1
|
Henry Z, Janin A, Nony S, Marmontel O, Charrière S, Moulin P, Marrec M, Wargny M, Cariou B, Di Filippo M. Minigene splicing reporter assay: a high-stake tool for genetic diagnosis in familial hypobetalipoproteinemia. Atherosclerosis 2025; 405:119236. [PMID: 40367726 DOI: 10.1016/j.atherosclerosis.2025.119236] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/24/2025] [Revised: 04/20/2025] [Accepted: 05/01/2025] [Indexed: 05/16/2025]
Abstract
BACKGROUND & AIMS Familial hypobetalipoproteinemia 1 (FHBL-SD2) is the most common monogenic form of primary hypocholesterolaemia, related to truncating variants in the APOB gene encoding apolipoprotein B. Due to its high level of complexity, variants of uncertain significance (VUS) require further investigations. This study aims to demonstrate the value of setting minigene assays in the FHBL-SD2's genetic diagnosis. METHODS Four APOB VUS occurring in patients with a FHBL-SD2 phenotype were considered. In silico analysis were performed with six software programs supposed to predict the potential splicing effect. Then, functional consequences were studied in vitro using a minigene splicing reporter assay. RESULTS An effect on splicing was predicted in silico for the 4 variants, with the activation of a cryptic acceptor site for c.694-13A>G and c.1471-6A>G variants, and the use of a cryptic donor site for c.1123A>G and c.1470G>A variants. Minigene study showed a complete effect on splicing for 3 mutations, confirming the in silico predictions. All of these transcripts result in premature truncated variants. Therefore, these variants were reclassified as likely pathogenic and causative of FHBL-SD2. However, no effect was shown either in HeLA and HuH7 cells for the c.1470G>A variant. CONCLUSIONS Minigene study appears to be a promising and valuable tool to enhance the diagnostic accuracy of FHBL-SD2. It emphasizes the challenge in interpreting VUS and underscores the importance of establishing a clear strategy to assess their significance. Therefore, promoting minigene studies would be beneficial to understand precisely the impact of splicing variants.
Collapse
Affiliation(s)
- Zoé Henry
- Service de Biochimie et Biologie Moléculaire, Laboratoire de Biologie Médicale MultiSites, Hospices Civils de Lyon, F-69677, Bron, France; Hospices Civils de Lyon, Fédération d'endocrinologie, Maladies Métaboliques, diabète et Nutrition, Hôpital Louis Pradel, Bron, France; CarMen Laboratory, INSERM, INRAE, Université Claude Bernard Lyon 1, F-69310, Pierre-Bénite, France
| | - Alexandre Janin
- Service de Biochimie et Biologie Moléculaire, Laboratoire de Biologie Médicale MultiSites, Hospices Civils de Lyon, F-69677, Bron, France; CNRS UMR5261, INSERM U1315, Laboratoire Physiopathologie et Génétique du Neurone et du Muscle, Institut NeuroMyoGène, Université Claude Bernard Lyon 1, Université de Lyon, F-69008, Lyon, France
| | - Séverine Nony
- Service de Biochimie et Biologie Moléculaire, Laboratoire de Biologie Médicale MultiSites, Hospices Civils de Lyon, F-69677, Bron, France
| | - Oriane Marmontel
- Service de Biochimie et Biologie Moléculaire, Laboratoire de Biologie Médicale MultiSites, Hospices Civils de Lyon, F-69677, Bron, France; CarMen Laboratory, INSERM, INRAE, Université Claude Bernard Lyon 1, F-69310, Pierre-Bénite, France
| | - Sybil Charrière
- Hospices Civils de Lyon, Fédération d'endocrinologie, Maladies Métaboliques, diabète et Nutrition, Hôpital Louis Pradel, Bron, France; CarMen Laboratory, INSERM, INRAE, Université Claude Bernard Lyon 1, F-69310, Pierre-Bénite, France
| | - Philippe Moulin
- Hospices Civils de Lyon, Fédération d'endocrinologie, Maladies Métaboliques, diabète et Nutrition, Hôpital Louis Pradel, Bron, France; CarMen Laboratory, INSERM, INRAE, Université Claude Bernard Lyon 1, F-69310, Pierre-Bénite, France
| | - Marie Marrec
- Nantes Université, CHU Nantes, CNRS, Inserm, L'institut du Thorax, F-44000, Nantes, France
| | - Matthieu Wargny
- Nantes Université, CHU Nantes, CNRS, Inserm, L'institut du Thorax, F-44000, Nantes, France; Nantes Université, CHU Nantes, Pôle Hospitalo-Universitaire 11: Santé Publique, Clinique des données, INSERM, CIC 1413, F-44000, Nantes, France
| | - Bertrand Cariou
- Nantes Université, CHU Nantes, CNRS, Inserm, L'institut du Thorax, F-44000, Nantes, France
| | - Mathilde Di Filippo
- Service de Biochimie et Biologie Moléculaire, Laboratoire de Biologie Médicale MultiSites, Hospices Civils de Lyon, F-69677, Bron, France; CarMen Laboratory, INSERM, INRAE, Université Claude Bernard Lyon 1, F-69310, Pierre-Bénite, France.
