carmen Festa
- Department of Pharmacy, University of Naples "Federico II", Via D. Montesano 49, 80131 Naples, Italy
| | - Bianca Fiorillo
- Department of Pharmacy, University of Naples "Federico II", Via D. Montesano 49, 80131 Naples, Italy
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, 1468 Madison Ave, New York, NY 10029, USA
| | - Francesco Saverio Di Leva
- Department of Pharmacy, University of Naples "Federico II", Via D. Montesano 49, 80131 Naples, Italy
| | - Rosalinda Roselli
- Department of Medicine and Surgery, University of Perugia, Piazza L. Severi, 1-06132 Perugia, Italy
| | - Silvia Marchianò
- Department of Medicine and Surgery, University of Perugia, Piazza L. Severi, 1-06132 Perugia, Italy
| | - Michele Biagioli
- Department of Medicine and Surgery, University of Perugia, Piazza L. Severi, 1-06132 Perugia, Italy
| | - Lucio Spinelli
- Department of Pharmacy, University of Naples "Federico II", Via D. Montesano 49, 80131 Naples, Italy
| | - Stefano Fiorucci
- Department of Medicine and Surgery, University of Perugia, Piazza L. Severi, 1-06132 Perugia, Italy
| | - Vittorio Limongelli
- Department of Pharmacy, University of Naples "Federico II", Via D. Montesano 49, 80131 Naples, Italy
- Faculty of Biomedical Sciences, Euler Institute, Università della Svizzera italiana (USI), Via G. Buffi 13, CH-6900 Lugano, Switzerland
| | - Angela Zampella
- Department of Pharmacy, University of Naples "Federico II", Via D. Montesano 49, 80131 Naples, Italy
| | - Simona De Marino
- Department of Pharmacy, University of Naples "Federico II", Via D. Montesano 49, 80131 Naples, Italy
| |
Collapse
|
9
|
Kim DH, Park JS, Choi HI, Kim CS, Bae EH, Ma SK, Kim SW. The role of the farnesoid X receptor in kidney health and disease: a potential therapeutic target in kidney diseases. Exp Mol Med 2023; 55:304-312. [PMID: 36737665 PMCID: PMC9981614 DOI: 10.1038/s12276-023-00932-2] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2022] [Revised: 11/22/2022] [Accepted: 12/01/2022] [Indexed: 02/05/2023] Open
Abstract
The prevalence of kidney diseases has been increasing worldwide due to the aging population and has results in an increased socioeconomic burden as well as increased morbidity and mortality. A deep understanding of the mechanisms underlying the physiological regulation of the kidney and the pathogenesis of related diseases can help identify potential therapeutic targets. The farnesoid X receptor (FXR, NR1H4) is a primary nuclear bile acid receptor that transcriptionally regulates bile acid homeostasis as well as glucose and lipid metabolism in multiple tissues. The roles of FXR in tissues other than hepatic and intestinal tissues are poorly understood. In studies over the past decade, FXR has been demonstrated to have a protective effect against kidney diseases through its anti-inflammatory and antifibrotic effects; it also plays roles in glucose and lipid metabolism in the kidney. In this review, we discuss the physiological role of FXR in the kidney and its pathophysiological roles in various kidney diseases, including acute kidney injury and chronic kidney diseases, diabetic nephropathy, and kidney fibrosis. Therefore, the regulatory mechanisms involving nuclear receptors, such as FXR, in the physiology and pathophysiology of the kidney and the development of agonists and antagonists for modulating FXR expression and activation should be elucidated to identify therapeutic targets for the treatment of kidney diseases.
Collapse
Affiliation(s)
- Dong-Hyun Kim
- Department of Internal Medicine, Chonnam National University Medical School, Gwangju, 61469, Korea.
| | - Jung Sun Park
- Department of Internal Medicine, Chonnam National University Medical School, Gwangju, 61469, Korea
| | - Hoon-In Choi
- Department of Internal Medicine, Chonnam National University Medical School, Gwangju, 61469, Korea
| | - Chang Seong Kim
- Department of Internal Medicine, Chonnam National University Medical School, Gwangju, 61469, Korea
| | - Eun Hui Bae
- Department of Internal Medicine, Chonnam National University Medical School, Gwangju, 61469, Korea
| | - Seong Kwon Ma
- Department of Internal Medicine, Chonnam National University Medical School, Gwangju, 61469, Korea
| | - Soo Wan Kim
- Department of Internal Medicine, Chonnam National University Medical School, Gwangju, 61469, Korea.
| |
Collapse
|
10
|
Yu L, Liu Y, Wang S, Zhang Q, Zhao J, Zhang H, Narbad A, Tian F, Zhai Q, Chen W. Cholestasis: exploring the triangular relationship of gut microbiota-bile acid-cholestasis and the potential probiotic strategies. Gut Microbes 2023; 15:2181930. [PMID: 36864554 PMCID: PMC9988349 DOI: 10.1080/19490976.2023.2181930] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/10/2022] [Accepted: 02/09/2023] [Indexed: 03/04/2023] Open
Abstract
Cholestasis is a condition characterized by the abnormal production or excretion of bile, and it can be induced by a variety of causes, the factors of which are extremely complex. Although great progress has been made in understanding cholestasis pathogenesis, the specific mechanisms remain unclear. Therefore, it is important to understand and distinguish cholestasis from different etiologies, which will also provide indispensable theoretical support for the development of corresponding therapeutic drugs. At present, the treatment of cholestasis mainly involves several bile acids (BAs) and their derivatives, most of which are in the clinical stage of development. Multiple lines of evidence indicate that ecological disorders of the gut microbiota are strongly related to the occurrence of cholestasis, in which BAs also play a pivotal role. Recent studies indicate that probiotics seem to have certain effects on cholestasis, but further confirmation from clinical trials is required. This paper reviews the etiology of and therapeutic strategies for cholestasis; summarizes the similarities and differences in inducement, symptoms, and mechanisms of related diseases; and provides information about the latest pharmacological therapies currently available and those under research for cholestasis. We also reviewed the highly intertwined relationship between gut microbiota-BA-cholestasis, revealing the potential role and possible mechanism of probiotics in the treatment of cholestasis.
Collapse
Affiliation(s)
- Leilei Yu
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, China
- School of Food Science and Technology, Jiangnan University, Wuxi, China
- International Joint Research Laboratory for Probiotics, Jiangnan University, Wuxi, Jiangsu, China
| | - Yaru Liu
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, China
- School of Food Science and Technology, Jiangnan University, Wuxi, China
| | - Shunhe Wang
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, China
- School of Food Science and Technology, Jiangnan University, Wuxi, China
| | - Qingsong Zhang
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, China
- School of Food Science and Technology, Jiangnan University, Wuxi, China
| | - Jianxin Zhao
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, China
- School of Food Science and Technology, Jiangnan University, Wuxi, China
- International Joint Research Laboratory for Probiotics, Jiangnan University, Wuxi, Jiangsu, China
- National Engineering Research Center for Functional Food, Jiangnan University, Wuxi, China
| | - Hao Zhang
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, China
- School of Food Science and Technology, Jiangnan University, Wuxi, China
- International Joint Research Laboratory for Probiotics, Jiangnan University, Wuxi, Jiangsu, China
- National Engineering Research Center for Functional Food, Jiangnan University, Wuxi, China
| | - Arjan Narbad
- International Joint Research Laboratory for Probiotics, Jiangnan University, Wuxi, Jiangsu, China
- Gut Health and Microbiome Institute Strategic Programme, Quadram Institute Bioscience, Norwich, UK
| | - Fengwei Tian
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, China
- School of Food Science and Technology, Jiangnan University, Wuxi, China
- International Joint Research Laboratory for Probiotics, Jiangnan University, Wuxi, Jiangsu, China
| | - Qixiao Zhai
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, China
- School of Food Science and Technology, Jiangnan University, Wuxi, China
- International Joint Research Laboratory for Probiotics, Jiangnan University, Wuxi, Jiangsu, China
| | - Wei Chen
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, China
- School of Food Science and Technology, Jiangnan University, Wuxi, China
- International Joint Research Laboratory for Probiotics, Jiangnan University, Wuxi, Jiangsu, China
- National Engineering Research Center for Functional Food, Jiangnan University, Wuxi, China
| |
Collapse
|
11
|
Ning Y, Zhou IY, Roberts JD, Rotile NJ, Akam E, Barrett SC, Sojoodi M, Barr MN, Punshon T, Pantazopoulos P, Drescher HK, Jackson BP, Tanabe KK, Caravan P. Molecular MRI quantification of extracellular aldehyde pairs for early detection of liver fibrogenesis and response to treatment. Sci Transl Med 2022; 14:eabq6297. [PMID: 36130015 PMCID: PMC10189657 DOI: 10.1126/scitranslmed.abq6297] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
Liver fibrosis plays a critical role in the evolution of most chronic liver diseases and is characterized by a buildup of extracellular matrix, which can progress to cirrhosis, hepatocellular carcinoma, liver failure, or death. Now, there are no noninvasive methods available to accurately assess disease activity (fibrogenesis) to sensitively detect early onset of fibrosis or to detect early response to treatment. Here, we hypothesized that extracellular allysine aldehyde (LysAld) pairs formed by collagen oxidation during active fibrosis could be a target for assessing fibrogenesis with a molecular probe. We showed that molecular magnetic resonance imaging (MRI) using an extracellular probe targeting these LysAld pairs acts as a noninvasive biomarker of fibrogenesis and demonstrated its high sensitivity and specificity in detecting fibrogenesis in toxin- and dietary-induced mouse models, a cholestasis rat model of liver fibrogenesis, and in human fibrotic liver tissues. Quantitative molecular MRI was highly correlated with fibrogenesis markers and enabled noninvasive detection of early onset fibrosis and response to antifibrotic treatment, showing high potential for clinical translation.
