1
|
Cazac-Panaite GD, Lăcătușu CM, Grigorescu ED, Foșălău AB, Onofriescu A, Mihai BM. Innovative Drugs First Implemented in Type 2 Diabetes Mellitus and Obesity and Their Effects on Metabolic Dysfunction-Associated Steatohepatitis (MASH)-Related Fibrosis and Cirrhosis. J Clin Med 2025; 14:1042. [PMID: 40004572 DOI: 10.3390/jcm14041042] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 01/06/2025] [Revised: 01/30/2025] [Accepted: 02/04/2025] [Indexed: 02/27/2025] Open
Abstract
Metabolic dysfunction-associated steatotic liver disease (MASLD), a progressive liver disease frequently associated with metabolic disorders such as type 2 diabetes mellitus (T2DM) and obesity, has the potential to progress symptomatically to liver cirrhosis and, in some cases, hepatocellular carcinoma. Hence, an urgent need arises to identify and approve new therapeutic options to improve patient outcomes. Research efforts have focused on either developing dedicated molecules or repurposing drugs already approved for other conditions, such as metabolic diseases. Among the latter, antidiabetic and anti-obesity agents have received the most extensive attention, with pivotal trial results anticipated shortly. However, the primary focus underlying successful regulatory approvals is demonstrating a substantial efficacy in improving liver fibrosis and preventing or ameliorating cirrhosis, the key advanced outcomes within MASLD progression. Besides liver steatosis, the ideal therapeutic candidate should reduce inflammation and fibrosis effectively. Although some agents have shown promise in lowering MASLD-related parameters, evidence of their impact on fibrosis and cirrhosis remains limited. This review aims to evaluate whether antidiabetic and anti-obesity drugs can be safely and effectively used in MASLD-related advanced fibrosis or cirrhosis in patients with T2DM. Our paper discusses the molecules closest to regulatory approval and the expectation that they can address the unmet needs of this increasingly prevalent disease.
Collapse
Affiliation(s)
- Georgiana-Diana Cazac-Panaite
- Unit of Diabetes, Nutrition, and Metabolic Diseases, Faculty of Medicine, "Grigore T. Popa" University of Medicine and Pharmacy, 700115 Iași, Romania
- Clinical Center of Diabetes, Nutrition and Metabolic Diseases, "Sf. Spiridon" County Clinical Emergency Hospital, 700111 Iași, Romania
| | - Cristina-Mihaela Lăcătușu
- Unit of Diabetes, Nutrition, and Metabolic Diseases, Faculty of Medicine, "Grigore T. Popa" University of Medicine and Pharmacy, 700115 Iași, Romania
- Clinical Center of Diabetes, Nutrition and Metabolic Diseases, "Sf. Spiridon" County Clinical Emergency Hospital, 700111 Iași, Romania
| | - Elena-Daniela Grigorescu
- Unit of Diabetes, Nutrition, and Metabolic Diseases, Faculty of Medicine, "Grigore T. Popa" University of Medicine and Pharmacy, 700115 Iași, Romania
- Clinical Center of Diabetes, Nutrition and Metabolic Diseases, "Sf. Spiridon" County Clinical Emergency Hospital, 700111 Iași, Romania
| | - Adina-Bianca Foșălău
- Unit of Diabetes, Nutrition, and Metabolic Diseases, Faculty of Medicine, "Grigore T. Popa" University of Medicine and Pharmacy, 700115 Iași, Romania
- Clinical Center of Diabetes, Nutrition and Metabolic Diseases, "Sf. Spiridon" County Clinical Emergency Hospital, 700111 Iași, Romania
| | - Alina Onofriescu
- Unit of Diabetes, Nutrition, and Metabolic Diseases, Faculty of Medicine, "Grigore T. Popa" University of Medicine and Pharmacy, 700115 Iași, Romania
- Clinical Center of Diabetes, Nutrition and Metabolic Diseases, "Sf. Spiridon" County Clinical Emergency Hospital, 700111 Iași, Romania
| | - Bogdan-Mircea Mihai
- Unit of Diabetes, Nutrition, and Metabolic Diseases, Faculty of Medicine, "Grigore T. Popa" University of Medicine and Pharmacy, 700115 Iași, Romania
- Clinical Center of Diabetes, Nutrition and Metabolic Diseases, "Sf. Spiridon" County Clinical Emergency Hospital, 700111 Iași, Romania
| |
Collapse
|
2
|
Brennan PN, MacMillan M, Manship T, Moroni F, Glover A, Troland D, MacPherson I, Graham C, Aird R, Semple SIK, Morris DM, Fraser AR, Pass C, McGowan NWA, Turner ML, Manson L, Lachlan NJ, Dillon JF, Kilpatrick AM, Campbell JDM, Fallowfield JA, Forbes SJ. Autologous macrophage therapy for liver cirrhosis: a phase 2 open-label randomized controlled trial. Nat Med 2025:10.1038/s41591-024-03406-8. [PMID: 39794616 DOI: 10.1038/s41591-024-03406-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 06/05/2024] [Accepted: 11/11/2024] [Indexed: 01/13/2025]
Abstract
Cirrhosis is a major cause of morbidity and mortality; however, there are no approved therapies except orthotopic liver transplantation. Preclinical studies showed that bone-marrow-derived macrophage injections reduce inflammation, resolve fibrosis and stimulate liver regeneration. In a multicenter, open-label, parallel-group, phase 2 randomized controlled trial ( ISRCTN10368050 ) in n = 51 adult patients with compensated cirrhosis and Model for End-Stage Liver Disease (MELD) score ≥10 and ≤17, we evaluated the efficacy of autologous monocyte-derived macrophage therapy (n = 27) compared to standard medical care (n = 24). The primary endpoint was the difference in baseline to day 90 change in MELD score (ΔMELD) between treatment and control groups (ΔΔMELD). Secondary endpoints included adverse clinical outcomes, non-invasive fibrosis biomarkers and health-related quality of life (HRQoL) at 90 d, 180 d and 360 d. The ΔΔMELD between day 0 and day 90 in the treatment group compared to controls was -0.87 (95% confidence interval: -1.79, 0.0; P = 0.06); therefore, the primary endpoint was not met. During 360-d follow-up, five of 24 participants in the control group developed a total of 10 severe adverse events, four of which were liver related, and three deaths (two liver related), whereas no liver-related severe adverse events or deaths occurred in the treatment group. Although no differences were observed in biomarkers or HRQoL, exploratory analysis showed anti-inflammatory serum cytokine profiles after macrophage infusion. This study reinforces the safety and potential efficacy of macrophage therapy in cirrhosis, supporting further investigation.