| |
Collapse
|
2
|
Wang JJ, Chen XY, Zhang YR, Shen Y, Zhu ML, Zhang J, Zhang JJ. Role of genetic variants and DNA methylation of lipid metabolism-related genes in metabolic dysfunction-associated steatotic liver disease. Front Physiol 2025; 16:1562848. [PMID: 40166716 PMCID: PMC11955510 DOI: 10.3389/fphys.2025.1562848] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2025] [Accepted: 02/25/2025] [Indexed: 04/02/2025] Open
Abstract
Metabolic dysfunction-associated steatotic liver disease (MASLD), is one of the most common chronic liver diseases, which encompasses a spectrum of diseases, from metabolic dysfunction-associated steatotic liver (MASL) to metabolic dysfunction-associated steatohepatitis (MASH), and may ultimately progress to MASH-related cirrhosis and hepatocellular carcinoma (HCC). MASLD is a complex disease that is influenced by genetic and environmental factors. Dysregulation of hepatic lipid metabolism plays a crucial role in the development and progression of MASLD. Therefore, the focus of this review is to discuss the links between the genetic variants and DNA methylation of lipid metabolism-related genes and MASLD pathogenesis. We first summarize the interplay between MASLD and the disturbance of hepatic lipid metabolism. Next, we focus on reviewing the role of hepatic lipid related gene loci in the onset and progression of MASLD. We summarize the existing literature around the single nucleotide polymorphisms (SNPs) associated with MASLD identified by genome-wide association studies (GWAS) and candidate gene analyses. Moreover, based on recent evidence from human and animal studies, we further discussed the regulatory function and associated mechanisms of changes in DNA methylation levels in the occurrence and progression of MASLD, with a particular emphasis on its regulatory role of lipid metabolism-related genes in MASLD and MASH. Furthermore, we review the alterations of hepatic DNA and blood DNA methylation levels associated with lipid metabolism-related genes in MASLD and MASH patients. Finally, we introduce potential value of the genetic variants and DNA methylation profiles of lipid metabolism-related genes in developing novel prognostic biomarkers and therapeutic targets for MASLD, intending to provide references for the future studies of MASLD.
Collapse
Affiliation(s)
- Jun-Jie Wang
- Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Diseases, Ministry of Education, Department of Basic Medicine, Gannan Medical University, Ganzhou, China
| | - Xiao-Yuan Chen
- Department of Publication Health and Health Management, Gannan Medical University, Ganzhou, China
| | - Yi-Rong Zhang
- Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Diseases, Ministry of Education, Department of Basic Medicine, Gannan Medical University, Ganzhou, China
| | - Yan Shen
- Department of Publication Health and Health Management, Gannan Medical University, Ganzhou, China
| | - Meng-Lin Zhu
- Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Diseases, Ministry of Education, Department of Basic Medicine, Gannan Medical University, Ganzhou, China
| | - Jun Zhang
- Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Diseases, Ministry of Education, Department of Basic Medicine, Gannan Medical University, Ganzhou, China
| | - Jun-Jie Zhang
- Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Diseases, Ministry of Education, Department of Basic Medicine, Gannan Medical University, Ganzhou, China
| |
Collapse
|
3
|
Chen R, Petrazzini BO, Duffy Á, Rocheleau G, Jordan D, Bansal M, Do R. Trans-ancestral rare variant association study with machine learning-based phenotyping for metabolic dysfunction-associated steatotic liver disease. Genome Biol 2025; 26:50. [PMID: 40065360 PMCID: PMC11892324 DOI: 10.1186/s13059-025-03518-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2024] [Accepted: 02/26/2025] [Indexed: 03/14/2025] Open
Abstract
BACKGROUND Genome-wide association studies (GWAS) have identified common variants associated with metabolic dysfunction-associated steatotic liver disease (MASLD). However, rare coding variant studies have been limited by phenotyping challenges and small sample sizes. We test associations of rare and ultra-rare coding variants with proton density fat fraction (PDFF) and MASLD case-control status in 736,010 participants of diverse ancestries from the UK Biobank, All of Us, and BioMe and performed a trans-ancestral meta-analysis. We then developed models to accurately predict PDFF and MASLD status in the UK Biobank and tested associations with these predicted phenotypes to increase statistical power. RESULTS The trans-ancestral meta-analysis with PDFF and MASLD case-control status identifies two single variants and two gene-level associations in APOB, CDH5, MYCBP2, and XAB2. Association testing with predicted phenotypes, which replicates more known genetic variants from GWAS than true phenotypes, identifies 16 single variants and 11 gene-level associations implicating 23 additional genes. Two variants were polymorphic only among African ancestry participants and several associations showed significant heterogeneity in ancestry and sex-stratified analyses. In total, we identified 27 genes, of which 3 are monogenic causes of steatosis (APOB, G6PC1, PPARG), 4 were previously associated with MASLD (APOB, APOC3, INSR, PPARG), and 23 had supporting clinical, experimental, and/or genetic evidence. CONCLUSIONS Our results suggest that trans-ancestral association analyses can identify ancestry-specific rare and ultra-rare coding variants in MASLD pathogenesis. Furthermore, we demonstrate the utility of machine learning in genetic investigations of difficult-to-phenotype diseases in trans-ancestral biobanks.