Collapse
Affiliation(s)
- Yingying Ning
- Athinoula A. Martinos Center for Biomedical Imaging, Institute for Innovation in Imaging, Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02129, USA
| | - Iris. Y. Zhou
- Athinoula A. Martinos Center for Biomedical Imaging, Institute for Innovation in Imaging, Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02129, USA
| | - Jesse D. Roberts
- Cardiovascular Research Center of the General Medical Services, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02129, USA
| | - Nicholas J. Rotile
- Athinoula A. Martinos Center for Biomedical Imaging, Institute for Innovation in Imaging, Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02129, USA
| | - Eman Akam
- Athinoula A. Martinos Center for Biomedical Imaging, Institute for Innovation in Imaging, Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02129, USA
| | - Stephen C. Barrett
- Division of Gastrointestinal and Oncologic Surgery, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
| | - Mozhdeh Sojoodi
- Division of Gastrointestinal and Oncologic Surgery, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
| | - Matthew N. Barr
- Department of Earth Sciences, Dartmouth College, Hanover, NH 03766, USA
| | - Tracy Punshon
- Department of Earth Sciences, Dartmouth College, Hanover, NH 03766, USA
| | - Pamela Pantazopoulos
- Athinoula A. Martinos Center for Biomedical Imaging, Institute for Innovation in Imaging, Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02129, USA
| | - Hannah K. Drescher
- Gastrointestinal Unit, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
| | - Brian P. Jackson
- Department of Earth Sciences, Dartmouth College, Hanover, NH 03766, USA
| | - Kenneth K. Tanabe
- Division of Gastrointestinal and Oncologic Surgery, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
| | - Peter Caravan
- Athinoula A. Martinos Center for Biomedical Imaging, Institute for Innovation in Imaging, Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02129, USA
| |
Collapse
|
12
|
Ahmad A, Adda N. Assessment of drug-drug interaction potential with EDP-305, a farnesoid X receptor agonist, in healthy subjects. Clin Transl Sci 2022; 15:2146-2158. [PMID: 35675500 PMCID: PMC9468552 DOI: 10.1111/cts.13348] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Revised: 04/29/2022] [Accepted: 05/21/2022] [Indexed: 01/25/2023] Open
Abstract
EDP-305 is a farnesoid X receptor (FXR) agonist that selectively activates FXR and is a potential treatment for patients with nonalcoholic steatohepatitis (NASH) with liver fibrosis. Results from preclinical studies indicate that CYP3A4 is the primary enzyme involved in EDP-305 metabolism and that EDP-305 has low potential to inhibit or induce cytochrome (CYP) isoenzymes and drug transporters. Four studies were conducted in healthy volunteers to evaluate the drug-drug interaction (DDI) potential of EDP-305 co-administered with drugs known to be substrates for drug metabolizing enzymes or transporters, and to assess the effect of inhibitors and inducers of CYP3A4 on EDP-305. Results suggest caution when substrates of CYP3A4 are administered concomitantly with EDP-305. A potential for increased exposure is apparent when CYP1A2 substrates with a narrow therapeutic index are administered with EDP-305. In contrast, substrates of drug transporters can be administered concomitantly with EDP-305 with a low potential for interactions. Coadministration of EDP-305 and a combined OC had no relevant effects on plasma concentrations of the combined OC. Co-administration of EDP-305 with strong or moderate inhibitors and inducers of CYP3A4 is not recommended. These results indicate low overall likelihood of interaction of EDP-305 and other substrates through CYP mediated interactions. The interaction potential of EDP-305 with drug transporters was low and of unlikely clinical significance. The EDP-305 DDI profile allows for convenient administration in patients with NASH and other patient populations with comorbidities, with minimal dose modification of concomitant medications.
Collapse
Affiliation(s)
- Alaa Ahmad
- Enanta Pharmaceuticals Inc.WatertownMassachusettsUSA
| | - Nathalie Adda
- Enanta Pharmaceuticals Inc.WatertownMassachusettsUSA
| |
Collapse
|
13
|
Duan S, Li X, Fan G, Liu R. Targeting bile acid signaling for the treatment of liver diseases: From bench to bed. Biomed Pharmacother 2022; 152:113154. [PMID: 35653887 DOI: 10.1016/j.biopha.2022.113154] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2022] [Revised: 05/13/2022] [Accepted: 05/15/2022] [Indexed: 11/02/2022] Open
Abstract
Liver diseases and related complications have become one of the leading causes of morbidity and mortality worldwide, yet effective medicine or approved treatment approach is still limited. Thus, novel therapy is urgently required to prevent or at least slow down the growing burden of liver transplantation or even death caused by malignant liver diseases. As the irreplaceable modulator of hepatic and intestinal signaling cascades, bile acids (BAs) play complex physiological as well as pathological roles in regulating energy and immune homeostasis in various liver diseases, including but not limited to metabolic diseases and cholangiopathies, making them highly attractive therapeutic targets. In the current review, recent progress in the research of enterohepatic circulation of BAs and potential therapeutic targets of BAs signaling, especially the development of currently available treatments, including agonizts of FXR and TGR5, analogs of FGF19, inhibitors of ASBT, and the regulation of gut microbiome through fecal microbiota transplantation were extensively summarized. Their protective effects, molecular mechanisms, and outcomes of clinical trials were highlighted. The structural features of these candidates and perspectives for their future development were further discussed. In conclusion, we believe that pharmacological therapies targeting BAs signaling represent promising and efficient strategies for the treatment of complex and multifactorial liver disorders.
Collapse
Affiliation(s)
- Shuni Duan
- School of Materia Medica, Beijing University of Chinese Medicine, 11 Bei San Huan Dong Lu, Beijing 100029, China
| | - Xiaojiaoyang Li
- School of Life Sciences, Beijing University of Chinese Medicine, 11 Bei San Huan Dong Lu, Beijing 100029, China
| | - Guifang Fan
- School of Materia Medica, Beijing University of Chinese Medicine, 11 Bei San Huan Dong Lu, Beijing 100029, China
| | - Runping Liu
- School of Materia Medica, Beijing University of Chinese Medicine, 11 Bei San Huan Dong Lu, Beijing 100029, China.
| |
Collapse
|
14
|
Liver Steatosis: A Marker of Metabolic Risk in Children. Int J Mol Sci 2022; 23:ijms23094822. [PMID: 35563210 PMCID: PMC9100068 DOI: 10.3390/ijms23094822] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2022] [Revised: 04/24/2022] [Accepted: 04/24/2022] [Indexed: 11/16/2022] Open
Abstract
Obesity is one of the greatest health challenges affecting children of all ages and ethnicities. Almost 19% of children and adolescents worldwide are overweight or obese, with an upward trend in the last decades. These reports imply an increased risk of fat accumulation in hepatic cells leading to a series of histological hepatic damages gathered under the acronym NAFLD (Non-Alcoholic Fatty Liver Disease). Due to the complex dynamics underlying this condition, it has been recently renamed as 'Metabolic Dysfunction Associated Fatty Liver Disease (MAFLD)', supporting the hypothesis that hepatic steatosis is a key component of the large group of clinical and laboratory abnormalities of Metabolic Syndrome (MetS). This review aims to share the latest scientific knowledge on MAFLD in children in an attempt to offer novel insights into the complex dynamics underlying this condition, focusing on the novel molecular aspects. Although there is still no treatment with a proven efficacy for this condition, starting from the molecular basis of the disease, MAFLD's therapeutic landscape is rapidly expanding, and different medications seem to act as modifiers of liver steatosis, inflammation, and fibrosis.
Collapse
|
15
|
Ratziu V, Rinella ME, Neuschwander-Tetri BA, Lawitz E, Denham D, Kayali Z, Sheikh A, Kowdley KV, Desta T, Elkhashab M, DeGrauw J, Goodwin B, Ahmad A, Adda N. EDP-305 in patients with NASH: A phase II double-blind placebo-controlled dose-ranging study. J Hepatol 2022; 76:506-517. [PMID: 34740705 DOI: 10.1016/j.jhep.2021.10.018] [Citation(s) in RCA: 52] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/21/2021] [Revised: 10/12/2021] [Accepted: 10/13/2021] [Indexed: 12/11/2022]
Abstract
BACKGROUND & AIMS EDP-305 is an oral farnesoid X receptor (FXR) agonist under development for the treatment of non-alcoholic steatohepatitis (NASH). Herein, we aimed to assess the efficacy, safety and tolerability of EDP-305 in patients with fibrotic NASH. METHODS In this double-blind phase II study, patients with fibrotic NASH (without cirrhosis), diagnosed by historical biopsy or phenotypically, were randomized to EDP-305 1 mg, EDP-305 2.5 mg, or placebo, for 12 weeks. The primary endpoint was mean change in alanine aminotransferase (ALT) from baseline to Week 12, and the key secondary endpoint was mean change in liver fat content from baseline to Week 12. RESULTS Between January 2018 and July 2019, 134 patients were randomized and 132 were evaluated. At Week 12, the least squares mean reductions from baseline in ALT for patients receiving 2.5 mg EDP-305 and 1 mg EDP-305 were -27.9 U/L (95% CI 0.03 to 24.9; p = 0.049) and -21.7 U/L (-5.8 to 18.3: p = 0.304), respectively, compared to -15.4 U/L for those receiving placebo. Absolute liver fat reduction was -7.1% (2.0-7.5; p = 0.0009) with 2.5 mg EDP-305, -3.3% with EDP-305 1 mg, and -2.4% with placebo. The most common (≥5%) adverse events were pruritus, nausea, vomiting, diarrhea, headache, and dizziness. Pruritus occurred in 50.9%, 9.1%, and 4.2% of patients in the 2.5 mg, 1 mg, and placebo groups, respectively, and led to study drug discontinuation in 20.8% of patients in the 2.5 mg group and 1.8% in the 1 mg group. CONCLUSIONS EDP-305 reduced ALT levels and liver fat content, providing support for a longer-term trial assessing histological endpoints in patients with NASH. CLINICALTRIALS. GOV NUMBER NCT03421431 LAY SUMMARY: Non-alcoholic fatty liver disease is a chronic hepatic disease that can progress to non-alcoholic steatohepatitis (NASH), which is associated with an increased risk of cirrhosis and liver cancer. Results from this phase II study support continued development of EDP-305, an oral farnesoid X receptor agonist, for the treatment of patients with NASH.
Collapse
Affiliation(s)
- Vlad Ratziu
- Sorbonne Université, ICAN, Hospital Pitié-Salpêtrière, INSERM UMRS 1138 CRC, Paris, France
| | - Mary E Rinella
- Northwestern University Feinberg School of Medicine, Chicago, IL, United States
| | | | - Eric Lawitz
- Texas Liver Institute, San Antonio, TX United States
| | - Douglas Denham
- Clinical Trials of Texas, Inc., San Antonio, TX, United States
| | - Zeid Kayali
- Inland Empire Liver Foundation, Rialto, CA, United States
| | - Aasim Sheikh
- GI Specialists of Georgia, Marietta, GA United States
| | - Kris V Kowdley
- Liver Institute Northwest and Elson S. Floyd College of Medicine, Washington State University, Seattle, WA, United States
| | - Taddese Desta
- Precision Research Institute, San Diego, CA, United States
| | | | - Jeffery DeGrauw
- Synexus - Wasatch Peak Family Practice, Layton, UT, United States
| | - Bryan Goodwin
- Enanta Pharmaceuticals, Inc, Watertown, MA, United States
| | - Alaa Ahmad
- Enanta Pharmaceuticals, Inc, Watertown, MA, United States
| | - Nathalie Adda
- Enanta Pharmaceuticals, Inc, Watertown, MA, United States.
| |
Collapse
|
16
|
Zhong Y, Mahoney RC, Khatun Z, Chen HH, Nguyen CT, Caravan P, Roberts JD. Lysyl oxidase regulation and protein aldehydes in the injured newborn lung. Am J Physiol Lung Cell Mol Physiol 2022; 322:L204-L223. [PMID: 34878944 PMCID: PMC8794022 DOI: 10.1152/ajplung.00158.2021] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
During newborn lung injury, excessive activity of lysyl oxidases (LOXs) disrupts extracellular matrix (ECM) formation. Previous studies indicate that TGFβ activation in the O2-injured mouse pup lung increases lysyl oxidase (LOX) expression. But how TGFβ regulates this, and whether the LOXs generate excess pulmonary aldehydes are unknown. First, we determined that O2-mediated lung injury increases LOX protein expression in TGFβ-stimulated pup lung interstitial fibroblasts. This regulation appeared to be direct; this is because TGFβ treatment also increased LOX protein expression in isolated pup lung fibroblasts. Then using a fibroblast cell line, we determined that TGFβ stimulates LOX expression at a transcriptional level via Smad2/3-dependent signaling. LOX is translated as a pro-protein that requires secretion and extracellular cleavage before assuming amine oxidase activity and, in some cells, reuptake with nuclear localization. We found that pro-LOX is processed in the newborn mouse pup lung. Also, O2-mediated injury was determined to increase pro-LOX secretion and nuclear LOX immunoreactivity particularly in areas populated with interstitial fibroblasts and exhibiting malformed ECM. Then, using molecular probes, we detected increased aldehyde levels in vivo in O2-injured pup lungs, which mapped to areas of increased pro-LOX secretion in lung sections. Increased activity of LOXs plays a critical role in the aldehyde generation; an inhibitor of LOXs prevented the elevation of aldehydes in the O2-injured pup lung. These results reveal new mechanisms of TGFβ and LOX in newborn lung disease and suggest that aldehyde-reactive probes might have utility in sensing the activation of LOXs in vivo during lung injury.