Collapse
Affiliation(s)
- Paul N Brennan
- Division of Molecular and Clinical Medicine, University of Dundee, Dundee, UK
- Centre for Regenerative Medicine, Institute for Regeneration and Repair, University of Edinburgh, Edinburgh, UK
| | - Mark MacMillan
- Centre for Regenerative Medicine, Institute for Regeneration and Repair, University of Edinburgh, Edinburgh, UK
| | - Thomas Manship
- Edinburgh Transplant Centre, Royal Infirmary of Edinburgh, Edinburgh, UK
| | | | - Alison Glover
- Scottish National Blood Transfusion Service (SNBTS), Edinburgh, UK
| | - Debbie Troland
- Centre for Regenerative Medicine, Institute for Regeneration and Repair, University of Edinburgh, Edinburgh, UK
| | - Iain MacPherson
- Division of Molecular and Clinical Medicine, University of Dundee, Dundee, UK
| | - Catriona Graham
- Wellcome Trust Clinical Research Facility, University of Edinburgh, Edinburgh, UK
| | - Rhona Aird
- Centre for Regenerative Medicine, Institute for Regeneration and Repair, University of Edinburgh, Edinburgh, UK
| | - Scott I K Semple
- Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, UK
| | - David M Morris
- Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, UK
| | | | - Chloe Pass
- Scottish National Blood Transfusion Service (SNBTS), Edinburgh, UK
| | - Neil W A McGowan
- Scottish National Blood Transfusion Service (SNBTS), Edinburgh, UK
| | - Marc L Turner
- Scottish National Blood Transfusion Service (SNBTS), Edinburgh, UK
| | - Lynn Manson
- Scottish National Blood Transfusion Service (SNBTS), Edinburgh, UK
| | | | - John F Dillon
- Division of Molecular and Clinical Medicine, University of Dundee, Dundee, UK
| | - Alastair M Kilpatrick
- Centre for Regenerative Medicine, Institute for Regeneration and Repair, University of Edinburgh, Edinburgh, UK
| | | | - Jonathan A Fallowfield
- Centre for Inflammation Research, Institute for Regeneration and Repair, University of Edinburgh, Edinburgh, UK
| | - Stuart J Forbes
- Centre for Regenerative Medicine, Institute for Regeneration and Repair, University of Edinburgh, Edinburgh, UK.
- Edinburgh Transplant Centre, Royal Infirmary of Edinburgh, Edinburgh, UK.
| |
Collapse
|
3
|
Fallowfield JA, Jimenez Ramos M. Emerging synthetic drugs for the treatment of hepatic cirrhosis: a 2024 update. Expert Opin Emerg Drugs 2024; 29:187-192. [PMID: 38603460 DOI: 10.1080/14728214.2024.2341878] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 01/23/2024] [Accepted: 04/08/2024] [Indexed: 04/13/2024]
Affiliation(s)
- Jonathan A Fallowfield
- Centre for Inflammation Research, Institute for Regeneration and Repair, University of Edinburgh, Edinburgh, UK
| | - Maria Jimenez Ramos
- Centre for Inflammation Research, Institute for Regeneration and Repair, University of Edinburgh, Edinburgh, UK
| |
Collapse
|
4
|
Lonardo A, Ballestri S, Mantovani A, Targher G, Bril F. Endpoints in NASH Clinical Trials: Are We Blind in One Eye? Metabolites 2024; 14:40. [PMID: 38248843 PMCID: PMC10820221 DOI: 10.3390/metabo14010040] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 12/08/2023] [Revised: 12/31/2023] [Accepted: 01/05/2024] [Indexed: 01/23/2024] Open
Abstract
This narrative review aims to illustrate the notion that nonalcoholic steatohepatitis (NASH), recently renamed metabolic dysfunction-associated steatohepatitis (MASH), is a systemic metabolic disorder featuring both adverse hepatic and extrahepatic outcomes. In recent years, several NASH trials have failed to identify effective pharmacological treatments and, therefore, lifestyle changes are the cornerstone of therapy for NASH. with this context, we analyze the epidemiological burden of NASH and the possible pathogenetic factors involved. These include genetic factors, insulin resistance, lipotoxicity, immuno-thrombosis, oxidative stress, reprogramming of hepatic metabolism, and hypoxia, all of which eventually culminate in low-grade chronic inflammation and increased risk of fibrosis progression. The possible explanations underlying the failure of NASH trials are also accurately examined. We conclude that the high heterogeneity of NASH, resulting from variable genetic backgrounds, exposure, and responses to different metabolic stresses, susceptibility to hepatocyte lipotoxicity, and differences in repair-response, calls for personalized medicine approaches involving research on noninvasive biomarkers. Future NASH trials should aim at achieving a complete assessment of systemic determinants, modifiers, and correlates of NASH, thus adopting a more holistic and unbiased approach, notably including cardiovascular-kidney-metabolic outcomes, without restricting therapeutic perspectives to histological surrogates of liver-related outcomes alone.
Collapse
Affiliation(s)
- Amedeo Lonardo
- AOU—Modena—Ospedale Civile di Baggiovara, 41126 Modena, Italy;
| | | | - Alessandro Mantovani
- Section of Endocrinology and Diabetes, Department of Medicine, University of Verona, Piazzale Stefani, 37126 Verona, Italy
| | - Giovanni Targher
- Department of Medicine, University of Verona, 37126 Verona, Italy;
- Metabolic Diseases Research Unit, IRCCS Sacro Cuore—Don Calabria Hospital, 37024 Negrar di Valpolicella, Italy
| | - Fernando Bril
- Department of Medicine, Heersink School of Medicine, University of Alabama at Birmingham (UAB), Birmingham, AL 35233, USA;
| |
Collapse
|
5
|
Knöchel J, Bergenholm L, Ibrahim E, Kechagias S, Hansson S, Liljeblad M, Nasr P, Carlsson B, Ekstedt M, Ueckert S. A Markov model of fibrosis development in nonalcoholic fatty liver disease predicts fibrosis progression in clinical cohorts. CPT Pharmacometrics Syst Pharmacol 2023; 12:2038-2049. [PMID: 37750001 PMCID: PMC10725269 DOI: 10.1002/psp4.13052] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 04/21/2023] [Revised: 08/28/2023] [Accepted: 09/06/2023] [Indexed: 09/27/2023] Open
Abstract
Disease progression in nonalcoholic steatohepatitis (NASH) is highly heterogenous and remains poorly understood. Fibrosis stage is currently the best predictor for development of end-stage liver disease and mortality. Better understanding and quantifying the impact of factors affecting NASH and fibrosis is essential to inform a clinical study design. We developed a population Markov model to describe the transition probability between fibrosis stages and mortality using a unique clinical nonalcoholic fatty liver disease cohort with serial biopsies over 3 decades. We evaluated covariate effects on all model parameters and performed clinical trial simulations to predict the fibrosis progression rate for external clinical cohorts. All parameters were estimated with good precision. Age and diagnosis of type 2 diabetes (T2D) were found to be significant predictors in the model. Increase in hepatic steatosis between visits was the most important predictor for progression of fibrosis. Fibrosis progression rate (FPR) was twofold higher for fibrosis stages 0 and 1 (F0-1) compared to fibrosis stage 2 and 3 (F2-3). A twofold increase in FPR was observed for T2D. A two-point steatosis worsening increased the FPR 11-fold. Predicted fibrosis progression was in good agreement with data from external clinical cohorts. Our fibrosis progression model shows that patient selection, particularly initial fibrosis stage distribution, can significantly impact fibrosis progression and as such the window for assessing drug efficacy in clinical trials. Our work highlights the increase in hepatic steatosis as the most important factor in increasing FPR, emphasizing the importance of well-defined lifestyle advise for reducing variability in NASH progression during clinical trials.