Collapse
Affiliation(s)
- Robert Chen
- The Charles Bronfman Institute for Personalized Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Medical Scientist Training Program, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Ben Omega Petrazzini
- The Charles Bronfman Institute for Personalized Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Center for Genomic Data Analytics, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Áine Duffy
- The Charles Bronfman Institute for Personalized Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Center for Genomic Data Analytics, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Ghislain Rocheleau
- The Charles Bronfman Institute for Personalized Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Center for Genomic Data Analytics, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Daniel Jordan
- The Charles Bronfman Institute for Personalized Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Center for Genomic Data Analytics, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Meena Bansal
- The Charles Bronfman Institute for Personalized Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Division of Liver Diseases, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Ron Do
- The Charles Bronfman Institute for Personalized Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- Center for Genomic Data Analytics, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| |
Collapse
|
4
|
Fan YH, Zhang S, Wang Y, Wang H, Li H, Bai L. Inter-organ metabolic interaction networks in non-alcoholic fatty liver disease. Front Endocrinol (Lausanne) 2025; 15:1494560. [PMID: 39850476 PMCID: PMC11754069 DOI: 10.3389/fendo.2024.1494560] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/11/2024] [Accepted: 12/16/2024] [Indexed: 01/25/2025] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) is a multisystem metabolic disorder, marked by abnormal lipid accumulation and intricate inter-organ interactions, which contribute to systemic metabolic imbalances. NAFLD may progress through several stages, including simple steatosis (NAFL), non-alcoholic steatohepatitis (NASH), cirrhosis, and potentially liver cancer. This disease is closely associated with metabolic disorders driven by overnutrition, with key pathological processes including lipid dysregulation, impaired lipid autophagy, mitochondrial dysfunction, endoplasmic reticulum (ER) stress, and local inflammation. While hepatic lipid metabolism in NAFLD is well-documented, further research into inter-organ communication mechanisms is crucial for a deeper understanding of NAFLD progression. This review delves into intrahepatic networks and tissue-specific signaling mediators involved in NAFLD pathogenesis, emphasizing their impact on distal organs.
Collapse
Affiliation(s)
- Yu-Hong Fan
- State Key Laboratory of New Targets Discovery and Drug Development for Major Diseases, Gannan Innovation and Translational Medicine Research Institute, Gannan Medical University, Ganzhou, China
| | - Siyao Zhang
- State Key Laboratory of New Targets Discovery and Drug Development for Major Diseases, Gannan Innovation and Translational Medicine Research Institute, Gannan Medical University, Ganzhou, China
| | - Ye Wang
- State Key Laboratory of New Targets Discovery and Drug Development for Major Diseases, Gannan Innovation and Translational Medicine Research Institute, Gannan Medical University, Ganzhou, China
| | - Hongni Wang
- State Key Laboratory of New Targets Discovery and Drug Development for Major Diseases, Gannan Innovation and Translational Medicine Research Institute, Gannan Medical University, Ganzhou, China
| | - Hongliang Li
- State Key Laboratory of New Targets Discovery and Drug Development for Major Diseases, Gannan Innovation and Translational Medicine Research Institute, Ganzhou, China
- Department of Cardiology, Renmin Hospital, Wuhan University, Wuhan, China
- Medical Science Research Center, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Lan Bai
- State Key Laboratory of New Targets Discovery and Drug Development for Major Diseases, Gannan Innovation and Translational Medicine Research Institute, Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Diseases, Ministry of Education, Gannan Medical University, Ganzhou, China
| |
Collapse
|
5
|
Sasaki K, Tada H, Komatsu T, Terada H, Endo Y, Ikewaki K, Uehara Y. A New Case of Abetalipoproteinemia Caused by Novel Compound Heterozygote Mutations in the MTTP Gene without Fat or Vitamin Malabsorption. J Atheroscler Thromb 2024; 31:1634-1640. [PMID: 38749717 PMCID: PMC11537785 DOI: 10.5551/jat.64730] [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: 11/22/2023] [Accepted: 03/24/2024] [Indexed: 11/06/2024] Open
Abstract
Abetalipoproteinemia (ABL) is a rare disease characterized by extremely low apolipoprotein B (apoB)-containing lipoprotein levels, dietary fat, and fat-soluble vitamin malabsorption, leading to gastrointestinal, neuromuscular, and ophthalmological symptoms. We herein report a case of ABL with novel compound heterozygous mutations in the microsomal triglyceride transfer protein gene (c.1686_1687del [p.Ser563TyrfsTer10] and c.1862T>C [p.Ile621Thr]), identified via panel sequencing. Although the patient had extremely reduced low-density lipoprotein cholesterol levels and a fatty liver, he did not exhibit other typical complications. Furthermore, unlike typical ABL, this patient had a preserved apoB-48 secretion and increased concentrations of high-density lipoprotein cholesterol, which may account for the normal serum fat-soluble vitamin levels.