Collapse
Affiliation(s)
- Ying Zhong
- 1Cardiovascular Research Center of the General Medical Services, Massachusetts General Hospital, Boston, Massachusetts,4Harvard Medical School, Harvard University, Cambridge, Massachusetts
| | - Rose C. Mahoney
- 1Cardiovascular Research Center of the General Medical Services, Massachusetts General Hospital, Boston, Massachusetts
| | - Zehedina Khatun
- 4Harvard Medical School, Harvard University, Cambridge, Massachusetts,5Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Boston, Massachusetts,6Division of Health Science Technology, Harvard-Massachusetts Institute of Technology, Cambridge, Massachusetts
| | - Howard H. Chen
- 4Harvard Medical School, Harvard University, Cambridge, Massachusetts,5Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Boston, Massachusetts,6Division of Health Science Technology, Harvard-Massachusetts Institute of Technology, Cambridge, Massachusetts
| | - Christopher T. Nguyen
- 1Cardiovascular Research Center of the General Medical Services, Massachusetts General Hospital, Boston, Massachusetts,4Harvard Medical School, Harvard University, Cambridge, Massachusetts,5Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Boston, Massachusetts
| | - Peter Caravan
- 4Harvard Medical School, Harvard University, Cambridge, Massachusetts,5Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Boston, Massachusetts,6Division of Health Science Technology, Harvard-Massachusetts Institute of Technology, Cambridge, Massachusetts,7The Institute for Innovation in Imaging, Massachusetts General Hospital, Boston, Massachusetts
| | - Jesse D. Roberts
- 1Cardiovascular Research Center of the General Medical Services, Massachusetts General Hospital, Boston, Massachusetts,2Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Boston, Massachusetts,3Department of Pediatrics, Massachusetts General Hospital, Boston, Massachusetts,4Harvard Medical School, Harvard University, Cambridge, Massachusetts
| |
Collapse
|
17
|
Albhaisi S, Sanyal AJ. Pharmacology of NASH. COMPREHENSIVE PHARMACOLOGY 2022:214-238. [DOI: 10.1016/b978-0-12-820472-6.00121-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
18
|
Negi CK, Babica P, Bajard L, Bienertova-Vasku J, Tarantino G. Insights into the molecular targets and emerging pharmacotherapeutic interventions for nonalcoholic fatty liver disease. Metabolism 2022; 126:154925. [PMID: 34740573 DOI: 10.1016/j.metabol.2021.154925] [Citation(s) in RCA: 147] [Impact Index Per Article: 49.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/12/2021] [Revised: 10/27/2021] [Accepted: 10/28/2021] [Indexed: 12/14/2022]
Abstract
Nonalcoholic fatty liver disease (NAFLD) is the most common form of chronic liver disease worldwide. With no Food and Drug Administration approved drugs, current treatment options include dietary restrictions and lifestyle modification. NAFLD is closely associated with metabolic disorders such as obesity, type 2 diabetes, and dyslipidemia. Hence, clinically various pharmacological approaches using existing drugs such as antidiabetic, anti-obesity, antioxidants, and cytoprotective agents have been considered in the management of NAFLD and nonalcoholic steatohepatitis (NASH). However, several pharmacological therapies aiming to alleviate NAFLD-NASH are currently being examined at various phases of clinical trials. Emerging data from these studies with drugs targeting diverse molecular mechanisms show promising outcomes. This review summarizes the current understanding of the pathogenic mechanisms of NAFLD and provides an insight into the pharmacological targets and emerging therapeutics with specific interventional mechanisms. In addition, we also discuss the importance and utility of new approach methodologies and regulatory perspectives for NAFLD-NASH drug development.
Collapse
Affiliation(s)
- Chander K Negi
- RECETOX, Faculty of Science, Masaryk University, Kotlarska 2, Brno, Czech Republic
| | - Pavel Babica
- RECETOX, Faculty of Science, Masaryk University, Kotlarska 2, Brno, Czech Republic.
| | - Lola Bajard
- RECETOX, Faculty of Science, Masaryk University, Kotlarska 2, Brno, Czech Republic
| | - Julie Bienertova-Vasku
- RECETOX, Faculty of Science, Masaryk University, Kotlarska 2, Brno, Czech Republic; Department of Pathological Physiology, Faculty of Medicine, Masaryk University, Brno, Czech Republic
| | - Giovanni Tarantino
- Department of Clinical Medicine and Surgery, Federico II University Medical School of Naples, Naples, Italy
| |
Collapse
|
19
|
Fang Y, Hegazy L, Finck BN, Elgendy B. Recent Advances in the Medicinal Chemistry of Farnesoid X Receptor. J Med Chem 2021; 64:17545-17571. [PMID: 34889100 DOI: 10.1021/acs.jmedchem.1c01017] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Farnesoid X receptor (FXR) is an important regulator of bile acid, lipid, amino acid, and glucose homeostasis, hepatic inflammation, regeneration, and fibrosis. FXR has been recognized as a promising drug target for various metabolic diseases such as lipid disorders, nonalcoholic fatty liver disease (NAFLD), nonalcoholic steatohepatitis (NASH), and chronic kidney disease. A large number of FXR ligands have been developed by pharmaceutical companies and academic institutions, and several candidates have progressed into clinical trials in the past decade. However, it is continually a challenge to discover drugs targeting FXR due to side effects associated with long-term administration. In this perspective, we summarize the research progress on medicinal chemistry of FXR modulators from 2018 to the present by discussing the diverse structures of synthetic FXR modulators including steroidal and non-steroidal ligands, their structure-activity relationships (SARs), and their therapeutic applications.
Collapse
Affiliation(s)
- Yuanying Fang
- Department of Pharmaceutical and Administrative Sciences, University of Health Sciences and Pharmacy, St. Louis, Missouri 63110, United States.,Center for Clinical Pharmacology, Washington University School of Medicine and St. Louis College of Pharmacy, St. Louis, Missouri 63110, United States
| | - Lamees Hegazy
- Department of Pharmaceutical and Administrative Sciences, University of Health Sciences and Pharmacy, St. Louis, Missouri 63110, United States.,Center for Clinical Pharmacology, Washington University School of Medicine and St. Louis College of Pharmacy, St. Louis, Missouri 63110, United States
| | - Brian N Finck
- Department of Medicine, Division of Geriatrics and Nutritional Science, Washington University School of Medicine, St. Louis, Missouri 63110, United States
| | - Bahaa Elgendy
- Department of Pharmaceutical and Administrative Sciences, University of Health Sciences and Pharmacy, St. Louis, Missouri 63110, United States.,Center for Clinical Pharmacology, Washington University School of Medicine and St. Louis College of Pharmacy, St. Louis, Missouri 63110, United States.,Chemistry Department, Faculty of Science, Benha University, Benha 13518, Egypt
| |
Collapse
|
20
|
Qu W, Ma T, Cai J, Zhang X, Zhang P, She Z, Wan F, Li H. Liver Fibrosis and MAFLD: From Molecular Aspects to Novel Pharmacological Strategies. Front Med (Lausanne) 2021; 8:761538. [PMID: 34746195 PMCID: PMC8568774 DOI: 10.3389/fmed.2021.761538] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2021] [Accepted: 09/27/2021] [Indexed: 12/11/2022] Open
Abstract
Metabolic-associated fatty liver disease (MAFLD) is a new disease definition, and this nomenclature MAFLD was proposed to renovate its former name, non-alcoholic fatty liver disease (NAFLD). MAFLD/NAFLD have shared and predominate causes from nutrition overload to persistent liver damage and eventually lead to the development of liver fibrosis and cirrhosis. Unfortunately, there is an absence of effective treatments to reverse MAFLD/NAFLD-associated fibrosis. Due to the significant burden of MAFLD/NAFLD and its complications, there are active investigations on the development of novel targets and pharmacotherapeutics for treating this disease. In this review, we cover recent discoveries in new targets and molecules for antifibrotic treatment, which target pathways intertwined with the fibrogenesis process, including lipid metabolism, inflammation, cell apoptosis, oxidative stress, and extracellular matrix formation. Although marked advances have been made in the development of antifibrotic therapeutics, none of the treatments have achieved the endpoints evaluated by liver biopsy or without significant side effects in a large-scale trial. In addition to the discovery of new druggable targets and pharmacotherapeutics, personalized medication, and combinatorial therapies targeting multiple profibrotic pathways could be promising in achieving successful antifibrotic interventions in patients with MAFLD/NAFLD.
Collapse
Affiliation(s)
- Weiyi Qu
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
- Institute of Model Animal, Wuhan University, Wuhan, China
| | - Tengfei Ma
- Institute of Model Animal, Wuhan University, Wuhan, China
- Department of Neurology, Huanggang Central Hospital, Huanggang, China
- Huanggang Institute of Translational Medicine, Huanggang Central Hospital, Huanggang, China
| | - Jingjing Cai
- Institute of Model Animal, Wuhan University, Wuhan, China
- Department of Cardiology, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Xiaojing Zhang
- Institute of Model Animal, Wuhan University, Wuhan, China
- School of Basic Medical Sciences, Wuhan University, Wuhan, China
| | - Peng Zhang
- Institute of Model Animal, Wuhan University, Wuhan, China
- School of Basic Medical Sciences, Wuhan University, Wuhan, China
| | - Zhigang She
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
- Institute of Model Animal, Wuhan University, Wuhan, China
| | - Feng Wan
- Department of Neurology, Huanggang Central Hospital, Huanggang, China
| | - Hongliang Li
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
- Institute of Model Animal, Wuhan University, Wuhan, China
- Huanggang Institute of Translational Medicine, Huanggang Central Hospital, Huanggang, China
| |
Collapse
|
21
|
Gastaldelli A, Stefan N, Häring HU. Liver-targeting drugs and their effect on blood glucose and hepatic lipids. Diabetologia 2021; 64:1461-1479. [PMID: 33877366 PMCID: PMC8187191 DOI: 10.1007/s00125-021-05442-2] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/09/2020] [Accepted: 12/18/2020] [Indexed: 12/16/2022]
Abstract
The global epidemic of non-alcoholic fatty liver disease (NAFLD) and steatohepatitis (NASH) and the high prevalence among individuals with type 2 diabetes has attracted the attention of clinicians specialising in liver disorders. Many drugs are in the pipeline for the treatment of NAFLD/NASH, and several glucose-lowering drugs are now being tested specifically for the treatment of liver disease. Among these are nuclear hormone receptor agonists (e.g. peroxisome proliferator-activated receptor agonists, farnesoid X receptor agonists and liver X receptor agonists), fibroblast growth factor-19 and -21, single, dual or triple incretins, sodium-glucose cotransporter inhibitors, drugs that modulate lipid or other metabolic pathways (e.g. inhibitors of fatty acid synthase, diacylglycerol acyltransferase-1, acetyl-CoA carboxylase and 11β-hydroxysteroid dehydrogenase type-1) or drugs that target the mitochondrial pyruvate carrier. We have reviewed the metabolic effects of these drugs in relation to improvement of diabetic hyperglycaemia and fatty liver disease, as well as peripheral metabolism and insulin resistance.
Collapse
Affiliation(s)
- Amalia Gastaldelli
- Institute of Clinical Physiology, National Research Council-CNR, Pisa, Italy.
| | - Norbert Stefan
- Department of Internal Medicine IV, University of Tübingen, Tübingen, Germany.