Collapse
Affiliation(s)
- Jane Knöchel
- Clinical Pharmacology and Quantitative PharmacologyClinical Pharmacology & Safety Sciences, R&D, AstraZenecaGothenburgSweden
| | - Linnéa Bergenholm
- DMPK, Research and Early Development, Cardiovascular, Renal and Metabolism (CVRM), BioPharmaceuticals R&D, AstraZenecaGothenburgSweden
| | - Eman Ibrahim
- Department of PharmacyUppsala UniversityUppsalaSweden
| | - Stergios Kechagias
- Department of Health, Medicine, and Caring SciencesLinköping UniversityLinköpingSweden
| | - Sara Hansson
- Translational Science and Experimental Medicine, Research and Early DevelopmentCardiovascular, Renal and Metabolism (CVRM), BioPharmaceuticals R&D, AstraZenecaGothenburgSweden
| | - Mathias Liljeblad
- Translational Science and Experimental Medicine, Research and Early DevelopmentCardiovascular, Renal and Metabolism (CVRM), BioPharmaceuticals R&D, AstraZenecaGothenburgSweden
| | - Patrik Nasr
- Department of Health, Medicine, and Caring SciencesLinköping UniversityLinköpingSweden
| | - Björn Carlsson
- Translational Science and Experimental Medicine, Research and Early DevelopmentCardiovascular, Renal and Metabolism (CVRM), BioPharmaceuticals R&D, AstraZenecaGothenburgSweden
| | - Mattias Ekstedt
- Department of Health, Medicine, and Caring SciencesLinköping UniversityLinköpingSweden
| | - Sebastian Ueckert
- Clinical Pharmacology and Quantitative PharmacologyClinical Pharmacology & Safety Sciences, R&D, AstraZenecaGothenburgSweden
| |
Collapse
|
6
|
Shi P, Zhu W, Fu J, Liang A, Zheng T, Wen Z, Wu X, Peng Y, Yuan S, Wu X. Avicularin alleviates acute liver failure by regulation of the TLR4/MyD88/NF-κB and Nrf2/HO-1/GPX4 pathways to reduce inflammation and ferroptosis. J Cell Mol Med 2023; 27:3326-3338. [PMID: 37644784 PMCID: PMC10623517 DOI: 10.1111/jcmm.17905] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 01/10/2023] [Revised: 05/04/2023] [Accepted: 08/02/2023] [Indexed: 08/31/2023] Open
Abstract
Acute liver failure (ALF) is an inflammation-mediated hepatocyte death process associated with ferroptosis. Avicularin (AL), a Chinese herbal medicine, exerts anti-inflammatory and antioxidative effects. However, the protective effect of AL and the mechanism on ALF have not been reported. Our in vivo results suggest that AL significantly alleviated lipopolysaccharide (LPS)/D-galactosamine (D-GalN)-induced hepatic pathological injury, liver enzymes, inflammatory cytokines, reactive oxygen species and iron levels and increased the antioxidant enzyme activities (malondialdehyde and glutathione). Our further in vitro experiments demonstrated that AL suppressed inflammatory response in LPS-stimulated RAW 264.7 cells via blocking the toll-like receptor 4 (TLR4)/myeloid differentiation protein-88 (MyD88)/nuclear factor kappa B (NF-κB) pathway. Moreover, AL attenuated ferroptosis in D-GalN-induced HepG2 cells by activating the nuclear factor erythroid 2-related factor 2 (Nrf2)/heme oxygenase 1 (HO-1)/glutathione peroxidase 4 (GPX4) pathway. Therefore, AL can alleviate inflammatory response and ferroptosis in LPS/D-GalN-induced ALF, and its protective effects are associated with blocking TLR4/MyD88/NF-κB pathway and activating Nrf2/HO-1/GPX4 pathway. Moreover, AL is a promising therapeutic option for ALF and should be clinically explored.
Collapse
Affiliation(s)
- Pei Shi
- Department of Infectious DiseasesThe First Affiliated Hospital of Nanchang UniversityNanchangChina
- Medical Innovation CenterThe First Affiliated Hospital of Nanchang UniversityNanchangChina
| | - Wentao Zhu
- Department of Infectious DiseasesThe First Affiliated Hospital of Nanchang UniversityNanchangChina
- Medical Innovation CenterThe First Affiliated Hospital of Nanchang UniversityNanchangChina
| | - Jiwei Fu
- Department of Infectious DiseasesThe First Affiliated Hospital of Nanchang UniversityNanchangChina
- Medical Innovation CenterThe First Affiliated Hospital of Nanchang UniversityNanchangChina
| | - An Liang
- Department of Infectious DiseasesThe First Affiliated Hospital of Nanchang UniversityNanchangChina
- Medical Innovation CenterThe First Affiliated Hospital of Nanchang UniversityNanchangChina
| | - Ting Zheng
- Department of Infectious DiseasesThe First Affiliated Hospital of Nanchang UniversityNanchangChina
| | - Zhilong Wen
- Department of Infectious DiseasesThe First Affiliated Hospital of Nanchang UniversityNanchangChina
| | - Xincheng Wu
- Department of Infectious DiseasesThe First Affiliated Hospital of Nanchang UniversityNanchangChina
| | - Yuchen Peng
- Department of Infectious DiseasesThe First Affiliated Hospital of Nanchang UniversityNanchangChina
| | - Songsong Yuan
- Department of Infectious DiseasesThe First Affiliated Hospital of Nanchang UniversityNanchangChina
| | - Xiaoping Wu
- Department of Infectious DiseasesThe First Affiliated Hospital of Nanchang UniversityNanchangChina
| |
Collapse
|
7
|
Heya MS, García-Ponce R, Soto BAM, Verde-Star MJ, Soto-Domínguez A, García-Hernandez DG, Saucedo-Cárdenas O, Hernández-Salazar M, Guillén-Meléndez GA. Green Alternatives in Treatment of Liver Diseases: the Challenges of Traditional Medicine and Green Nanomedicine. Chem Biodivers 2023; 20:e202300463. [PMID: 37531499 DOI: 10.1002/cbdv.202300463] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 04/01/2023] [Revised: 07/21/2023] [Accepted: 08/01/2023] [Indexed: 08/04/2023]
Abstract
Over the last decade, liver diseases have become a global problem, with approximately two million deaths per year. The high increase in the mortality rate of these diseases is mostly related to the limitations in the understanding of the evolutionary clinical cases of liver diseases, the low delivery of drugs in the liver, the non-specific administration of drugs, and the side effects generated at the systemic level by conventional therapeutic agents. Today it is common knowledge that phytochemicals have a high curative potential, even in the prevention and/or reversibility of liver disorders; however, even using these green molecules, researchers continue to deal with the same challenges implemented with conventional therapeutic agents, which limits the pharmacological potential of these friendly molecules. On the other hand, the latest advances in nanotechnology have proven that the use of nanocarriers as a delivery system for green active ingredients, as well as conventional ones, increases the pharmacological potential of these active ingredients due to their physicochemical characteristics (size, Zeta potential, etc.,) moldable depending on the therapeutic objective; in addition to the above, it should be noted that in recent years, nanoparticles have been developed for the specific delivery of drugs towards a specific target (stellar cells, hepatocytes, Kupffer cells), depending on the clinical state of the disease in the patient. The present review addresses the challenges of traditional medicine and green nanomedicine as alternatives in the treatment of liver diseases.