Collapse
Affiliation(s)
- Kei Sasaki
- Center for Preventive, Anti-aging and Regenerative Medicine, Fukuoka University Hospital, Fukuoka, Japan
- Division of Anti-aging and Vascular Medicine, Department of Internal Medicine, National Defense Medical College, Tokorozawa, Japan
- Department of Internal Medicine, Self-Defense Forces Fukuoka Hospital, Kasuga, Japan
| | - Hayato Tada
- Department of Cardiology, Kanazawa University Graduate School of Medicine, Kanazawa, Japan
| | - Tomohiro Komatsu
- Center for Preventive, Anti-aging and Regenerative Medicine, Fukuoka University Hospital, Fukuoka, Japan
| | - Hisato Terada
- Department of Internal Medicine, Self-Defense Forces Fukuoka Hospital, Kasuga, Japan
| | - Yasuhiro Endo
- Division of Anti-aging and Vascular Medicine, Department of Internal Medicine, National Defense Medical College, Tokorozawa, Japan
| | - Katsunori Ikewaki
- Division of Anti-aging and Vascular Medicine, Department of Internal Medicine, National Defense Medical College, Tokorozawa, Japan
| | - Yoshinari Uehara
- Center for Preventive, Anti-aging and Regenerative Medicine, Fukuoka University Hospital, Fukuoka, Japan
| |
Collapse
|
6
|
Makhmudova U, Steinhagen-Thiessen E, Volpe M, Landmesser U. Advances in nucleic acid-targeted therapies for cardiovascular disease prevention. Cardiovasc Res 2024; 120:1107-1125. [PMID: 38970537 DOI: 10.1093/cvr/cvae136] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Revised: 04/14/2024] [Accepted: 05/03/2024] [Indexed: 07/08/2024] Open
Abstract
Nucleic acid-based therapies are being rapidly developed for prevention and management of cardiovascular diseases (CVD). Remarkable advancements have been achieved in the delivery, safety, and effectiveness of these therapeutics in the past decade. These therapies can also modulate therapeutic targets that cannot be sufficiently addressed using traditional drugs or antibodies. Among the nucleic acid-targeted therapeutics under development for CVD prevention are RNA-targeted approaches, including antisense oligonucleotides (ASO), small interfering RNAs (siRNA), and novel genome editing techniques. Genetic studies have identified potential therapeutic targets that are suggested to play a causative role in development and progression of CVD. RNA- and DNA-targeted therapeutics can be particularly well delivered to the liver, where atherogenic lipoproteins and angiotensinogen (AGT) are produced. Current targets in lipid metabolism include proprotein convertase subtilisin/kexin type 9 (PCSK9), apolipoprotein A (ApoA), apolipoprotein C3 (ApoC3), angiopoietin-like 3 (ANGPTL3). Several large-scale clinical development programs for nucleic acid-targeted therapies in cardiovascular prevention are under way, which may also be attractive from a therapy adherence point of view, given the long action of these therapeutics. In addition to genome editing, the concept of gene transfer is presently under assessment in preclinical and clinical investigations as a potential approach for addressing low-density lipoprotein receptor deficiency. Furthermore, ongoing research is exploring the use of RNA-targeted therapies to treat arterial hypertension by reducing hepatic angiotensinogen (AGT) production. This review summarizes the rapid translation of siRNA and ASO therapeutics as well as gene editing into clinical studies to treat dyslipidemia and arterial hypertension for CVD prevention. It also outlines potential innovative therapeutic options that are likely relevant to the future of cardiovascular medicine.
Collapse
Affiliation(s)
- Umidakhon Makhmudova
- Department of Cardiology, Angiology, and Intensive Care Medicine, Deutsches Herzzentrum der Charité, Berlin, Hindenburgdamm 30, 12203 Berlin, Germany
- Friede Springer Cardiovascular Prevention Center at Charité, Hindenburgdamm 30, 12203 Berlin, Germany
- Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Klinik/Centrum, Charitéplatz 1, 10117 Berlin, Germany
| | - Elisabeth Steinhagen-Thiessen
- Friede Springer Cardiovascular Prevention Center at Charité, Hindenburgdamm 30, 12203 Berlin, Germany
- Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Klinik/Centrum, Charitéplatz 1, 10117 Berlin, Germany
- Department of Endocrinology and Metabolic Diseases, Charite Universitätsmedizin Berlin, Augustenburger Platz 1, 13353 Berlin, Germany
| | - Massimo Volpe
- Department of Clinical and Molecular Medicine, Sapienza University of Rome, Via di Grottarossa 1035-1039, Rome 00189, Italy
- Cardiology Department, IRCCS San Raffaele Roma, Via di Valcannuta 250, Rome 00166, Italy
| | - Ulf Landmesser
- Department of Cardiology, Angiology, and Intensive Care Medicine, Deutsches Herzzentrum der Charité, Berlin, Hindenburgdamm 30, 12203 Berlin, Germany
- Friede Springer Cardiovascular Prevention Center at Charité, Hindenburgdamm 30, 12203 Berlin, Germany
- Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Klinik/Centrum, Charitéplatz 1, 10117 Berlin, Germany
- Berlin Institute of Health (BIH), Charité - Universitätsmedizin Berlin, Charitéplatz 1, 10117 Berlin, Germany
- German Centre for Cardiovascular Research, DZHK, Partner Site Berlin, Germany
| |
Collapse
|
7
|
Guay SP, Paquette M, Girard L, Desgagné V, Gosse G, Poulin V, Bouchard L, Baass A. High carrier frequency for abetalipoproteinemia and evidence of a founder variant in a French-Canadian population. J Clin Lipidol 2024; 18:e625-e630. [PMID: 38908974 DOI: 10.1016/j.jacl.2024.04.132] [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: 02/20/2024] [Revised: 04/22/2024] [Accepted: 04/23/2024] [Indexed: 06/24/2024]
Abstract
Abetalipoproteinemia (ABL) is a rare recessive genetic disease caused by bi-allelic pathogenic variants in the microsomal triglyceride transfer protein (MTTP) gene. This disease is characterized by a deficiency in the secretion of apolipoprotein B-containing lipoproteins. Patients with ABL present with neurological, hematological, and gastrointestinal symptoms due to fat malabsorption and a deficiency in liposoluble vitamins. In this report, we present a total of four ABL cases, including three new cases, all originating from the same French-Canadian founder population in Saguenay-Lac-Saint-Jean, Québec, Canada. These individuals are homozygous for the same pathogenic variant in the MTTP gene (c.419dup, p.Asn140Lysfs*2). We found that this variant is more common than anticipated in this population, with an estimated carrier frequency of 1:203. Early diagnosis is essential to initiate treatment known to prevent complications associated with ABL. Population carrier screening or newborn screening for ABL should be considered in this French-Canadian founder population.
Collapse
Affiliation(s)
- Simon-Pierre Guay
- Genetic Dyslipidemias Clinic of the Montreal Clinical Research Institute, Montréal, Québec, Canada (Drs Guay, Paquette, Gosse, Poulin, and Baass); Department of Medicine, Division of Endocrinology, Université de Montréal, Montréal, Québec, Canada (Dr Guay); Department of Pediatrics, Division of Medical Genetics, Université de Sherbrooke, Québec, Canada (Dr Guay)
| | - Martine Paquette
- Genetic Dyslipidemias Clinic of the Montreal Clinical Research Institute, Montréal, Québec, Canada (Drs Guay, Paquette, Gosse, Poulin, and Baass)
| | - Lysanne Girard
- Department of Biochemistry and Functional Genomics, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, Québec, Canada (Drs Girard, Desgagné, and Bouchard)
| | - Véronique Desgagné
- Department of Biochemistry and Functional Genomics, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, Québec, Canada (Drs Girard, Desgagné, and Bouchard); Clinical Department of Laboratory Medicine, Centre intégré universitaire de santé et de services sociaux (CIUSSS) du Saguenay-Lac-Saint-Jean - Hôpital de Chicoutimi, Saguenay, Québec, Canada (Drs Desgagné, Bouchard, and Baass)
| | - Géraldine Gosse
- Genetic Dyslipidemias Clinic of the Montreal Clinical Research Institute, Montréal, Québec, Canada (Drs Guay, Paquette, Gosse, Poulin, and Baass)
| | - Valérie Poulin
- Genetic Dyslipidemias Clinic of the Montreal Clinical Research Institute, Montréal, Québec, Canada (Drs Guay, Paquette, Gosse, Poulin, and Baass)
| | - Luigi Bouchard
- Department of Biochemistry and Functional Genomics, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, Québec, Canada (Drs Girard, Desgagné, and Bouchard); Clinical Department of Laboratory Medicine, Centre intégré universitaire de santé et de services sociaux (CIUSSS) du Saguenay-Lac-Saint-Jean - Hôpital de Chicoutimi, Saguenay, Québec, Canada (Drs Desgagné, Bouchard, and Baass)
| | - Alexis Baass
- Genetic Dyslipidemias Clinic of the Montreal Clinical Research Institute, Montréal, Québec, Canada (Drs Guay, Paquette, Gosse, Poulin, and Baass); Department of Medicine, Divisions of Experimental Medicine and Medical Biochemistry, McGill University, Montréal, Québec, Canada (Dr Baass).
| |
Collapse
|
8
|
Wakabayashi T, Takahashi M, Okazaki H, Okazaki S, Yokote K, Tada H, Ogura M, Ishigaki Y, Yamashita S, Harada-Shiba M, on behalf of the Committee on Primary Dyslipidemia under the Research Program on Rare and Intractable Disease of the Ministry of Health, Labour and Welfare of Japan. Current Diagnosis and Management of Familial Hypobetalipoproteinemia 1. J Atheroscler Thromb 2024; 31:1005-1023. [PMID: 38710625 PMCID: PMC11224688 DOI: 10.5551/jat.rv22018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Accepted: 03/28/2024] [Indexed: 05/08/2024] Open
Abstract
Familial hypobetalipoproteinemia (FHBL) 1 is a rare genetic disorder with an autosomal codominant mode of inheritance and is caused by defects in the apolipoprotein (apo) B (APOB) gene that disable lipoprotein formation. ApoB proteins are required for the formation of very low-density lipoproteins (VLDLs), chylomicrons, and their metabolites. VLDLs transport cholesterol and triglycerides from the liver to the peripheral tissues, whereas chylomicrons transport absorbed lipids and fat-soluble vitamins from the intestine. Homozygous or compound heterozygotes of FHBL1 (HoFHBL1) are extremely rare, and defects in APOB impair VLDL and chylomicron secretion, which result in marked hypolipidemia with malabsorption of fat and fat-soluble vitamins, leading to various complications such as growth disorders, acanthocytosis, retinitis pigmentosa, and neuropathy. Heterozygotes of FHBL1 are relatively common and are generally asymptomatic, except for moderate hypolipidemia and possible hepatic steatosis. If left untreated, HoFHBL1 can cause severe complications and disabilities that are pathologically and phenotypically similar to abetalipoproteinemia (ABL) (an autosomal recessive disorder) caused by mutations in the microsomal triglyceride transfer protein (MTTP) gene. Although HoFHBL1 and ABL cannot be distinguished from the clinical manifestations and laboratory findings of the proband, moderate hypolipidemia in first-degree relatives may help diagnose HoFHBL1. There is currently no specific treatment for HoFHBL1. Palliative therapy including high-dose fat-soluble vitamin supplementation may prevent or delay complications. Registry research on HoFHBL1 is currently ongoing to better understand the disease burden and unmet needs of this life-threatening disease with few therapeutic options.
Collapse
Affiliation(s)
- Tetsuji Wakabayashi
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Jichi Medical University, Tochigi, Japan
| | - Manabu Takahashi
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Jichi Medical University, Tochigi, Japan
| | - Hiroaki Okazaki
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Jichi Medical University, Tochigi, Japan
| | - Sachiko Okazaki
- Division for Health Service Promotion, The University of Tokyo, Tokyo, Japan
| | | | - Hayato Tada
- Department of Cardiovascular Medicine, Kanazawa University Graduate School of Medical Sciences, Ishikawa, Japan
| | - Masatsune Ogura
- Department of Clinical Laboratory Technology, Faculty of Medical Science, Juntendo University, Tokyo, Japan
| | - Yasushi Ishigaki
- Division of Diabetes, Metabolism and Endocrinology, Department of Internal Medicine, Iwate Medical University, Iwate, Japan
| | - Shizuya Yamashita
- Department of Cardiology, Rinku General Medical Center, Osaka, Japan
| | - Mariko Harada-Shiba
- Cardiovascular Center, Osaka Medical and Pharmaceutical University, Osaka, Japan
| | - on behalf of the Committee on Primary Dyslipidemia under the Research Program on Rare and Intractable Disease of the Ministry of Health, Labour and Welfare of Japan
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Jichi Medical University, Tochigi, Japan
- Division for Health Service Promotion, The University of Tokyo, Tokyo, Japan
- Chiba University, Chiba, Japan
- Department of Cardiovascular Medicine, Kanazawa University Graduate School of Medical Sciences, Ishikawa, Japan
- Department of Clinical Laboratory Technology, Faculty of Medical Science, Juntendo University, Tokyo, Japan
- Division of Diabetes, Metabolism and Endocrinology, Department of Internal Medicine, Iwate Medical University, Iwate, Japan
- Department of Cardiology, Rinku General Medical Center, Osaka, Japan
- Cardiovascular Center, Osaka Medical and Pharmaceutical University, Osaka, Japan
| |
Collapse
|
9
|
Koenig AB, Tan A, Abdelaal H, Monge F, Younossi ZM, Goodman ZD. Review article: Hepatic steatosis and its associations with acute and chronic liver diseases. Aliment Pharmacol Ther 2024; 60:167-200. [PMID: 38845486 DOI: 10.1111/apt.18059] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/04/2024] [Revised: 04/23/2024] [Accepted: 05/13/2024] [Indexed: 06/28/2024]
Abstract
BACKGROUND Hepatic steatosis is a common finding in liver histopathology and the hallmark of metabolic dysfunction-associated steatotic liver disease (MASLD), formerly known as non-alcoholic fatty liver disease (NAFLD), whose global prevalence is rising. AIMS To review the histopathology of hepatic steatosis and its mechanisms of development and to identify common and rare disease associations. METHODS We reviewed literature on the basic science of lipid droplet (LD) biology and clinical research on acute and chronic liver diseases associated with hepatic steatosis using the PubMed database. RESULTS A variety of genetic and environmental factors contribute to the development of chronic hepatic steatosis or steatotic liver disease, which typically appears macrovesicular. Microvesicular steatosis is associated with acute mitochondrial dysfunction and liver failure. Fat metabolic processes in hepatocytes whose dysregulation leads to the development of steatosis include secretion of lipoprotein particles, uptake of remnant lipoprotein particles or free fatty acids from blood, de novo lipogenesis, oxidation of fatty acids, lipolysis and lipophagy. Hepatic insulin resistance is a key feature of MASLD. Seipin is a polyfunctional protein that facilitates LD biogenesis. Assembly of hepatitis C virus takes place on LD surfaces. LDs make important, functional contact with the endoplasmic reticulum and other organelles. CONCLUSIONS Diverse liver pathologies are associated with hepatic steatosis, with MASLD being the most important contributor. The biogenesis and dynamics of LDs in hepatocytes are complex and warrant further investigation. Organellar interfaces permit co-regulation of lipid metabolism to match generation of potentially toxic lipid species with their LD depot storage.
Collapse
Affiliation(s)
- Aaron B Koenig
- Beatty Liver and Obesity Research Program, Inova Health System, Falls Church, Virginia, USA
| | - Albert Tan
- Beatty Liver and Obesity Research Program, Inova Health System, Falls Church, Virginia, USA
- Center for Liver Diseases, Inova Fairfax Hospital, Falls Church, Virginia, USA
| | - Hala Abdelaal
- Beatty Liver and Obesity Research Program, Inova Health System, Falls Church, Virginia, USA
- Center for Liver Diseases, Inova Fairfax Hospital, Falls Church, Virginia, USA
| | - Fanny Monge
- Beatty Liver and Obesity Research Program, Inova Health System, Falls Church, Virginia, USA
- Center for Liver Diseases, Inova Fairfax Hospital, Falls Church, Virginia, USA
| | - Zobair M Younossi
- Beatty Liver and Obesity Research Program, Inova Health System, Falls Church, Virginia, USA
- The Global NASH Council, Center for Outcomes Research in Liver Diseases, Washington, DC, USA
| | - Zachary D Goodman
- Beatty Liver and Obesity Research Program, Inova Health System, Falls Church, Virginia, USA
- Center for Liver Diseases, Inova Fairfax Hospital, Falls Church, Virginia, USA
| |
Collapse
|
10
|
Chouik Y, Di Filippo M, Radenne S, Dumortier J, Moulin P, Levrero M. Combination of heterozygous APOB gene mutation with PNPLA3 and TM6SF2 variants promotes steatotic liver disease, cirrhosis and HCC development. Liver Int 2024; 44:1474-1477. [PMID: 38421084 DOI: 10.1111/liv.15837] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/03/2023] [Revised: 12/15/2023] [Accepted: 12/26/2023] [Indexed: 03/02/2024]
Affiliation(s)
- Yasmina Chouik
- INSERM U1052, CNRS UMR_5286, Cancer Research Center of Lyon, Lyon, France
- University of Lyon, Université Claude-Bernard 1, UMR_S1052, Lyon, France
- Institute of Hepatology Lyon, Lyon, France
- Department of Hepatology, Hôpital Croix-Rousse, Hospices Civils de Lyon, Lyon, France
| | - Mathilde Di Filippo
- Department of Biochemistry and Molecular Biology, Hospices Civils de Lyon, Lyon, France
- CarMeN Laboratory, UMR INSERM U1060/INRAE U1397, Claude Bernard Lyon1 University, Bron, France
| | - Sylvie Radenne
- Department of Hepatology, Hôpital Croix-Rousse, Hospices Civils de Lyon, Lyon, France
| | - Jérôme Dumortier
- INSERM U1052, CNRS UMR_5286, Cancer Research Center of Lyon, Lyon, France
- University of Lyon, Université Claude-Bernard 1, UMR_S1052, Lyon, France
- Institute of Hepatology Lyon, Lyon, France
- Department of Hepatology, Hospices Civils de Lyon, Hôpital Universitaire Edouard Herriot, Lyon, France
| | - Philippe Moulin
- CarMeN Laboratory, UMR INSERM U1060/INRAE U1397, Claude Bernard Lyon1 University, Bron, France
- Department of Endocrinology, Metabolic Disease, Diabetes and Nutrition, Hospices Civils de Lyon, Lyon, France
| | - Massimo Levrero
- INSERM U1052, CNRS UMR_5286, Cancer Research Center of Lyon, Lyon, France
- University of Lyon, Université Claude-Bernard 1, UMR_S1052, Lyon, France
- Institute of Hepatology Lyon, Lyon, France
- Department of Hepatology, Hôpital Croix-Rousse, Hospices Civils de Lyon, Lyon, France
- Department of Medicine SCIAC, University of Rome La Sapienza, Rome, Italy
- The Italian Institute of Technology (IIT) Center for Life Nanosciences (CLNS), University of Rome La Sapienza, Rome, Italy
| |
Collapse
|
11
|
Newberry EP, Molitor EA, Liu A, Chong K, Liu X, Alonso C, Mato JM, Davidson NO. Impaired Hepatic Very Low-Density Lipoprotein Secretion Promotes Tumorigenesis and Is Accelerated with Fabp1 Deletion. THE AMERICAN JOURNAL OF PATHOLOGY 2024; 194:958-974. [PMID: 38417694 PMCID: PMC11156158 DOI: 10.1016/j.ajpath.2024.02.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/26/2023] [Revised: 01/17/2024] [Accepted: 02/09/2024] [Indexed: 03/01/2024]
Abstract
Genetic polymorphisms that impair very low-density lipoprotein (VLDL) secretion are linked to hepatic steatosis, fibrosis, and hepatocellular cancer. Liver-specific deletion of microsomal triglyceride transfer protein (Mttp-LKO) impairs VLDL assembly, promoting hepatic steatosis and fibrosis, which are attenuated in Mttp-LKO X Fabp1-null [Fabp1/Mttp double knockout (DKO)] mice. The current study examined the impact of impaired VLDL secretion in Mttp-LKO mice on hepatocellular cancer incidence and progression in comparison to Fabp1/Mttp DKO mice. Diethylnitrosamine-treated Mttp-LKO mice exhibited steatosis with increased tumor burden compared with flox controls, whereas diethylnitrosamine-treated Fabp1/Mttp DKO mice exhibited a paradoxical increase in tumor burden and >50% mortality by 50 weeks. Serum high-density lipoprotein cholesterol was elevated in both Mttp-LKO and Fabp1/Mttp DKO mice, with increased intratumoral expression of apolipoprotein A1 and apolipoprotein E. Lipidomic surveys revealed progressive enrichment in distinct triglyceride species in livers from Mttp-LKO mice with further enrichment in Fabp1/Mttp DKO mice. RNA sequencing revealed mRNA changes suggesting altered monocarboxylic acid use and increased aerobic glycolysis, whereas hepatocytes from Fabp1/Mttp DKO mice exhibited increased capacity to use glucose and glutamine. These metabolic shifts were accompanied by reduced expression of HNF1 homeobox A (HNF1a), which correlated with tumor burden. Taken together, these findings demonstrate that hepatic tumorigenesis is increased in mice with impaired VLDL secretion and further accelerated via pathways including altered fatty acid compartmentalization and shifts in hepatic energy use.
Collapse
Affiliation(s)
- Elizabeth P Newberry
- Department of Medicine, Washington University School of Medicine, St. Louis, Missouri
| | - Elizabeth A Molitor
- Department of Medicine, Washington University School of Medicine, St. Louis, Missouri
| | - Allen Liu
- Department of Medicine, Washington University School of Medicine, St. Louis, Missouri
| | - Kamyar Chong
- Department of Medicine, Washington University School of Medicine, St. Louis, Missouri
| | - Xiuli Liu
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, Missouri
| | - Cristina Alonso
- OWL Metabolomics, Parque Tecnológico de Bizkaia, Derio, Spain
| | - Jose M Mato
- CIC bioGUNE, Parque Tecnológico de Bizkaia, Derio, Spain
| | - Nicholas O Davidson
- Department of Medicine, Washington University School of Medicine, St. Louis, Missouri.
| |
Collapse
|
12
|
Moll TO, Klemek ML, Farber SA. Directly Measuring Atherogenic Lipoprotein Kinetics in Zebrafish with the Photoconvertible LipoTimer Reporter. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.29.596423. [PMID: 38853962 PMCID: PMC11160697 DOI: 10.1101/2024.05.29.596423] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2024]
Abstract
Lipoprotein kinetics are a crucial factor in understanding lipoprotein metabolism since a prolonged time in circulation can contribute to the atherogenic character of apolipoprotein-B (ApoB)-containing lipoproteins (B-lps). Here, we report a method to directly measure lipoprotein kinetics in live developing animals. We developed a zebrafish geneticly encoded reporter, LipoTimer, in which endogenous ApoBb.1 is fused to the photoconvertible fluorophore Dendra2 which shift its emission profile from green to red upon UV exposure. By quantifying the red population of ApoB-Dendra2 over time, we found that B-lp turnover in wild-type larvae becomes faster as development proceeds. Mutants with impaired B-lp uptake or lipolysis present with increased B-lp levels and half-life. In contrast, mutants with impaired B-lp triglyceride loading display slightly fewer and smaller-B-lps, which have a significantly shorter B-lp half-life. Further, we showed that chronic high-cholesterol feeding is associated with a longer B-lp half-life in wild-type juveniles but does not lead to changes in B-lp half-life in lipolysis deficient apoC2 mutants. These data support the hypothesis that B-lp lipolysis is suppressed by the flood of intestinal-derived B-lps that follow a high-fat meal.
Collapse
Affiliation(s)
- Tabea O.C. Moll
- Johns Hopkins University, Baltimore, Maryland, United States of America
| | | | - Steven A. Farber
- Johns Hopkins University, Baltimore, Maryland, United States of America
| |
Collapse
|
13
|
Zeng T, Lv J, Liang J, Xie B, Liu L, Tan Y, