15
views
0
recommends
+1 Recommend
0 collections
    0
    shares
      • Record: found
      • Abstract: found
      • Article: found
      Is Open Access

      Identification and characterisation of a rare MTTP variant underlying hereditary non-alcoholic fatty liver disease

      research-article
      1 , 2 , , 3 , 4 , , 5 , , 6 , 5 , 7 , 5 , 7 , 8 , 9 , 3 , 4 , 5 , 5 , 6 , 6 , § , 10 , 11 , 11 , 12 , 13 , 14 , 14 , , 15 , ∗∗ , 16 , 16 , 15 , , 3 , 4 , , 1 , 2 , ,
      JHEP Reports
      Elsevier
      Microsomal triglyceride transfer protein, Abetalipoproteinaemia, hiPSC-derived hepatocytes, Lipoprotein ApoB

      Read this article at

      Bookmark
          There is no author summary for this article yet. Authors can add summaries to their articles on ScienceOpen to make them more accessible to a non-specialist audience.

          Abstract

          Background & Aims

          Non-alcoholic fatty liver disease (NAFLD) is a complex trait with an estimated prevalence of 25% globally. We aimed to identify the genetic variant underlying a four-generation family with progressive NAFLD leading to cirrhosis, decompensation, and development of hepatocellular carcinoma in the absence of common risk factors such as obesity and type 2 diabetes.

          Methods

          Exome sequencing and genome comparisons were used to identify the likely causal variant. We extensively characterised the clinical phenotype and post-prandial metabolic responses of family members with the identified novel variant in comparison with healthy non-carriers and wild-type patients with NAFLD. Variant-expressing hepatocyte-like cells (HLCs) were derived from human-induced pluripotent stem cells generated from homozygous donor skin fibroblasts and restored to wild-type using CRISPR-Cas9. The phenotype was assessed using imaging, targeted RNA analysis, and molecular expression arrays.

          Results

          We identified a rare causal variant c.1691T>C p.I564T (rs745447480) in MTTP, encoding microsomal triglyceride transfer protein (MTP), associated with progressive NAFLD, unrelated to metabolic syndrome and without characteristic features of abetalipoproteinaemia. HLCs derived from a homozygote donor had significantly lower MTP activity and lower lipoprotein ApoB secretion than wild-type cells, while having similar levels of MTP mRNA and protein. Cytoplasmic triglyceride accumulation in HLCs triggered endoplasmic reticulum stress, secretion of pro-inflammatory mediators, and production of reactive oxygen species.

          Conclusions

          We have identified and characterised a rare causal variant in MTTP, and homozygosity for MTTP p .I564T is associated with progressive NAFLD without any other manifestations of abetalipoproteinaemia. Our findings provide insights into mechanisms driving progressive NAFLD.

          Impact and Implications

          A rare genetic variant in the gene MTTP has been identified as responsible for the development of severe non-alcoholic fatty liver disease in a four-generation family with no typical disease risk factors. A cell line culture created harbouring this variant gene was characterised to understand how this genetic variation leads to a defect in liver cells, which results in accumulation of fat and processes that promote disease. This is now a useful model for studying the disease pathways and to discover new ways to treat common types of fatty liver disease.

          Graphical abstract

          Highlights

          • A rare MTTP variant, p.I564T, leads to progressive fatty liver disease and cirrhosis.

          • Hepatocyte-like cells expressing MTP 564T have reduced liver lipoprotein secretion capability.

          • Reactive oxygen species, NF-κB pathway, and extracellular matrix remodelling drive lipotoxicity.

          Related collections

          Most cited references40

          • Record: found
          • Abstract: not found
          • Article: not found

          VMD: Visual molecular dynamics

            Bookmark
            • Record: found
            • Abstract: found
            • Article: not found

            Global epidemiology of nonalcoholic fatty liver disease-Meta-analytic assessment of prevalence, incidence, and outcomes.

            Nonalcoholic fatty liver disease (NAFLD) is a major cause of liver disease worldwide. We estimated the global prevalence, incidence, progression, and outcomes of NAFLD and nonalcoholic steatohepatitis (NASH). PubMed/MEDLINE were searched from 1989 to 2015 for terms involving epidemiology and progression of NAFLD. Exclusions included selected groups (studies that exclusively enrolled morbidly obese or diabetics or pediatric) and no data on alcohol consumption or other liver diseases. Incidence of hepatocellular carcinoma (HCC), cirrhosis, overall mortality, and liver-related mortality were determined. NASH required histological diagnosis. All studies were reviewed by three independent investigators. Analysis was stratified by region, diagnostic technique, biopsy indication, and study population. We used random-effects models to provide point estimates (95% confidence interval [CI]) of prevalence, incidence, mortality and incidence rate ratios, and metaregression with subgroup analysis to account for heterogeneity. Of 729 studies, 86 were included with a sample size of 8,515,431 from 22 countries. Global prevalence of NAFLD is 25.24% (95% CI: 22.10-28.65) with highest prevalence in the Middle East and South America and lowest in Africa. Metabolic comorbidities associated with NAFLD included obesity (51.34%; 95% CI: 41.38-61.20), type 2 diabetes (22.51%; 95% CI: 17.92-27.89), hyperlipidemia (69.16%; 95% CI: 49.91-83.46%), hypertension (39.34%; 95% CI: 33.15-45.88), and metabolic syndrome (42.54%; 95% CI: 30.06-56.05). Fibrosis progression proportion, and mean annual rate of progression in NASH were 40.76% (95% CI: 34.69-47.13) and 0.09 (95% CI: 0.06-0.12). HCC incidence among NAFLD patients was 0.44 per 1,000 person-years (range, 0.29-0.66). Liver-specific mortality and overall mortality among NAFLD and NASH were 0.77 per 1,000 (range, 0.33-1.77) and 11.77 per 1,000 person-years (range, 7.10-19.53) and 15.44 per 1,000 (range, 11.72-20.34) and 25.56 per 1,000 person-years (range, 6.29-103.80). Incidence risk ratios for liver-specific and overall mortality for NAFLD were 1.94 (range, 1.28-2.92) and 1.05 (range, 0.70-1.56).
              Bookmark
              • Record: found
              • Abstract: found
              • Article: not found

              Genome engineering using the CRISPR-Cas9 system.

              Targeted nucleases are powerful tools for mediating genome alteration with high precision. The RNA-guided Cas9 nuclease from the microbial clustered regularly interspaced short palindromic repeats (CRISPR) adaptive immune system can be used to facilitate efficient genome engineering in eukaryotic cells by simply specifying a 20-nt targeting sequence within its guide RNA. Here we describe a set of tools for Cas9-mediated genome editing via nonhomologous end joining (NHEJ) or homology-directed repair (HDR) in mammalian cells, as well as generation of modified cell lines for downstream functional studies. To minimize off-target cleavage, we further describe a double-nicking strategy using the Cas9 nickase mutant with paired guide RNAs. This protocol provides experimentally derived guidelines for the selection of target sites, evaluation of cleavage efficiency and analysis of off-target activity. Beginning with target design, gene modifications can be achieved within as little as 1-2 weeks, and modified clonal cell lines can be derived within 2-3 weeks.
                Bookmark

                Author and article information

                Contributors
                Journal
                JHEP Rep
                JHEP Rep
                JHEP Reports
                Elsevier
                2589-5559
                23 April 2023
                August 2023
                23 April 2023
                : 5
                : 8
                : 100764
                Affiliations
                [1 ]National Institute of Health Research (NIHR) Nottingham Biomedical Research Centre, Nottingham University Hospitals NHS Trust & University of Nottingham, Nottingham, UK
                [2 ]Nottingham Digestive Diseases Centre, Translational Medical Sciences, School of Medicine, University of Nottingham, Nottingham, UK
                [3 ]Translational Medical Sciences, School of Medicine, University of Nottingham, Nottingham, UK
                [4 ]University of Nottingham Biodiscovery Institute, University of Nottingham, Nottingham, UK
                [5 ]Genetic Epidemiology Group, Department of Population Health Sciences, University of Leicester, Leicester, UK
                [6 ]Clinical Genetics Department, University Hospitals Leicester NHS Trust, Leicester, UK
                [7 ]NIHR Leicester Respiratory Biomedical Research Centre, Glenfield Hospital, Leicester, UK
                [8 ]School of Biosciences, University of Nottingham, Nottingham, UK
                [9 ]School of Veterinary Medicine and Science, University of Nottingham, Nottingham, UK
                [10 ]Leicester Cancer Research Centre, University of Leicester, Leicester, UK
                [11 ]Ulverscroft Eye Unit, Department of Neuroscience, Psychology and Behaviour, University of Leicester, Leicester, UK
                [12 ]Department of Chemical Pathology and Metabolic Diseases, University Hospitals of Leicester NHS Trust, Leicester, UK
                [13 ]Department of Cardiovascular Sciences, University of Leicester, Leicester, UK
                [14 ]Department of Gastroenterology, University Hospitals of Leicester NHS Trust, Leicester, UK
                [15 ]Department of Genetics and Genome Biology, University of Leicester, Leicester, UK
                [16 ]Population Health and Research Institute, Trivandrum, India
                Author notes
                []Corresponding author. Address: Nottingham Digestive Diseases Centre, University of Nottingham, Queens Medical Centre Campus, Nottingham NG7 2UH, UK. guru.aithal@ 123456nottingham.ac.uk
                [†]

                These authors share first authorship.

                [‡]

                These authors share senior authorship.

                [¶]

                Current institution: Treliske Hospital, Truro, Cornwall, UK.

                [§]

                Current institution: Department of Genetics, Nottingham University Hospitals NHS Trust, Nottingham, UK.

                [∗∗]

                Current institution: Department of Biosciences, School of Science and Technology, Nottingham Trent University, Clifton Campus, Nottingham, UK.

                Article
                S2589-5559(23)00095-2 100764
                10.1016/j.jhepr.2023.100764
                10362796
                37484212
                85a140a3-5754-495b-a8c6-e10ab8d1cbac
                © 2023 The Authors

                This is an open access article under the CC BY license (http://creativecommons.org/licenses/by/4.0/).

                History
                : 31 January 2023
                : 28 March 2023
                : 11 April 2023
                Categories
                Research Article

                microsomal triglyceride transfer protein,abetalipoproteinaemia,hipsc-derived hepatocytes,lipoprotein apob

                Comments

                Comment on this article