Collapse
Affiliation(s)
- Michel Stephane Heya
- Faculty of Public Health and Nutrition, Universidad Autónoma de Nuevo León, Ave. Pedro de Alba S/N & Ave. Manuel L. Barragán, San Nicolas de los Garza, 66451, Nuevo León, México
| | - Romario García-Ponce
- Biological Science School, Universidad Autónoma de Nuevo León, Ave., Pedro de Alba S/N & Ave. Manuel L. Barragán, San Nicolás de los Garza, 66451, Nuevo León, México
| | - Beatriz Amari Medina Soto
- Department of Microbiology, Faculty of Veterinary Medicine and Zootechnics., Universidad Autónoma de Nuevo León, Francisco Villa S/N, Ex Hacienda El Canadá, Gral. Escobedo, Nuevo León, México
| | - María Julia Verde-Star
- Biological Science School, Universidad Autónoma de Nuevo León, Ave., Pedro de Alba S/N & Ave. Manuel L. Barragán, San Nicolás de los Garza, 66451, Nuevo León, México
| | - Adolfo Soto-Domínguez
- Department of Histology, Faculty of Medicine, Universidad Autónoma de Nuevo León, Madero y Aguirre Pequeño S/N, Mitras Centro, 64460, Monterrey, Nuevo León, México
| | - David Gilberto García-Hernandez
- Biological Science School, Universidad Autónoma de Nuevo León, Ave., Pedro de Alba S/N & Ave. Manuel L. Barragán, San Nicolás de los Garza, 66451, Nuevo León, México
| | - Odila Saucedo-Cárdenas
- Department of Histology, Faculty of Medicine, Universidad Autónoma de Nuevo León, Madero y Aguirre Pequeño S/N, Mitras Centro, 64460, Monterrey, Nuevo León, México
| | - Marcelo Hernández-Salazar
- Faculty of Public Health and Nutrition, Universidad Autónoma de Nuevo León, Ave. Pedro de Alba S/N & Ave. Manuel L. Barragán, San Nicolas de los Garza, 66451, Nuevo León, México
| | - Gloria Arely Guillén-Meléndez
- Department of Histology, Faculty of Medicine, Universidad Autónoma de Nuevo León, Madero y Aguirre Pequeño S/N, Mitras Centro, 64460, Monterrey, Nuevo León, México
| |
Collapse
|
8
|
Brennan PN, Elsharkawy AM, Kendall TJ, Loomba R, Mann DA, Fallowfield JA. Antifibrotic therapy in nonalcoholic steatohepatitis: time for a human-centric approach. Nat Rev Gastroenterol Hepatol 2023; 20:679-688. [PMID: 37268740 PMCID: PMC10236408 DOI: 10.1038/s41575-023-00796-x] [Citation(s) in RCA: 31] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Academic Contribution Register] [Accepted: 05/09/2023] [Indexed: 06/04/2023]
Abstract
Nonalcoholic steatohepatitis (NASH) might soon become the leading cause of end-stage liver disease and indication for liver transplantation worldwide. Fibrosis severity is the only histological predictor of liver-related morbidity and mortality in NASH identified to date. Moreover, fibrosis regression is associated with improved clinical outcomes. However, despite numerous clinical trials of plausible drug candidates, an approved antifibrotic therapy remains elusive. Increased understanding of NASH susceptibility and pathogenesis, emerging human multiomics profiling, integration of electronic health record data and modern pharmacology techniques hold enormous promise in delivering a paradigm shift in antifibrotic drug development in NASH. There is a strong rationale for drug combinations to boost efficacy, and precision medicine strategies targeting key genetic modifiers of NASH are emerging. In this Perspective, we discuss why antifibrotic effects observed in NASH pharmacotherapy trials have been underwhelming and outline potential approaches to improve the likelihood of future clinical success.
Collapse
Affiliation(s)
- Paul N Brennan
- Institute for Regeneration & Repair, University of Edinburgh, Edinburgh, UK
- Division of Molecular and Clinical Medicine, University of Dundee, Dundee, UK
| | - Ahmed M Elsharkawy
- Liver Unit and NIHR Biomedical Research Centre, University Hospitals Birmingham NHS Foundation Trust, Birmingham, UK
| | - Timothy J Kendall
- Institute for Regeneration & Repair, University of Edinburgh, Edinburgh, UK
- Edinburgh Pathology, University of Edinburgh, Edinburgh, UK
| | - Rohit Loomba
- NAFLD Research Centre, Division of Gastroenterology and Hepatology, UC San Diego School of Medicine, La Jolla, CA, USA
| | - Derek A Mann
- Fibrosis Research Group, Newcastle University, Newcastle, UK.