- Institute of Diabetes Research and Metabolic Diseases of the Helmholtz Center Munich, Tübingen, Germany.
- German Center for Diabetes Research, Neuherberg, Germany.
| | - Hans-Ulrich Häring
- Department of Internal Medicine IV, University of Tübingen, Tübingen, Germany
- Institute of Diabetes Research and Metabolic Diseases of the Helmholtz Center Munich, Tübingen, Germany
- German Center for Diabetes Research, Neuherberg, Germany
| |
Collapse
|
22
|
Clinical Management of Primary Biliary Cholangitis-Strategies and Evolving Trends. Clin Rev Allergy Immunol 2021; 59:175-194. [PMID: 31713023 DOI: 10.1007/s12016-019-08772-7] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
PBC is a chronic progressive autoimmune disorder involving the destruction of intrahepatic small bile ducts, cholestasis, fibrosis, and ultimately cirrhosis if left untreated. It is largely driven by the autoimmune response, but bile acids and the intestinal microbiota are implicated in disease progression as well. The only drugs licensed for PBC are UDCA and OCA. UDCA as a first-line and OCA as a second-line therapy are safe and effective, but the lack of response in a significant portion of patients and inadequate control of symptoms such as fatigue and pruritus remain as concerns. Liver transplantation is an end-stage therapy for many patients refractory to UDCA, which gives excellent survival rates but also moderate to high recurrence rates. The limited options for FDA-approved PBC therapies necessitate the development of alternative approaches. Currently, a wide variety of experimental drugs exist targeting immunological and physiological aspects of PBC to suppress inflammation. Immunological therapies include drugs targeting immune molecules in the B cell and T cell response, and specific cytokines and chemokines implicated in inflammation. Drugs targeting bile acids are also noteworthy as bile acids can perpetuate hepatic inflammation and lead to fibrosis over time. These include FXR agonists, ASBT inhibitors, and PPAR agonists such as bezafibrate and fenofibrate. Nonetheless, many of these drugs can only delay disease progression and fail to enhance patients' quality of life. Nanomedicine shows great potential for treatment of autoimmune diseases, as it provides a new approach that focuses on tolerance induction rather than immunosuppression. Tolerogenic nanoparticles carrying immune-modifying agents can be engineered to safely and effectively target the antigen-specific immune response in autoimmune diseases. These may work well with PBC especially, given the anatomical features and immunological specificity of the disease. Nanobiological therapy is thus an area of highly promising research for future treatment of PBC.
Collapse
|
23
|
Abstract
Our understanding of nonalcoholic fatty liver disease pathophysiology continues to advance rapidly. Accordingly, the field has moved from describing the clinical phenotype through the presence of nonalcoholic steatohepatitis (NASH) and degree of fibrosis to deep phenotyping with a description of associated comorbidities, genetic polymorphisms and environmental influences that could be associated with disease progression. These insights have fuelled a robust therapeutic pipeline across a variety of new targets to resolve steatohepatitis or reverse fibrosis, or both. Additionally, some of these therapies have beneficial effects that extend beyond the liver, such as effects on glycaemic control, lipid profile and weight loss. In addition, emerging therapies for NASH cirrhosis would have to demonstrate either reversal of fibrosis with associated reduction in portal hypertension or at least delay the progression with eventual decrease in liver-related outcomes. For non-cirrhotic NASH, it is the expectation that reversal of fibrosis by one stage or resolution of NASH with no worsening in fibrosis will need to be accompanied by overall survival benefits. In this Review, we summarize NASH therapies that have progressed to phase II and beyond. We also discuss some of the potential clinical challenges with the use of these new therapies when approved.
Collapse
|
24
|
Li H, Guan Y, Han C, Zhang Y, Liu Q, Wei W, Ma Y. The pathogenesis, models and therapeutic advances of primary biliary cholangitis. Biomed Pharmacother 2021; 140:111754. [PMID: 34044277 DOI: 10.1016/j.biopha.2021.111754] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2021] [Revised: 05/17/2021] [Accepted: 05/19/2021] [Indexed: 12/30/2022] Open
Abstract
Primary biliary cholangitis (PBC) is an autoimmune disease characterized by the destruction of intrahepatic small bile ducts and the presence of antimitochondrial antibody (AMA), eventually progresses to liver fibrosis and cirrhosis. Genetic predisposition and environmental factors are involved in the occurrence of PBC, and the epitopes exposure and the imbalance of autoimmune tolerance are the last straw. The apoptosis of biliary epithelial cell (BEC) leads to the release of autoantigen epitopes, which activate the immune system, and the disorder of innate and adaptive immunity eventually leads to the start of disease. Animal models have unique advantages in investigating the pathogenesis and drug exploitation of PBC. Multiple models have been reported, and spontaneous model and induced model have been widely used in relevant research of PBC in recent years. Currently, the only drugs licensed for PBC are ursodesoxycholic acid (UDCA) and obeticholic acid (OCA). In the last few years, as the learned more about the pathogenesis of PBC, more and more targets have been discovered, and multiple targeted drugs are being in developed. In this review, the pathogenesis, murine models and treatment strategies of PBC were summarized, and the current research status was discussed to provide insights for the further study of PBC.
Collapse
Affiliation(s)
- Hao Li
- Institute of Clinical Pharmacology, Anhui Medical University, Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Anhui Collaborative Innovation Center of Anti-inflammatory and Immune Medicine, Hefei, China
| | - Yanling Guan
- Institute of Clinical Pharmacology, Anhui Medical University, Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Anhui Collaborative Innovation Center of Anti-inflammatory and Immune Medicine, Hefei, China
| | - Chenchen Han
- Institute of Clinical Pharmacology, Anhui Medical University, Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Anhui Collaborative Innovation Center of Anti-inflammatory and Immune Medicine, Hefei, China
| | - Yu Zhang
- Institute of Clinical Pharmacology, Anhui Medical University, Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Anhui Collaborative Innovation Center of Anti-inflammatory and Immune Medicine, Hefei, China
| | - Qian Liu
- Institute of Clinical Pharmacology, Anhui Medical University, Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Anhui Collaborative Innovation Center of Anti-inflammatory and Immune Medicine, Hefei, China
| | - Wei Wei
- Institute of Clinical Pharmacology, Anhui Medical University, Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Anhui Collaborative Innovation Center of Anti-inflammatory and Immune Medicine, Hefei, China.
| | - Yang Ma
- Institute of Clinical Pharmacology, Anhui Medical University, Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Anhui Collaborative Innovation Center of Anti-inflammatory and Immune Medicine, Hefei, China.
| |
Collapse
|
25
|
Jiang L, Zhang H, Xiao D, Wei H, Chen Y. Farnesoid X receptor (FXR): Structures and ligands. Comput Struct Biotechnol J 2021; 19:2148-2159. [PMID: 33995909 PMCID: PMC8091178 DOI: 10.1016/j.csbj.2021.04.029] [Citation(s) in RCA: 103] [Impact Index Per Article: 25.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2020] [Revised: 04/10/2021] [Accepted: 04/10/2021] [Indexed: 02/07/2023] Open
Abstract
Farnesoid X receptor (FXR) is a bile acid activated nuclear receptor (BAR) and is mainly expressed in the liver and intestine. Upon ligand binding, FXR regulates key genes involved in the metabolic process of bile acid synthesis, transport and reabsorption and is also involved in the metabolism of carbohydrates and lipids. Because of its important functions, FXR is considered as a promising drug target for the therapy of bile acid-related liver diseases. With the approval of obeticholic acid (OCA) as the first small molecule to target FXR, many other small molecules are being evaluated in clinical trials. This review summarizes the structures of FXR, especially its ligand binding domain, and the development of small molecules (including agonists and antagonists) targeting FXR.
Collapse
Affiliation(s)
- Longying Jiang
- Department of Pathology, NHC Key Laboratory of Cancer Proteomics, Laboratory of Structural Biology, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
- Department of Oncology, NHC Key Laboratory of Cancer Proteomics, Laboratory of Structural Biology, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
| | - Huajun Zhang
- Department of Oncology, NHC Key Laboratory of Cancer Proteomics, Laboratory of Structural Biology, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
| | - Desheng Xiao
- Department of Pathology, NHC Key Laboratory of Cancer Proteomics, Laboratory of Structural Biology, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
| | - Hudie Wei
- Department of Pathology, NHC Key Laboratory of Cancer Proteomics, Laboratory of Structural Biology, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
- Department of Oncology, NHC Key Laboratory of Cancer Proteomics, Laboratory of Structural Biology, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
| | - Yongheng Chen
- Department of Pathology, NHC Key Laboratory of Cancer Proteomics, Laboratory of Structural Biology, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
- Department of Oncology, NHC Key Laboratory of Cancer Proteomics, Laboratory of Structural Biology, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
| |
Collapse
|
26
|
dos Santos Ferreira D, Arora G, Gieseck RL, Rotile NJ, Waghorn PA, Tanabe KK, Wynn TA, Caravan P, Fuchs BC. Molecular Magnetic Resonance Imaging of Liver Fibrosis and Fibrogenesis Is Not Altered by Inflammation. Invest Radiol 2021; 56:244-251. [PMID: 33109919 PMCID: PMC7956154 DOI: 10.1097/rli.0000000000000737] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
METHODS Three groups of mice that develop either mild type 2 inflammation and fibrosis (wild type), severe fibrosis with exacerbated type 2 inflammation (Il10-/-Il12b-/-Il13ra2-/-), or minimal fibrosis with marked type 1 inflammation (Il4ra∂/∂) after infection with S. mansoni were imaged using both probes for determination of signal enhancement. Schistosoma mansoni-infected wild-type mice developed chronic liver fibrosis. RESULTS The liver MR signal enhancement after either probe administration was significantly higher in S. mansoni-infected wild-type mice compared with naive animals. The S. mansoni-infected Il4ra∂/∂ mice presented with little liver signal enhancement after probe injection despite the presence of substantial inflammation. Schistosoma mansoni-infected Il10-/-Il12b-/-Il13ra2-/- mice presented with marked fibrosis, which correlated to increased signal enhancement after injection of either probe. CONCLUSIONS Both MR probes, EP-3533 and Gd-Hyd, were specific for fibrosis in this model of chronic liver disease regardless of the presence or severity of the underlying inflammation. These results, in addition to previous findings, show the potential application of both molecular MR probes for detection and quantification of fibrosis from various etiologies.