- Department of Gastroenterology and Hepatology, School of Medicine, Koç University, Istanbul, Turkey.
| | | |
Collapse
|
9
|
Wu Z, Zhou H, Liu D, Deng F. Alterations in the gut microbiota and the efficacy of adjuvant probiotic therapy in liver cirrhosis. Front Cell Infect Microbiol 2023; 13:1218552. [PMID: 37483387 PMCID: PMC10361729 DOI: 10.3389/fcimb.2023.1218552] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 05/07/2023] [Accepted: 06/15/2023] [Indexed: 07/25/2023] Open
Abstract
Background Liver cirrhosis is the end stage of various chronic liver diseases (CLDs). The gut microbiota can impact the liver environment and trigger chronic liver inflammation through the gut-liver axis. Alteration of the gut microbiota has become an effective strategy in the biological treatment of cirrhosis. Methods Twenty-eight patients with liver cirrhosis and 16 healthy individuals were included, and fresh stool samples were collected. We analyzed changes in the gut microbiota between groups by 16S rRNA sequencing and evaluated the association between microbiota alterations and hepatic function. Additionally, 102 cirrhotic patients were retrospectively enrolled and divided into a probiotic group (n=44) and a nonprobiotic group (n=58) in addition to standard treatment for cirrhosis. Patients were monitored for hematological parameters and hepatic function during the six-month follow-up. Results The gut microbiota profile of patients with cirrhosis was greatly different from that of healthy individuals, presenting with significantly reduced α diversity and decreased abundance of representative SCFA-producing bacteria including Firmicutes, Coprococcus and Clostridium IV. The pathogenic bacteria Gammaproteobacteria, Veillonella, and Bacilli were greatly enriched in cirrhotic patients. Additionally, patients with decompensated cirrhosis (DCPC) had a significantly reduced abundance of Oscillibacter compared to compensated cirrhosis (CPC), which is also a SCFA-producing bacteria, and the lower Firmicutes to Bacteroidetes ratio and enhanced MDR values were also shown in DCPC patients compared to CPC patients. In addition, the abundance of Firmicutes was negatively related to hepatic function in cirrhotic patients, including the levels of ALT, AST, and DBIL. From the retrospective study, we found that biochemical improvements in alanine transaminase (ALT) and total bilirubin (TBIL) were obtained in DCPC patients who received oral probiotic therapy compared with the nonprobiotic group. Conclusion Severe microbial dysbiosis existed in patients with liver cirrhosis, especially patients who reached the decompensatory stage. SCFA-producing bacteria were significantly reduced in cirrhosis. Altered gut microbiota cause changes in functional modules, which may contribute to cirrhosis progression and are associated with clinical prognosis. Adjuvant probiotic supplementation to enhance SCFA-producing bacteria can be a prospective therapy for patients with cirrhosis.
Collapse
Affiliation(s)
- Zengrong Wu
- Department of Gastroenterology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
- Research Center of Digestive Disease, Central South University, Changsha, Hunan, China
| | - Hejun Zhou
- Department of Gastroenterology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
- Research Center of Digestive Disease, Central South University, Changsha, Hunan, China
| | - Deliang Liu
- Department of Gastroenterology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
- Research Center of Digestive Disease, Central South University, Changsha, Hunan, China
| | - Feihong Deng
- Department of Gastroenterology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
- Research Center of Digestive Disease, Central South University, Changsha, Hunan, China
| |
Collapse
|
10
|
Hamberger F, Mederacke YS, Mederacke I. An inducible model for genetic manipulation and fate-tracing of PDGFRβ-expressing fibrogenic cells in the liver. Sci Rep 2023; 13:7322. [PMID: 37147343 PMCID: PMC10162963 DOI: 10.1038/s41598-023-34353-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 11/15/2022] [Accepted: 04/27/2023] [Indexed: 05/07/2023] Open
Abstract
Myofibroblasts are the source of extracellular matrix protein during liver fibrogenesis. Fibroblasts, hepatic stellate cells (HSCs) and vascular smooth muscle cells are mesenchymal subpopulations in the liver that are characterized by the expression of PDGFRβ and contribute to the pool of these myofibroblasts. Conditional knockout models are important to better understand the function of specific liver cell populations including mesenchymal cells. While there is a limited number of constitutive mouse models for liver mesenchymal cell specific transgene expression, there is no established model for inducible gene targeting in HSCs or PDGFRβ-expressing mesenchymal cell populations in the liver. To address this, we investigated whether the tamoxifen inducible PDGFRβ-P2A-CreERT2 mouse can be used as a reliable tool to specifically express transgens in liver mesenchymal cells. Our data demonstrate, that PDGFRβ-P2A-CreERT2 specifically and efficiently marks over 90% of retinoid positive HSCs in healthy and fibrotic liver in mice upon tamoxifen injection, and that those cells give rise to Col1a1-expressing myofibroblasts in different models of liver fibrosis. Together with a negligible background recombination of only about 0.33%, this confirms that the PDGFRβ-P2A-CreERT2 mouse is nearly as efficient as established constitutive LratCre and PDGFRβ-Cre mouse models for recombination in HSCs, and that it is a powerful model for mesenchymal liver cell studies that require an inducible Cre approach.
Collapse
Affiliation(s)
- Florian Hamberger
- Department of Gastroenterology, Hepatology, Infectious Diseases and Endocrinology, Hannover Medical School, Carl-Neuberg-Str. 1, 30625, Hannover, Germany
| | - Young-Seon Mederacke
- Department of Gastroenterology, Hepatology, Infectious Diseases and Endocrinology, Hannover Medical School, Carl-Neuberg-Str. 1, 30625, Hannover, Germany
| | - Ingmar Mederacke
- Department of Gastroenterology, Hepatology, Infectious Diseases and Endocrinology, Hannover Medical School, Carl-Neuberg-Str. 1, 30625, Hannover, Germany.
| |
Collapse
|
11
|
Xiao S, Pan X, Huang X, Liu Y, Wen SW, Liu A. Gene polymorphisms of inflammatory factors in liver cirrhosis. Front Genet 2023; 14:1140427. [PMID: 37101651 PMCID: PMC10123281 DOI: 10.3389/fgene.2023.1140427] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 01/09/2023] [Accepted: 03/27/2023] [Indexed: 04/28/2023] Open
Abstract
Introduction: Studies on the association between gene polymorphisms of various inflammatory factors and liver cirrhosis have been inconsistent. The purpose of this study was to comprehensively summarize the available evidence on the association between gene polymorphisms of inflammatory factors and liver cirrhosis through a systematic review. Methods: We searched databases of PubMed, EMBASE, Web of Science, and the Cochrane Library for relevant articles published from building databases to 25 September 2022. A systematic review and meta-analysis were performed to investigate the association between gene polymorphisms of various inflammatory factors and liver cirrhosis. Odds ratios (OR) and 95% confidence intervals (CI) were used to assess the strength of association. Results: A total of 43 articles were included in the systematic review and of them, 22 articles were included in the meta-analysis. The gene polymorphisms of IL-10-1082 GA + AA vs. GG (OR = 1.43, 95% CI = 1.12-1.83), IL-10-1082 AA vs. GG (OR = 2.03, 95% CI = 1.36-3.02), IL-18 -137 GG vs. CC (OR = 3.84, 95% CI = 1.29-11.40), TGF-β1 -509 T vs. C (OR = 2.52, 95% CI = 1.42-4.48), and IFN-γ +874 T vs. A (OR = 1.98, 95% CI = 1.32-2.98) were associated with liver cirrhosis significantly and no association was observed in other gene polymorphisms included in the meta-analysis. The review of inflammatory factors gene polymorphisms that were only reported by a single study indicated 19 gene polymorphisms were risk factors and 4 gene polymorphisms were protective factors for liver cirrhosis, while the association between other 27 gene polymorphisms and liver cirrhosis were not statistically significant. Discussion: This study suggests that IL-10 -1082G/A, IL-18 -137G/C, TGF-β1 -509T/C, and IFN-γ +874T/A were potentially associated with the risk of liver cirrhosis susceptibility. These findings may provide comprehensive evidence for genetic susceptibility and immunogenetic pathology of liver cirrhosis.
Collapse
Affiliation(s)
- Sailan Xiao
- Department of Epidemiology and Health Statistics, Xiangya School of Public Health, Central South University, Changsha, China
| | - Xiongfeng Pan
- Department of Epidemiology and Health Statistics, Xiangya School of Public Health, Central South University, Changsha, China
| | - Xun Huang
- Department of Epidemiology and Health Statistics, Xiangya School of Public Health, Central South University, Changsha, China
| | - Yamin Liu
- Department of Epidemiology and Health Statistics, Xiangya School of Public Health, Central South University, Changsha, China
| | - Shi Wu Wen
- Clinical Epidemiology Program, Ottawa Hospital Research Institute, Ottawa, ON, Canada
- Department of Obstetrics and Gynaecology, University of Ottawa Faculty of Medicine, Ottawa, ON, Canada
- School of Epidemiology and Public Health, University of Ottawa Faculty of Medicine, Ottawa, ON, Canada
| | - Aizhong Liu
- Department of Epidemiology and Health Statistics, Xiangya School of Public Health, Central South University, Changsha, China
- Hunan Provincial Key Laboratory of Clinical Epidemiology, Xiangya School of Public Health, Central South University, Changsha, China
| |
Collapse
|
12
|
Abouelezz HM, Shehatou GS, Shebl AM, Salem HA. A standardized pomegranate fruit extract ameliorates thioacetamide-induced liver fibrosis in rats via AGE-RAGE-ROS signaling. Heliyon 2023; 9:e14256. [PMID: 36938469 PMCID: PMC10015255 DOI: 10.1016/j.heliyon.2023.e14256] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 07/29/2022] [Revised: 02/22/2023] [Accepted: 02/28/2023] [Indexed: 03/07/2023] Open
Abstract
This work aimed to investigate a possible mechanism that may mediate the hepatoprotective effects of pomegranate fruit extract (PFE) against thioacetamide (THIO)-induced liver fibrosis in rats. Male Sprague Dawley rats were randomly allocated into four groups (n = 8 each): control; PFE (150 mg/kg/day, orally); THIO (200 mg/kg, i.p, 3 times a week); and THIO and PFE-treated groups. Oral PFE treatment decreased liver/body weight ratio by 12.4%, diminished serum function levels of ALT, AST, ALP, LDH, and total bilirubin, increased serum albumin, boosted hepatic GSH (by 35.6%) and SOD (by 17.5%), and significantly reduced hepatic levels of ROS, MDA, 4-HNE, AGEs, and RAGE in THIO-fibrotic rats relative to untreated THIO group. Moreover, PFE administration downregulated the hepatic levels of profibrotic TGF-β1 (by 23.0%, P < 0.001) and TIMP-1 (by 41.5%, P < 0.001), attenuated α-SMA protein expression, decreased serum HA levels (by 41.3%), and reduced the hepatic levels of the fibrosis markers hydroxyproline (by 26.0%, P < 0.001), collagen type IV (by 44.3%, P < 0.001) and laminin (by 43.4%, P < 0.001) compared to the untreated THIO group. The histopathological examination has corroborated these findings, where PFE decreased hepatic nodule incidence, attenuated portal necroinflammation and reduced extent of fibrosis. These findings may suggest that oral PFE administration could slow the progression of hepatic fibrogenesis via reducing hepatic levels of AGEs, RAGE, ROS, TGF-β1, and TIMP-1.
Collapse
Affiliation(s)
- Hadeer M. Abouelezz
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Mansoura University, Mansoura, Egypt
- Corresponding author.
| | - George S.G. Shehatou
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Mansoura University, Mansoura, Egypt
- Department of Pharmacology and Biochemistry, Faculty of Pharmacy, Delta University for Science and Technology, Gamasa City, Egypt
| | - Abdelhadi M. Shebl
- Department of Pathology, Faculty of Medicine, Mansoura University, Mansoura, Egypt
| | - Hatem A. Salem
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Mansoura University, Mansoura, Egypt
| |
Collapse
|
13
|
Toprakhisar B, Verfaillie CM, Kumar M. Advances in Recellularization of Decellularized Liver Grafts with Different Liver (Stem) Cells: Towards Clinical Applications. Cells 2023; 12:301. [PMID: 36672236 PMCID: PMC9856398 DOI: 10.3390/cells12020301] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 12/06/2022] [Revised: 12/22/2022] [Accepted: 12/28/2022] [Indexed: 01/15/2023] Open
Abstract
Liver transplantation is currently the only curative therapy for patients with acute or chronic liver failure. However, a dramatic gap between the number of available liver grafts and the number of patients on the transplantation waiting list emphasizes the need for valid liver substitutes. Whole-organ engineering is an emerging field of tissue engineering and regenerative medicine. It aims to generate transplantable and functional organs to support patients on transplantation waiting lists until a graft becomes available. It comprises two base technologies developed in the last decade; (1) organ decellularization to generate a three-dimensional (3D) extracellular matrix scaffold of an organ, and (2) scaffold recellularization to repopulate both the parenchymal and vascular compartments of a decellularized organ. In this review article, recent advancements in both technologies, in relation to liver whole-organ engineering, are presented. We address the potential sources of hepatocytes and non-parenchymal liver cells for repopulation studies, and the role of stem-cell-derived liver progeny is discussed. In addition, different cell seeding strategies, possible graft modifications, and methods used to evaluate the functionality of recellularized liver grafts are outlined. Based on the knowledge gathered from recent transplantation studies, future directions are summarized.
Collapse
Affiliation(s)
- Burak Toprakhisar
- Stem Cell Institute, Department of Stem Cell and Developmental Biology, KU Leuven, 3000 Leuven, Belgium
| | | | | |
Collapse
|
14
|
Ma J, Zhao Q, Chen M, Wang W, He B, Jiang Y, Li Y. microRNA-122 inhibits hepatic stellate cell proliferation and activation in vitro and represses carbon tetrachloride-induced liver cirrhosis in mice. Ann Hepatol 2022; 27:100700. [PMID: 35338010 DOI: 10.1016/j.aohep.2022.100700] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Academic Contribution Register] [Received: 12/31/2021] [Revised: 02/24/2022] [Accepted: 02/24/2022] [Indexed: 02/04/2023]
Abstract
OBJECTIVE This study aimed to determine the roles of microRNA (miR)-122 in the activation of hepatic stellate cells (HSCs) and liver cirrhosis. METHODS Rat primary HSCs were incubated with transforming growth factor-beta (TGF-β), during which miR-122 and EphB2 expression was measured. miR-122 mimic and/or pcDNA3.1 EphB2 was transfected into TGF-β-induced HSCs. A mouse model of liver cirrhosis was established via an intraperitoneal injection of carbon tetrachloride (CCl4), followed by the injection of miR-122 agomir. Levels of serum alanine transaminase (ALT) and aspartate aminotransferase (AST) were measured. Fibronectin (FN), alpha smooth muscle actin (α-SMA), Collagen I, miR-122, and EphB2 expression was evaluated in liver tissues and HSCs. Cell proliferation was measured using CCK-8 assay. Interactions between miR-122 and EphB2 were assessed using dual luciferase reporter assay. RESULTS miR-122 (0.15-fold) was downregulated and EphB2 (mRNA: 5.06-fold; protein: 2.35-fold) was upregulated after TGF-β induction of HSCs. Overexpressed miR-122 decreased proliferation and EphB2 (mRNA: 0.46-fold; protein: 0.62-fold), FN (mRNA: 0.45-fold; protein: 0.64-fold), α-SMA (mRNA: 0.48-fold; protein: 0.51-fold), and Collagen I (mRNA: 0.44-fold; protein: 0.51-fold) expression in HSCs, which was abrogated by EphB2 upregulation. miR-122 expression was reduced by 0.21-fold and serum ALT and AST levels were enhanced in mice following 8-week CCl4 induction along with increased expression of FN, α-SMA, and Collagen I in liver tissues, which was blocked by miR-122 overexpression. Moreover, EphB2 was a target gene of miR-122. CONCLUSION miR-122 curtails HSC proliferation and activation by targeting EphB2 and suppresses liver cirrhosis in mice.
Collapse
Affiliation(s)
- Jing Ma
- Department of Infectious Diseases, the Second Xiangya Hospital of Central South University, Changsha, Hunan 410011, P.R. China
| | - Qiqian Zhao
- Department of Infectious Diseases, the Second Xiangya Hospital of Central South University, Changsha, Hunan 410011, P.R. China
| | - Mengxuan Chen
- Department of Infectious Diseases, the Second Xiangya Hospital of Central South University, Changsha, Hunan 410011, P.R. China
| | - Weihang Wang
- Department of Infectious Diseases, the Second Xiangya Hospital of Central South University, Changsha, Hunan 410011, P.R. China
| | - Bo He
- Department of Infectious Diseases, the Second Xiangya Hospital of Central South University, Changsha, Hunan 410011, P.R. China
| | - Yongfang Jiang
- Department of Infectious Diseases, the Second Xiangya Hospital of Central South University, Changsha, Hunan 410011, P.R. China
| | - Yi Li
- Department of Infectious Diseases, the Second Xiangya Hospital of Central South University, Changsha, Hunan 410011, P.R. China.
| |
Collapse
|
15
|
Akbari B, Baghaei-Yazdi N, Bahmaie M, Mahdavi Abhari F. The role of plant-derived natural antioxidants in reduction of oxidative stress. Biofactors 2022; 48:611-633. [PMID: 35229925 DOI: 10.1002/biof.1831] [Citation(s) in RCA: 135] [Impact Index Per Article: 45.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Academic Contribution Register] [Received: 12/02/2021] [Revised: 01/31/2022] [Accepted: 02/10/2022] [Indexed: 12/15/2022]
Abstract
Free radicals are a group of damaging molecules produced during the normal metabolism of cells in the human body. Exposure to ultraviolet radiation, cigarette smoking, and other environmental pollutants enhances free radicals in the human body. The destructive effects of free radicals may also cause harm to membranes, enzymes, and DNA, leading to several human diseases such as cancer, atherosclerosis, malaria, coronavirus disease (COVID-19), rheumatoid arthritis, and neurodegenerative illnesses. This process occurs when there is an imbalance between free radicals and antioxidant defenses. Since antioxidants scavenge free radicals and repair damaged cells, increasing the consumption of fruits and vegetables containing high antioxidant values is recommended to slow down oxidative stress in the body. Additionally, natural products demonstrated a wide range of biological impacts such as anti-inflammatory, anti-aging, anti-atherosclerosis, and anti-cancer properties. Hence, in this review article, our goal is to explore the role of natural therapeutic antioxidant effects to reduce oxidative stress in the diseases.
Collapse
Affiliation(s)
- Behnaz Akbari
- School of Medicine, Department of Anatomy & Neurobiology, Boston University, Boston, Massachusetts, USA
| | - Namdar Baghaei-Yazdi
- College of Liberal Arts & Sciences, School of Life Sciences, University of Westminster, London, UK
| | - Manochehr Bahmaie
- Department of Chemistry, Islamic Azad University, North Tehran Branch, Tehran, Iran
| | | |
Collapse
|
16
|
Oakley F. Interrogating mechanisms of liver fibrosis with omics. Nat Rev Gastroenterol Hepatol 2022; 19:89-90. [PMID: 34903888 DOI: 10.1038/s41575-021-00567-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Academic Contribution Register] [Indexed: 12/08/2022]
Affiliation(s)
- Fiona Oakley
- Newcastle Fibrosis Research Group, Biosciences Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, UK.
| |
Collapse
|
17
|
Ramos MJ, Bandiera L, Menolascina F, Fallowfield JA. In vitro models for non-alcoholic fatty liver disease: Emerging platforms and their applications. iScience 2022; 25:103549. [PMID: 34977507 PMCID: PMC8689151 DOI: 10.1016/j.isci.2021.103549] [Citation(s) in RCA: 48] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 02/07/2023] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) represents a global healthcare challenge, affecting 1 in 4 adults, and death rates are predicted to rise inexorably. The progressive form of NAFLD, non-alcoholic steatohepatitis (NASH), can lead to fibrosis, cirrhosis, and hepatocellular carcinoma. However, no medical treatments are licensed for NAFLD-NASH. Identifying efficacious therapies has been hindered by the complexity of disease pathogenesis, a paucity of predictive preclinical models and inadequate validation of pharmacological targets in humans. The development of clinically relevant in vitro models of the disease will pave the way to overcome these challenges. Currently, the combined application of emerging technologies (e.g., organ-on-a-chip/microphysiological systems) and control engineering approaches promises to unravel NAFLD biology and deliver tractable treatment candidates. In this review, we will describe advances in preclinical models for NAFLD-NASH, the recent introduction of novel technologies in this space, and their importance for drug discovery endeavors in the future.
Collapse
Affiliation(s)
- Maria Jimenez Ramos
- Centre for Inflammation Research, The University of Edinburgh, The Queen's Medical Research Institute, Edinburgh EH16 4TJ, UK
| | - Lucia Bandiera
- Institute for Bioengineering, The University of Edinburgh, Edinburgh EH9 3BF, UK.,Synthsys - Centre for Synthetic and Systems Biology, The University of Edinburgh, Edinburgh EH9 3BF, UK
| | - Filippo Menolascina
- Institute for Bioengineering, The University of Edinburgh, Edinburgh EH9 3BF, UK.,Synthsys - Centre for Synthetic and Systems Biology, The University of Edinburgh, Edinburgh EH9 3BF, UK
| | - Jonathan Andrew Fallowfield
- Centre for Inflammation Research, The University of Edinburgh, The Queen's Medical Research Institute, Edinburgh EH16 4TJ, UK
| |
Collapse
|
18
|
Ramos MJ, Bandiera L, Menolascina F, Fallowfield JA. In vitro models for non-alcoholic fatty liver disease: Emerging platforms and their applications. iScience 2022; 25:103549. [PMID: 34977507 DOI: 10.1016/j.isci] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 05/23/2023] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) represents a global healthcare challenge, affecting 1 in 4 adults, and death rates are predicted to rise inexorably. The progressive form of NAFLD, non-alcoholic steatohepatitis (NASH), can lead to fibrosis, cirrhosis, and hepatocellular carcinoma. However, no medical treatments are licensed for NAFLD-NASH. Identifying efficacious therapies has been hindered by the complexity of disease pathogenesis, a paucity of predictive preclinical models and inadequate validation of pharmacological targets in humans. The development of clinically relevant in vitro models of the disease will pave the way to overcome these challenges. Currently, the combined application of emerging technologies (e.g., organ-on-a-chip/microphysiological systems) and control engineering approaches promises to unravel NAFLD biology and deliver tractable treatment candidates. In this review, we will describe advances in preclinical models for NAFLD-NASH, the recent introduction of novel technologies in this space, and their importance for drug discovery endeavors in the future.
Collapse
Affiliation(s)
- Maria Jimenez Ramos
- Centre for Inflammation Research, The University of Edinburgh, The Queen's Medical Research Institute, Edinburgh EH16 4TJ, UK
| | - Lucia Bandiera
- Institute for Bioengineering, The University of Edinburgh, Edinburgh EH9 3BF, UK
- Synthsys - Centre for Synthetic and Systems Biology, The University of Edinburgh, Edinburgh EH9 3BF, UK
| | - Filippo Menolascina
- Institute for Bioengineering, The University of Edinburgh, Edinburgh EH9 3BF, UK
- Synthsys - Centre for Synthetic and Systems Biology, The University of Edinburgh, Edinburgh EH9 3BF, UK
| | - Jonathan Andrew Fallowfield
- Centre for Inflammation Research, The University of Edinburgh, The Queen's Medical Research Institute, Edinburgh EH16 4TJ, UK
| |
Collapse
|
19
|
Brennan PN, MacMillan M, Manship T, Moroni F, Glover A, Graham C, Semple S, Morris DM, Fraser AR, Pass C, McGowan NWA, Turner ML, Lachlan N, Dillon JF, Campbell JDM, Fallowfield JA, Forbes SJ. Study protocol: a multicentre, open-label, parallel-group, phase 2, randomised controlled trial of autologous macrophage therapy for liver cirrhosis (MATCH). BMJ Open 2021; 11:e053190. [PMID: 34750149 PMCID: PMC8576470 DOI: 10.1136/bmjopen-2021-053190] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Academic Contribution Register] [Indexed: 01/06/2023] Open
Abstract
INTRODUCTION Liver cirrhosis is a growing global healthcare challenge. Cirrhosis is characterised by severe liver fibrosis, organ dysfunction and complications related to portal hypertension. There are no licensed antifibrotic or proregenerative medicines and liver transplantation is a scarce resource. Hepatic macrophages can promote both liver fibrogenesis and fibrosis regression. The safety and feasibility of peripheral infusion of ex vivo matured autologous monocyte-derived macrophages in patients with compensated cirrhosis has been demonstrated. METHODS AND ANALYSIS The efficacy of autologous macrophage therapy, compared with standard medical care, will be investigated in a cohort of adult patients with compensated cirrhosis in a multicentre, open-label, parallel-group, phase 2, randomised controlled trial. The primary outcome is the change in Model for End-Stage Liver Disease score at 90 days. The trial will provide the first high-quality examination of the efficacy of autologous macrophage therapy in improving liver function, non-invasive fibrosis markers and other clinical outcomes in patients with compensated cirrhosis. ETHICS AND DISSEMINATION The trial will be conducted according to the ethical principles of the Declaration of Helsinki 2013 and has been approved by Scotland A Research Ethics Committee (reference 15/SS/0121), National Health Service Lothian Research and Development department and the Medicine and Health Care Regulatory Agency-UK. Final results will be presented in peer-reviewed journals and at relevant conferences. TRIAL REGISTRATION NUMBERS ISRCTN10368050 and EudraCT; reference 2015-000963-15.
Collapse
Affiliation(s)
- Paul Noel Brennan
- Centre for Regenerative Medicine, The University of Edinburgh Medical School, Edinburgh, UK
| | - Mark MacMillan
- Centre for Regenerative Medicine, The University of Edinburgh Medical School, Edinburgh, UK
| | | | | | - Alison Glover
- Scottish National Blood Transfusion Service, Edinburgh, UK
| | - Catriona Graham
- Deanery of Clinical Sciences, The University of Edinburgh, Edinburgh, UK
| | - Scott Semple
- Centre for Cardiovascular Science, The University of Edinburgh Deanery of Clinical Sciences, Edinburgh, UK
| | - David M Morris
- Centre for Cardiovascular Science, The University of Edinburgh Deanery of Clinical Sciences, Edinburgh, UK
| | | | - Chloe Pass
- Tissues, Cells and Advanced Therapeutics, SNBTS, Edinburgh, UK
| | | | - Marc L Turner
- Tissues, Cells and Advanced Therapeutics, SNBTS, Edinburgh, UK
| | - Neil Lachlan
- Department of Gastroenterology, NHS Greater Glasgow and Clyde, Glasgow, UK
| | - John F Dillon
- Liver Group, University of Dundee Division of Cardiovascular and Diabetes Medicine, Dundee, UK
| | | | - Jonathan Andrew Fallowfield
- Queen's Medical Research Institute, University of Edinburgh MRC Centre for Inflammation Research, Edinburgh, UK
| | - Stuart J Forbes
- Centre for Regenerative Medicine, The University of Edinburgh Deanery of Clinical Sciences, Edinburgh, UK
| |
Collapse
|