Collapse
Affiliation(s)
- Diego dos Santos Ferreira
- Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02129 USA
| | - Gunisha Arora
- Division of Surgical Oncology, Massachusetts General Hospital Cancer Center and Harvard Medical School, Boston, MA 02114 USA
| | - Richard L. Gieseck
- Laboratory of Parasitic Disease, National Institute of Allergy and Infectious Diseases, National Institutes of Health, 5601 Fishers Ln, Bethesda, MD, 20892, United States
| | - Nicholas J. Rotile
- Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02129 USA
| | - Philip A. Waghorn
- Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02129 USA
| | - Kenneth K. Tanabe
- Division of Surgical Oncology, Massachusetts General Hospital Cancer Center and Harvard Medical School, Boston, MA 02114 USA
| | - Thomas A. Wynn
- Laboratory of Parasitic Disease, National Institute of Allergy and Infectious Diseases, National Institutes of Health, 5601 Fishers Ln, Bethesda, MD, 20892, United States
| | - Peter Caravan
- Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02129 USA
- The Institute for Innovation in Imaging (i), Department of Radiology, Massachusetts General Hospital, Boston, MA 02129 USA
| | - Bryan C. Fuchs
- Division of Surgical Oncology, Massachusetts General Hospital Cancer Center and Harvard Medical School, Boston, MA 02114 USA
| |
Collapse
|
27
|
Quantitative, noninvasive MRI characterization of disease progression in a mouse model of non-alcoholic steatohepatitis. Sci Rep 2021; 11:6105. [PMID: 33731798 PMCID: PMC7971064 DOI: 10.1038/s41598-021-85679-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2020] [Accepted: 02/28/2021] [Indexed: 12/17/2022] Open
Abstract
Non-alcoholic steatohepatitis (NASH) is an increasing cause of chronic liver disease characterized by steatosis, inflammation, and fibrosis which can lead to cirrhosis, hepatocellular carcinoma, and mortality. Quantitative, noninvasive methods for characterizing the pathophysiology of NASH at both the preclinical and clinical level are sorely needed. We report here a multiparametric magnetic resonance imaging (MRI) protocol with the fibrogenesis probe Gd-Hyd to characterize fibrotic disease activity and steatosis in a common mouse model of NASH. Mice were fed a choline-deficient, L-amino acid-defined, high-fat diet (CDAHFD) to induce NASH with advanced fibrosis. Mice fed normal chow and CDAHFD underwent MRI after 2, 6, 10 and 14 weeks to measure liver T1, T2*, fat fraction, and dynamic T1-weighted Gd-Hyd enhanced imaging of the liver. Steatosis, inflammation, and fibrosis were then quantified by histology. NASH and fibrosis developed quickly in CDAHFD fed mice with strong correlation between morphometric steatosis quantification and liver fat estimated by MRI (r = 0.90). Sirius red histology and collagen quantification confirmed increasing fibrosis over time (r = 0.82). Though baseline T1 and T2* measurements did not correlate with fibrosis, Gd-Hyd signal enhancement provided a measure of the extent of active fibrotic disease progression and correlated strongly with lysyl oxidase expression. Gd-Hyd MRI accurately detects fibrogenesis in a mouse model of NASH with advanced fibrosis and can be combined with other MR measures, like fat imaging, to more accurately assess disease burden.
Collapse
|
28
|
Rupcic Rubin V, Bojanic K, Smolic M, Rubin J, Tabll A, Smolic R. An Update on Efficacy and Safety of Emerging Hepatic Antifibrotic Agents. J Clin Transl Hepatol 2021. [PMID: 33604256 DOI: 10.14218/jcth.2020.00040.] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/11/2022] Open
Abstract
Liver fibrosis represents a response to chronic liver injury. Metabolic dysfunction-associated fatty liver disease and metabolic dysfunction-associated steatohepatitis are the most common chronic liver diseases, both with increasing incidence. Therefore, there is a great impetus for development of agents targeting these conditions. Accumulating data on possible treatment options for liver fibrosis are emerging in the literature. However, despite extensive research and much effort in the field, approved agents for liver fibrosis are still lacking. In this critical review, we have summarized the main data about specific treatment options for liver fibrosis gained from ongoing clinical trials, with an emphasis on efficacy and safety of these agents.
Collapse
Affiliation(s)
- Vinka Rupcic Rubin
- Department of Gynaecology and Obstetrics, Osijek University Hospital Centre, Faculty of Medicine, Josip Juraj Strossmayer University of Osijek, Osijek, Croatia
| | - Kristina Bojanic
- Department of Biophysics and Radiology, Faculty of Dental Medicine and Health Osijek, J. J. Strossmayer University of Osijek, Osijek, Croatia.,Department of Biophysics and Radiology, Faculty of Medicine Osijek, J. J. Strossmayer University of Osijek, Osijek, Croatia.,Department of Radiology, Health Center Osijek, Osijek, Croatia
| | - Martina Smolic
- Department of Pharmacology and Biochemistry, Faculty of Dental Medicine and Health Osijek, J. J. Strossmayer University of Osijek, Osijek, Croatia.,Department of Pharmacology, Faculty of Medicine Osijek, J. J. Strossmayer University of Osijek, Osijek, Croatia
| | - Jurica Rubin
- Department of Medicine, Division of Gastroenterology/Hepatology, University Hospital Osijek, Osijek, Croatia
| | - Ashraf Tabll
- Microbial Biotechnology Department, Genetic Engineering and Biotechnology Research Division, National Research Centre, Dokki, Egypt.,Department of immunology, Egypt Center for Research and Regenerative
| | - Robert Smolic
- Department of Medicine, Division of Gastroenterology/Hepatology, University Hospital Osijek, Osijek, Croatia.,Department of Pathophysiology, Physiology and Immunology, Faculty of Dental Medicine and Health Osijek, J. J. Strossmayer University of Osijek, Osijek, Croatia.,Department of Pathophysiology, Faculty of Medicine Osijek, J. J. Strossmayer University of Osijek, Osijek, Croatia
| |
Collapse
|
29
|
Rupcic Rubin V, Bojanic K, Smolic M, Rubin J, Tabll A, Smolic R. An Update on Efficacy and Safety of Emerging Hepatic Antifibrotic Agents. J Clin Transl Hepatol 2021; 9:60-70. [PMID: 33604256 PMCID: PMC7868700 DOI: 10.14218/jcth.2020.00040] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/03/2020] [Revised: 11/22/2020] [Accepted: 12/09/2020] [Indexed: 02/07/2023] Open
Abstract
Liver fibrosis represents a response to chronic liver injury. Metabolic dysfunction-associated fatty liver disease and metabolic dysfunction-associated steatohepatitis are the most common chronic liver diseases, both with increasing incidence. Therefore, there is a great impetus for development of agents targeting these conditions. Accumulating data on possible treatment options for liver fibrosis are emerging in the literature. However, despite extensive research and much effort in the field, approved agents for liver fibrosis are still lacking. In this critical review, we have summarized the main data about specific treatment options for liver fibrosis gained from ongoing clinical trials, with an emphasis on efficacy and safety of these agents.
Collapse
Affiliation(s)
- Vinka Rupcic Rubin
- Department of Gynaecology and Obstetrics, Osijek University Hospital Centre, Faculty of Medicine, Josip Juraj Strossmayer University of Osijek, Osijek, Croatia
| | - Kristina Bojanic
- Department of Biophysics and Radiology, Faculty of Dental Medicine and Health Osijek, J. J. Strossmayer University of Osijek, Osijek, Croatia
- Department of Biophysics and Radiology, Faculty of Medicine Osijek, J. J. Strossmayer University of Osijek, Osijek, Croatia
- Department of Radiology, Health Center Osijek, Osijek, Croatia
| | - Martina Smolic
- Department of Pharmacology and Biochemistry, Faculty of Dental Medicine and Health Osijek, J. J. Strossmayer University of Osijek, Osijek, Croatia
- Department of Pharmacology, Faculty of Medicine Osijek, J. J. Strossmayer University of Osijek, Osijek, Croatia
| | - Jurica Rubin
- Department of Medicine, Division of Gastroenterology/Hepatology, University Hospital Osijek, Osijek, Croatia
| | - Ashraf Tabll
- Microbial Biotechnology Department, Genetic Engineering and Biotechnology Research Division, National Research Centre, Dokki, Egypt
- Department of immunology, Egypt Center for Research and Regenerative
| | - Robert Smolic
- Department of Medicine, Division of Gastroenterology/Hepatology, University Hospital Osijek, Osijek, Croatia
- Department of Pathophysiology, Physiology and Immunology, Faculty of Dental Medicine and Health Osijek, J. J. Strossmayer University of Osijek, Osijek, Croatia
- Department of Pathophysiology, Faculty of Medicine Osijek, J. J. Strossmayer University of Osijek, Osijek, Croatia
| |
Collapse
|
30
|
Abstract
Molecular magnetic resonance (MR) imaging utilizes molecular probes to provide added biochemical or cellular information to what can already be achieved with anatomical and functional MR imaging. This review provides an overview of molecular MR and focuses specifically on molecular MR contrast agents that provide contrast by shortening the T1 time. We describe the requirements for a successful molecular MR contrast agent and the challenges for clinical translation. The review highlights work from the last 5 years and places an emphasis on new contrast agents that have been validated in multiple preclinical models. Applications of molecular MR include imaging of inflammation, fibrosis, fibrogenesis, thromboembolic disease, and cancers. Molecular MR is positioned to move beyond detection of disease to the quantitative staging of disease and measurement of treatment response.
Collapse
Affiliation(s)
| | | | - Peter Caravan
- The Institute for Innovation in Imaging, A. A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Harvard Medical School, Charlestown, Massachusetts 02129, USA
| |
Collapse
|
31
|
Zhou IY, Montesi SB, Akam EA, Caravan P. Molecular Imaging of Fibrosis. Mol Imaging 2021. [DOI: 10.1016/b978-0-12-816386-3.00077-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
|
32
|
Molecular Magnetic Resonance Imaging of Fibrin Deposition in the Liver as an Indicator of Tissue Injury and Inflammation. Invest Radiol 2020; 55:209-216. [PMID: 31895219 DOI: 10.1097/rli.0000000000000631] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
RATIONALE AND OBJECTIVES Liver inflammation is associated with nonalcoholic steatohepatitis and other pathologies, but noninvasive methods to assess liver inflammation are limited. Inflammation causes endothelial disruption and leakage of plasma proteins into the interstitial space and can result in extravascular coagulation with fibrin deposition. Here we assess the feasibility of using the established fibrin-specific magnetic resonance probe EP-2104R for the noninvasive imaging of fibrin as a marker of liver inflammation. METHODS Weekly 100 mg/kg diethylnitrosamine (DEN) dosing was used to generate liver fibrosis in male rats; control animals received vehicle. Magnetic resonance imaging at 1.5 T with EP-2104R, a matched non-fibrin-binding control linear peptide, or the collagen-specific probe EP-3533 was performed at 1 day or 7 days after the last DEN administration. Imaging data were compared with quantitative histological measures of fibrosis and inflammation. RESULTS After 4 or 5 DEN administrations, the liver becomes moderately fibrotic, and fibrosis is the same if the animal is killed 1 day (Ishak score, 3.62 ± 0.31) or 7 days (Ishak score, 3.82 ± 0.25) after the last DEN dose, but inflammation is significantly higher at 1 day compared with 7 days after the last DEN dose (histological activity index from 0-4, 3.54 ± 0.14 vs 1.61 ± 0.16, respectively; P < 0.0001). Peak EP-2104R signal enhancement was significantly higher in animals imaged at 1 day post-DEN compared with 7 days post-DEN or control rats (29.0% ± 3.2% vs 22.4% ± 2.0% vs 17.0% ± 0.2%, respectively; P = 0.017). Signal enhancement with EP-2104R was significantly higher than control linear peptide at 1 day post-DEN but not at 7 days post-DEN indicating specific fibrin binding during the inflammatory phase. Collagen molecular magnetic resonance with EP-3533 showed equivalent T1 change when imaging rats 1 day or 7 days post-DEN, consistent with equivalent fibrosis. CONCLUSIONS EP-2104R can specifically detect fibrin associated with inflammation in a rat model of liver inflammation and fibrosis.
Collapse
|
33
|
Gochanour EM, Kowdley KV. Investigational drugs in early phase development for primary biliary cholangitis. Expert Opin Investig Drugs 2020; 30:131-141. [PMID: 33249947 DOI: 10.1080/13543784.2021.1857364] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Introduction: With a large percentage of patients having an incomplete response or intolerance to current FDA approved medications, new therapies for the treatment of primary biliary cholangitis are in great demand. Areas covered: In this review, we assess currently available drugs as well as promising new therapies for the treatment of primary biliary cholangitis. A literature search was performed with the following search terms: 'PBC treatment,' 'PBC therapeutics,' 'PBC clinical trials,' and included original articles, meta-analyses, and systematic reviews from 1 January 1981, to 1 January 2020. ClinicalTrials.gov was accessed for data from ongoing trials. Expert opinion: Targeted drug therapies offer an alternative for patients who are unable to meet their therapeutic goals with either of the two currently approved treatment options. Specifically, new drugs targeting bile-acid regulation, immune-modulation, and fibrogenic pathways are currently in development with multiple agents showing encouraging early results with the ultimate goal of developing therapies that will achieve high rates of biochemical remission, will be well tolerated, and improve symptoms and quality of life in patients with primary biliary cholangitis. Based on a review of the current literature, PPAR agonists appear to be promising agents, along with FGF19 analogs and FXR agonists.
Collapse
|
34
|
Zhou IY, Tanabe KK, Fuchs BC, Caravan P. Collagen-targeted molecular imaging in diffuse liver diseases. Abdom Radiol (NY) 2020; 45:3545-3556. [PMID: 32737546 DOI: 10.1007/s00261-020-02677-2] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2020] [Revised: 07/10/2020] [Accepted: 07/18/2020] [Indexed: 12/14/2022]
Abstract
Liver fibrosis is a common pathway shared by all progressive chronic liver diseases (CLD) regardless of the underlying etiologies. With liver biopsy being the gold standard in assessing fibrosis degree, there is a large unmet clinical need to develop non-invasive imaging tools that can directly and repeatedly quantify fibrosis throughout the liver for a more accurate assessment of disease burden, progression, and treatment response. Type I collagen is a particularly attractive target for molecular imaging as its excessive deposition is specific to fibrosis, and it is present in concentrations suitable for many imaging modalities. Novel molecular MRI contrast agents designed to bind with collagen provide direct quantification of collagen deposition, which have been validated across animal species and liver injury models. Collagen-targeted molecular imaging probes hold great promise not only as a tool for initial staging and surveillance of fibrosis progression, but also as a marker of fibrosis regression in drug trials.
Collapse
Affiliation(s)
- Iris Y Zhou
- Athinoula A. Martinos Center for Biomedical Imaging, Charlestown, MA, USA
- Harvard Medical School, 149 13th St, Boston, MA, 02129, USA
- Institute for Innovation in Imaging (i3), Department of Radiology, Massachusetts General Hospital, Charlestown, MA, USA
| | - Kenneth K Tanabe
- Division of Surgical Oncology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | | | - Peter Caravan
- Athinoula A. Martinos Center for Biomedical Imaging, Charlestown, MA, USA.
- Harvard Medical School, 149 13th St, Boston, MA, 02129, USA.
- Institute for Innovation in Imaging (i3), Department of Radiology, Massachusetts General Hospital, Charlestown, MA, USA.
| |
Collapse
|
35
|
Zhou IY, Catalano OA, Caravan P. Advances in functional and molecular MRI technologies in chronic liver diseases. J Hepatol 2020; 73:1241-1254. [PMID: 32585160 PMCID: PMC7572718 DOI: 10.1016/j.jhep.2020.06.020] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/25/2020] [Revised: 06/11/2020] [Accepted: 06/15/2020] [Indexed: 02/06/2023]
Abstract
MRI has emerged as the most comprehensive non-invasive diagnostic tool for liver diseases. In recent years, the value of MRI in hepatology has been significantly enhanced by a wide range of contrast agents, both clinically available and under development, that add functional information to anatomically detailed morphological images, or increase the distinction between normal and pathological tissues by targeting molecular and cellular events. Several classes of contrast agents are available for contrast-enhanced hepatic MRI, including i) conventional non-specific extracellular fluid contrast agents for assessing tissue perfusion; ii) hepatobiliary-specific contrast agents that are taken up by functioning hepatocytes and excreted through the biliary system for evaluating hepatobiliary function; iii) superparamagnetic iron oxide particles that accumulate in Kupffer cells; and iv) novel molecular contrast agents that are biochemically targeted to specific molecular/cellular processes for staging liver diseases or detecting treatment responses. The use of different functional and molecular MRI methods enables the non-invasive assessment of disease burden, progression, and treatment response in a variety of liver diseases. A high diagnostic performance can be achieved with MRI by combining imaging biomarkers.
Collapse
Affiliation(s)
- Iris Y Zhou
- Athinoula A. Martinos Center for Biomedical Imaging, Charlestown, MA, United States; Harvard Medical School, Boston, MA, USA; Institute for Innovation in Imaging (i(3)), Department of Radiology, Massachusetts General Hospital, Charlestown, MA, USA
| | - Onofrio A Catalano
- Athinoula A. Martinos Center for Biomedical Imaging, Charlestown, MA, United States; Harvard Medical School, Boston, MA, USA; Division of Abdominal Imaging, Department of Radiology, Massachusetts General Hospital, Boston, MA, United States
| | - Peter Caravan
- Athinoula A. Martinos Center for Biomedical Imaging, Charlestown, MA, United States; Harvard Medical School, Boston, MA, USA; Institute for Innovation in Imaging (i(3)), Department of Radiology, Massachusetts General Hospital, Charlestown, MA, USA.
| |
Collapse
|
36
|
Abstract
The worldwide prevalence of non-alcoholic fatty liver disease is around 25%, and that of nonalcoholic steatohepatitis (NASH) ranges from 1.5% to 6.45%. Patients with NASH, especially those with fibrosis, are at higher risk for adverse outcomes such as cirrhosis and liver-related mortality. Although vitamin E, pioglitazone, and liraglutide improved liver histology in randomized trials, there are currently no Food and Drug Administration-approved drugs for NASH. Five pharmacologic agents-obeticholic acid, elafibranor, cenicriviroc, resmetirom, and aramchol-are being evaluated in large, histology-based phase 3 trials. Within 2 to 4 years, new and effective drugs for the treatment of NASH are expected. Additionally, many phase 2 trials are ongoing for various agents. Based on the results of phase 2 and 3 trials, combination treatments are also being investigated. Future treatment strategies will comprise drug combinations and precision medicine based on the different phenotypes of NASH and treatment response of the individual patient.
Collapse
Affiliation(s)
- Soung Won Jeong
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Soonchunhyang University Seoul Hospital, Soonchunhyang University College of Medicine, Seoul, Korea
- Corresponding author: Soung Won Jeong Division of Gastroenterology and Hepatology, Department of Internal Medicine, Soonchunhyang University Seoul Hospital, Soonchunhyang University College of Medicine, 59 Daesagwan-ro, Yongsan-gu, Seoul 04401, Korea E-mail:
| |
Collapse
|
37
|
Li C, Yang J, Wang Y, Qi Y, Yang W, Li Y. Farnesoid X Receptor Agonists as Therapeutic Target for Cardiometabolic Diseases. Front Pharmacol 2020; 11:1247. [PMID: 32982723 PMCID: PMC7479173 DOI: 10.3389/fphar.2020.01247] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2020] [Accepted: 07/29/2020] [Indexed: 12/12/2022] Open
Abstract
Cardiometabolic diseases are characterized as a combination of multiple risk factors for cardiovascular disease (CVD) and metabolic diseases including diabetes mellitus and dyslipidemia. Cardiometabolic diseases are closely associated with cell glucose and lipid metabolism, inflammatory response and mitochondrial function. Farnesoid X Receptor (FXR), a metabolic nuclear receptor, are found to be activated by primary BAs such as chenodeoxycholic acid (CDCA), cholic acid (CA) and synthetic agonists such as obeticholic acid (OCA). FXR plays crucial roles in regulating cholesterol homeostasis, lipid metabolism, glucose metabolism, and intestinal microorganism. Recently, emerging evidence suggests that FXR agonists are functional for metabolic syndrome and cardiovascular diseases and are considered as a potential therapeutic agent. This review will discuss the pathological mechanism of cardiometabolic disease and reviews the potential mechanisms of FXR agonists in the treatment of cardiometabolic disease.
Collapse
Affiliation(s)
- Chao Li
- Experimental Center, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Jie Yang
- Cardiovascular Department, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Yu Wang
- Cardiovascular Department, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Yingzi Qi
- School of Health, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Wenqing Yang
- Experimental Center, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Yunlun Li
- Experimental Center, Shandong University of Traditional Chinese Medicine, Jinan, China.,Cardiovascular Department, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
| |
Collapse
|
38
|
Perino A, Demagny H, Velazquez-Villegas L, Schoonjans K. Molecular Physiology of Bile Acid Signaling in Health, Disease, and Aging. Physiol Rev 2020; 101:683-731. [PMID: 32790577 DOI: 10.1152/physrev.00049.2019] [Citation(s) in RCA: 237] [Impact Index Per Article: 47.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Over the past two decades, bile acids (BAs) have become established as important signaling molecules that enable fine-tuned inter-tissue communication from the liver, their site of production, over the intestine, where they are modified by the gut microbiota, to virtually any organ, where they exert their pleiotropic physiological effects. The chemical variety of BAs, to a large extent determined by the gut microbiome, also allows for a complex fine-tuning of adaptive responses in our body. This review provides an overview of the mechanisms by which BA receptors coordinate several aspects of physiology and highlights new therapeutic strategies for diseases underlying pathological BA signaling.
Collapse
Affiliation(s)
- Alessia Perino
- Institute of Bioengineering, Ecole Polytechnique Fédérale de Lausanne, Lausanne (EPFL), Switzerland
| | - Hadrien Demagny
- Institute of Bioengineering, Ecole Polytechnique Fédérale de Lausanne, Lausanne (EPFL), Switzerland
| | - Laura Velazquez-Villegas
- Institute of Bioengineering, Ecole Polytechnique Fédérale de Lausanne, Lausanne (EPFL), Switzerland
| | - Kristina Schoonjans
- Institute of Bioengineering, Ecole Polytechnique Fédérale de Lausanne, Lausanne (EPFL), Switzerland
| |
Collapse
|
39
|
Zhou IY, Clavijo Jordan V, Rotile NJ, Akam E, Krishnan S, Arora G, Krishnan H, Slattery H, Warner N, Mercaldo N, Farrar CT, Wellen J, Martinez R, Schlerman F, Tanabe KK, Fuchs BC, Caravan P. Advanced MRI of Liver Fibrosis and Treatment Response in a Rat Model of Nonalcoholic Steatohepatitis. Radiology 2020; 296:67-75. [PMID: 32343209 DOI: 10.1148/radiol.2020192118] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Background Liver biopsy is the reference standard to diagnose nonalcoholic steatohepatitis (NASH) but is invasive with potential complications. Purpose To evaluate molecular MRI with type 1 collagen-specific probe EP-3533 and allysine-targeted fibrogenesis probe Gd-Hyd, MR elastography, and native T1 to characterize fibrosis and to assess treatment response in a rat model of NASH. Materials and Methods MRI was performed prospectively (June-November 2018) in six groups of male Wistar rats (a) age- and (b) weight-matched animals received standard chow (n = 12 per group); (c) received choline-deficient, l-amino acid-defined, high-fat diet (CDAHFD) for 6 weeks or (d) 9 weeks (n = 8 per group); (e) were fed 6 weeks of CDAHFD and switched to standard chow for 3 weeks (n = 12); (f) were fed CDAHFD for 9 weeks with daily treatment of elafibranor beginning at week 6 (n = 14). Differences in imaging measurements and tissue analyses among groups were tested with one-way analysis of variance. The ability of each imaging measurement to stage fibrosis was quantified by using area under the receiver operating characteristic curve (AUC) with quantitative digital pathology (collagen proportionate area [CPA]) as reference standard. Optimal cutoff values for distinguishing advanced fibrosis were used to assess treatment response. Results AUC for distinguishing fibrotic (CPA >4.8%) from nonfibrotic (CPA ≤4.8%) livers was 0.95 (95% confidence interval [CI]: 0.91, 1.00) for EP-3533, followed by native T1, Gd-Hyd, and MR elastography with AUCs of 0.90 (95% CI: 0.83, 0.98), 0.84 (95% CI: 0.74, 0.95), and 0.65 (95% CI: 0.51, 0.79), respectively. AUCs for discriminating advanced fibrosis (CPA >10.3%) were 0.86 (95% CI: 0.76, 0.97), 0.96 (95% CI: 0.90, 1.01), 0.84 (95% CI: 0.70, 0.98), and 0.74 (95% CI: 0.63, 0.86) for EP-3533, Gd-Hyd, MR elastography, and native T1, respectively. Gd-Hyd MRI had the highest accuracy (24 of 26, 92%; 95% CI: 75%, 99%) in identifying responders and nonresponders in the treated groups compared with MR elastography (23 of 26, 88%; 95% CI: 70%, 98%), EP-3533 (20 of 26, 77%; 95% CI: 56%, 91%), and native T1 (14 of 26, 54%; 95% CI: 33%, 73%). Conclusion Collagen-targeted molecular MRI most accurately detected early onset of fibrosis, whereas the fibrogenesis probe Gd-Hyd proved most accurate for detecting treatment response. © RSNA, 2020 Online supplemental material is available for this article.
Collapse
Affiliation(s)
- Iris Y Zhou
- From the Department of Radiology, Athinoula A. Martinos Center for Biomedical Imaging, Institute for Innovation in Imaging (I.Y.Z., V.C.J., N.J.R., E.A., H.K., H.S., N.W., C.T.F., P.C.), Division of Surgical Oncology (S.K., G.A., K.K.T., B.C.F.), and Institute for Technology Assessment, Department of Radiology (N.M.), Massachusetts General Hospital and Harvard Medical School, Charlestown, 149 13th St, Boston, MA 02129; and Pfizer, Cambridge, Mass (J.W., R.M., F.S.)
| | - Veronica Clavijo Jordan
- From the Department of Radiology, Athinoula A. Martinos Center for Biomedical Imaging, Institute for Innovation in Imaging (I.Y.Z., V.C.J., N.J.R., E.A., H.K., H.S., N.W., C.T.F., P.C.), Division of Surgical Oncology (S.K., G.A., K.K.T., B.C.F.), and Institute for Technology Assessment, Department of Radiology (N.M.), Massachusetts General Hospital and Harvard Medical School, Charlestown, 149 13th St, Boston, MA 02129; and Pfizer, Cambridge, Mass (J.W., R.M., F.S.)
| | - Nicholas J Rotile
- From the Department of Radiology, Athinoula A. Martinos Center for Biomedical Imaging, Institute for Innovation in Imaging (I.Y.Z., V.C.J., N.J.R., E.A., H.K., H.S., N.W., C.T.F., P.C.), Division of Surgical Oncology (S.K., G.A., K.K.T., B.C.F.), and Institute for Technology Assessment, Department of Radiology (N.M.), Massachusetts General Hospital and Harvard Medical School, Charlestown, 149 13th St, Boston, MA 02129; and Pfizer, Cambridge, Mass (J.W., R.M., F.S.)
| | - Eman Akam
- From the Department of Radiology, Athinoula A. Martinos Center for Biomedical Imaging, Institute for Innovation in Imaging (I.Y.Z., V.C.J., N.J.R., E.A., H.K., H.S., N.W., C.T.F., P.C.), Division of Surgical Oncology (S.K., G.A., K.K.T., B.C.F.), and Institute for Technology Assessment, Department of Radiology (N.M.), Massachusetts General Hospital and Harvard Medical School, Charlestown, 149 13th St, Boston, MA 02129; and Pfizer, Cambridge, Mass (J.W., R.M., F.S.)
| | - Smitha Krishnan
- From the Department of Radiology, Athinoula A. Martinos Center for Biomedical Imaging, Institute for Innovation in Imaging (I.Y.Z., V.C.J., N.J.R., E.A., H.K., H.S., N.W., C.T.F., P.C.), Division of Surgical Oncology (S.K., G.A., K.K.T., B.C.F.), and Institute for Technology Assessment, Department of Radiology (N.M.), Massachusetts General Hospital and Harvard Medical School, Charlestown, 149 13th St, Boston, MA 02129; and Pfizer, Cambridge, Mass (J.W., R.M., F.S.)
| | - Gunisha Arora
- From the Department of Radiology, Athinoula A. Martinos Center for Biomedical Imaging, Institute for Innovation in Imaging (I.Y.Z., V.C.J., N.J.R., E.A., H.K., H.S., N.W., C.T.F., P.C.), Division of Surgical Oncology (S.K., G.A., K.K.T., B.C.F.), and Institute for Technology Assessment, Department of Radiology (N.M.), Massachusetts General Hospital and Harvard Medical School, Charlestown, 149 13th St, Boston, MA 02129; and Pfizer, Cambridge, Mass (J.W., R.M., F.S.)
| | - Hema Krishnan
- From the Department of Radiology, Athinoula A. Martinos Center for Biomedical Imaging, Institute for Innovation in Imaging (I.Y.Z., V.C.J., N.J.R., E.A., H.K., H.S., N.W., C.T.F., P.C.), Division of Surgical Oncology (S.K., G.A., K.K.T., B.C.F.), and Institute for Technology Assessment, Department of Radiology (N.M.), Massachusetts General Hospital and Harvard Medical School, Charlestown, 149 13th St, Boston, MA 02129; and Pfizer, Cambridge, Mass (J.W., R.M., F.S.)
| | - Hannah Slattery
- From the Department of Radiology, Athinoula A. Martinos Center for Biomedical Imaging, Institute for Innovation in Imaging (I.Y.Z., V.C.J., N.J.R., E.A., H.K., H.S., N.W., C.T.F., P.C.), Division of Surgical Oncology (S.K., G.A., K.K.T., B.C.F.), and Institute for Technology Assessment, Department of Radiology (N.M.), Massachusetts General Hospital and Harvard Medical School, Charlestown, 149 13th St, Boston, MA 02129; and Pfizer, Cambridge, Mass (J.W., R.M., F.S.)
| | - Noah Warner
- From the Department of Radiology, Athinoula A. Martinos Center for Biomedical Imaging, Institute for Innovation in Imaging (I.Y.Z., V.C.J., N.J.R., E.A., H.K., H.S., N.W., C.T.F., P.C.), Division of Surgical Oncology (S.K., G.A., K.K.T., B.C.F.), and Institute for Technology Assessment, Department of Radiology (N.M.), Massachusetts General Hospital and Harvard Medical School, Charlestown, 149 13th St, Boston, MA 02129; and Pfizer, Cambridge, Mass (J.W., R.M., F.S.)
| | - Nathaniel Mercaldo
- From the Department of Radiology, Athinoula A. Martinos Center for Biomedical Imaging, Institute for Innovation in Imaging (I.Y.Z., V.C.J., N.J.R., E.A., H.K., H.S., N.W., C.T.F., P.C.), Division of Surgical Oncology (S.K., G.A., K.K.T., B.C.F.), and Institute for Technology Assessment, Department of Radiology (N.M.), Massachusetts General Hospital and Harvard Medical School, Charlestown, 149 13th St, Boston, MA 02129; and Pfizer, Cambridge, Mass (J.W., R.M., F.S.)
| | - Christian T Farrar
- From the Department of Radiology, Athinoula A. Martinos Center for Biomedical Imaging, Institute for Innovation in Imaging (I.Y.Z., V.C.J., N.J.R., E.A., H.K., H.S., N.W., C.T.F., P.C.), Division of Surgical Oncology (S.K., G.A., K.K.T., B.C.F.), and Institute for Technology Assessment, Department of Radiology (N.M.), Massachusetts General Hospital and Harvard Medical School, Charlestown, 149 13th St, Boston, MA 02129; and Pfizer, Cambridge, Mass (J.W., R.M., F.S.)
| | - Jeremy Wellen
- From the Department of Radiology, Athinoula A. Martinos Center for Biomedical Imaging, Institute for Innovation in Imaging (I.Y.Z., V.C.J., N.J.R., E.A., H.K., H.S., N.W., C.T.F., P.C.), Division of Surgical Oncology (S.K., G.A., K.K.T., B.C.F.), and Institute for Technology Assessment, Department of Radiology (N.M.), Massachusetts General Hospital and Harvard Medical School, Charlestown, 149 13th St, Boston, MA 02129; and Pfizer, Cambridge, Mass (J.W., R.M., F.S.)
| | - Robert Martinez
- From the Department of Radiology, Athinoula A. Martinos Center for Biomedical Imaging, Institute for Innovation in Imaging (I.Y.Z., V.C.J., N.J.R., E.A., H.K., H.S., N.W., C.T.F., P.C.), Division of Surgical Oncology (S.K., G.A., K.K.T., B.C.F.), and Institute for Technology Assessment, Department of Radiology (N.M.), Massachusetts General Hospital and Harvard Medical School, Charlestown, 149 13th St, Boston, MA 02129; and Pfizer, Cambridge, Mass (J.W., R.M., F.S.)
| | - Franklin Schlerman
- From the Department of Radiology, Athinoula A. Martinos Center for Biomedical Imaging, Institute for Innovation in Imaging (I.Y.Z., V.C.J., N.J.R., E.A., H.K., H.S., N.W., C.T.F., P.C.), Division of Surgical Oncology (S.K., G.A., K.K.T., B.C.F.), and Institute for Technology Assessment, Department of Radiology (N.M.), Massachusetts General Hospital and Harvard Medical School, Charlestown, 149 13th St, Boston, MA 02129; and Pfizer, Cambridge, Mass (J.W., R.M., F.S.)
| | - Kenneth K Tanabe
- From the Department of Radiology, Athinoula A. Martinos Center for Biomedical Imaging, Institute for Innovation in Imaging (I.Y.Z., V.C.J., N.J.R., E.A., H.K., H.S., N.W., C.T.F., P.C.), Division of Surgical Oncology (S.K., G.A., K.K.T., B.C.F.), and Institute for Technology Assessment, Department of Radiology (N.M.), Massachusetts General Hospital and Harvard Medical School, Charlestown, 149 13th St, Boston, MA 02129; and Pfizer, Cambridge, Mass (J.W., R.M., F.S.)
| | - Bryan C Fuchs
- From the Department of Radiology, Athinoula A. Martinos Center for Biomedical Imaging, Institute for Innovation in Imaging (I.Y.Z., V.C.J., N.J.R., E.A., H.K., H.S., N.W., C.T.F., P.C.), Division of Surgical Oncology (S.K., G.A., K.K.T., B.C.F.), and Institute for Technology Assessment, Department of Radiology (N.M.), Massachusetts General Hospital and Harvard Medical School, Charlestown, 149 13th St, Boston, MA 02129; and Pfizer, Cambridge, Mass (J.W., R.M., F.S.)
| | - Peter Caravan
- From the Department of Radiology, Athinoula A. Martinos Center for Biomedical Imaging, Institute for Innovation in Imaging (I.Y.Z., V.C.J., N.J.R., E.A., H.K., H.S., N.W., C.T.F., P.C.), Division of Surgical Oncology (S.K., G.A., K.K.T., B.C.F.), and Institute for Technology Assessment, Department of Radiology (N.M.), Massachusetts General Hospital and Harvard Medical School, Charlestown, 149 13th St, Boston, MA 02129; and Pfizer, Cambridge, Mass (J.W., R.M., F.S.)
| |
Collapse
|
40
|
Sumida Y, Yoneda M, Ogawa Y, Yoneda M, Okanoue T, Nakajima A. Current and new pharmacotherapy options for non-alcoholic steatohepatitis. Expert Opin Pharmacother 2020; 21:953-967. [PMID: 32237916 DOI: 10.1080/14656566.2020.1744564] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
INTRODUCTION There is an unmet medical need for an effective anti-fibrotic treatment for NASH with advanced fibrosis. AREAS COVERED The authors review the current and novel agents for the treatment of NASH with fibrosis. They also consider the potential future strategies of combination therapies. EXPERT OPINION Farnesoid X receptor (FXR) agonist (obeticholic acid [OCA]) significantly ameliorated hepatic fibrosis in NASH stage 2/3 fibrosis in an interim analysis of phase 3 trial. Because OCA has several drawbacks such as itching and elevated low-density lipoprotein-cholesterol (LDL-C), non-bile acid FXR agonists are now under development. Selonsertib (apoptosis signaling kinase 1 inhibitor), emricasan (an irreversible pan-caspase inhibitor), and simtsuzumab (a monoclonal antibody against lysyl oxidase-like 2) were discontinued because of no efficacy over placebo. Peroxisome proliferator-activator receptor α/δ agonists, C-C motif chemokine receptor-2/5 antagonists, and thyroid β receptor agonist are ongoing in phase 3 trials. A variety of agents including fibroblast growth factor (FGF)-21 and FGF-19 agonists, as well as acetyl-CoA carboxylase inhibitors, are also expected. Among antidiabetic agents, semaglutide, a novel GLP-1 RA, is ongoing for NASH stage 1-3 fibrosis in a phase 2 trial. Furthermore, the combination of GLP-RA/glucagon receptor agonist and GLP-RA/gastrointestinal peptide agonist are promising future options.
Collapse
Affiliation(s)
- Yoshio Sumida
- Division of Hepatology and Pancreatology, Department of Internal Medicine, Aichi Medical University , Kasugai, Japan
| | - Masato Yoneda
- Department of Gastroenterology and Hepatology, Yokohama City University , Yokohama, Japan
| | - Yuji Ogawa
- Department of Gastroenterology and Hepatology, Yokohama City University , Yokohama, Japan
| | - Masashi Yoneda
- Division of Hepatology and Pancreatology, Department of Internal Medicine, Aichi Medical University , Kasugai, Japan
| | - Takeshi Okanoue
- Department of Gastroenterology, Suita Hospital , Osaka, Japan
| | - Atsushi Nakajima
- Department of Gastroenterology and Hepatology, Yokohama City University , Yokohama, Japan
| |
Collapse
|
41
|
Yao Q, Li S, Li X, Wang F, Tu C. Myricetin Modulates Macrophage Polarization and Mitigates Liver Inflammation and Fibrosis in a Murine Model of Nonalcoholic Steatohepatitis. Front Med (Lausanne) 2020; 7:71. [PMID: 32195263 PMCID: PMC7065264 DOI: 10.3389/fmed.2020.00071] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2019] [Accepted: 02/19/2020] [Indexed: 12/12/2022] Open
Abstract
This study aimed to investigate the beneficial effects of myricetin in a diet-induced nonalcoholic steatohepatitis (NASH) model and the underlying mechanism. C57BL/6J mice were fed a standard chow or the choline-deficient, L-amino acid-defined, high-fat diet (CDAHFD) for 8 weeks with the treatment of myricetin (100 mg/kg) or vehicle by daily gavage. Hepatic inflammation, steatosis, fibrosis, and hepatic stellate cells (HSC) activation were assessed. We also analyzed M1 and M2 macrophages and its related markers in livers from NASH mice and in RAW264.7 macrophages stimulated by lipopolysaccharide (LPS) or interleukin 4 (IL-4) in vitro. Furthermore, we determined the effect of myricetin on the triggering receptor expressed on myeloid cells-1 (TREM-1), toll like receptor (TLR) 2 and 4, and myeloid differentiation factor 88 (MyD88) signaling both in livers from mice and in RAW264.7 cells stimulated by LPS. Our results revealed that myricetin remarkably ameliorated hepatic steatosis, inflammation, and inhibited hepatic macrophage infiltration in CDAHFD-fed mice. Myricetin-treated to CDAHFD-fed mice also inhibited liver fibrosis and HSC activation when compared with vehicle-treated to those mice. Moreover, myricetin inhibited M1 macrophage polarization and its relative markers in livers of NASH mice while induced M2 polarization. Similarly, in vitro study, myricetin inhibited the LPS-induced mRNA expression of M1 macrophages marker genes and induced IL-4-induced M2 macrophage marker genes in RAW264.7 macrophages. Mechanically, myricetin inhibited the expression of TREM-1 and TLR2/4-MyD88 signaling molecules in livers from NASH mice and in RAW264.7 macrophages stimulated by LPS in vitro. Additionally, myricetin inhibited the activation of nuclear factor (NF)-κB signaling and the phosphorylation of the signal transducer and activation of transcription 3 (STAT3) in LPS-stimulated RAW264.7 macrophages. Taken together, our data indicated that myricetin modulated the polarization of macrophages via inhibiting the TREM-1-TLR2/4-MyD88 signaling molecules in macrophages and therefore mitigated NASH and hepatic fibrosis in the CDAHFD-diet-induced NASH model in mice.
Collapse
Affiliation(s)
- Qunyan Yao
- Department of Gastroenterology and Hepatology, Zhongshan Hospital, Fudan University, Shanghai, China
- Shanghai Institute of Liver Diseases, Shanghai, China
| | - Shuyu Li
- Department of Gastroenterology and Hepatology, Zhongshan Hospital, Fudan University, Shanghai, China
- Shanghai Institute of Liver Diseases, Shanghai, China
| | - Xi Li
- Department of Geriatrics, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Fu Wang
- Shanghai Medical College, Fudan University, Shanghai, China
| | - Chuantao Tu
- Department of Gastroenterology, Shanghai Public Health Clinical Center, Fudan University, Shanghai, China
- *Correspondence: Chuantao Tu
| |
Collapse
|
42
|
Shah RA, Kowdley KV. Current and potential treatments for primary biliary cholangitis. Lancet Gastroenterol Hepatol 2020; 5:306-315. [DOI: 10.1016/s2468-1253(19)30343-7] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/31/2019] [Revised: 07/31/2019] [Accepted: 08/01/2019] [Indexed: 02/07/2023]
|
43
|
Abstract
Though ursodeoxycholic acid (UDCA) remains the baseline treatment for most cholestatic liver diseases, UDCA treatment leaves approximately one-third of patients with primary biliary cholangitis (PBC) and all patients with primary sclerosing cholangitis (PSC) at risk for disease progression. New anticholestatic agents, including nuclear receptor agonists, choleretics, and bile acid synthesis suppressors, will likely increase response rates to therapy in PBC and PSC. Strategies that target early immune-mediated injury have so far been disappointing, hampered by the lack of biomarkers to detect early disease states, which then could profit from immunomodulatory therapy. Future concepts need to personalize treatments according to disease stage, progression, and phase, and to combine multiple drugs to target different pathogenic pathways.
Collapse
Affiliation(s)
- Martin Wagner
- Division of Gastroenterology and Hepatology, Department of Medicine, Medical University of Graz, 8036 Graz, Austria
| | - Peter Fickert
- Division of Gastroenterology and Hepatology, Department of Medicine, Medical University of Graz, 8036 Graz, Austria
| |
Collapse
|
44
|
Roth JD, Veidal SS, Fensholdt LKD, Rigbolt KTG, Papazyan R, Nielsen JC, Feigh M, Vrang N, Young M, Jelsing J, Adorini L, Hansen HH. Combined obeticholic acid and elafibranor treatment promotes additive liver histological improvements in a diet-induced ob/ob mouse model of biopsy-confirmed NASH. Sci Rep 2019; 9:9046. [PMID: 31227742 PMCID: PMC6588626 DOI: 10.1038/s41598-019-45178-z] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2019] [Accepted: 05/29/2019] [Indexed: 12/13/2022] Open
Abstract
Obeticholic acid (OCA) and elafibranor (ELA) are selective and potent agonists for the farnesoid X receptor (FXR) and dual peroxisome proliferator-activated receptor α/δ (PPAR-α/δ), respectively. Both agents have demonstrated clinical efficacy in nonalcoholic steatohepatitis (NASH). The present study used OCA and ELA to compare the effects of mono- and combination therapies on metabolic and histological endpoints in Lepob/ob mice with established diet-induced and biopsy-confirmed NASH (ob/ob-NASH). ob/ob-NASH mice were fed the AMLN diet high in trans-fat, fructose and cholesterol for 15 weeks, whereafter they received vehicle, OCA (30 mg/kg, PO, QD), ELA (3, 10 mg/kg, PO, QD), or combinations (OCA + ELA) for eight weeks. Within-subject comparisons were performed on histomorphometric changes, including fractional area of liver fat, galectin-3 and Col1a1. OCA and ELA monotherapies improved all quantitative histopathological parameters and OCA + ELA combinations exerted additive effects on metabolic and histological endpoints. In agreement with their different molecular mechanisms of action, OCA and ELA monotherapies elicited distinct hepatic gene expression profiles and their combination led to profound transcriptome changes associated with further improvements in lipid handling and insulin signaling, suppression of immune responses and reduced extracellular matrix formation. In conclusion, these findings provide preclinical proof-of-concept for combined FXR and PPAR-α/δ agonist-based therapies in NASH.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Mark Young
- Intercept Pharmaceuticals, San Diego, CA, USA
| | | | | | | |
Collapse
|
45
|
Montesi SB, Désogère P, Fuchs BC, Caravan P. Molecular imaging of fibrosis: recent advances and future directions. J Clin Invest 2019; 129:24-33. [PMID: 30601139 DOI: 10.1172/jci122132] [Citation(s) in RCA: 95] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Fibrosis, the progressive accumulation of connective tissue that occurs in response to injury, causes irreparable organ damage and may result in organ failure. The few available antifibrotic treatments modify the rate of fibrosis progression, but there are no available treatments to reverse established fibrosis. Thus, more effective therapies are urgently needed. Molecular imaging is a promising biomedical methodology that enables noninvasive visualization of cellular and subcellular processes. It provides a unique means to monitor and quantify dysregulated molecular fibrotic pathways in a noninvasive manner. Molecular imaging could be used for early detection, disease staging, and prognostication, as well as for assessing disease activity and treatment response. As fibrotic diseases are often molecularly heterogeneous, molecular imaging of a specific pathway could be used for patient stratification and cohort enrichment with the goal of improving clinical trial design and feasibility and increasing the ability to detect a definitive outcome for new therapies. Here we review currently available molecular imaging probes for detecting fibrosis and fibrogenesis, the active formation of new fibrous tissue, and their application to models of fibrosis across organ systems and fibrotic processes. We provide our opinion as to the potential roles of molecular imaging in human fibrotic diseases.
Collapse
Affiliation(s)
| | - Pauline Désogère
- Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA.,Athinoula A. Martinos Center for Biomedical Imaging and.,Institute for Innovation in Imaging, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Bryan C Fuchs
- Division of Surgical Oncology, Massachusetts General Hospital Cancer Center and Harvard Medical School, Boston, Massachusetts, USA
| | - Peter Caravan
- Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA.,Athinoula A. Martinos Center for Biomedical Imaging and.,Institute for Innovation in Imaging, Massachusetts General Hospital, Boston, Massachusetts, USA
| |
Collapse
|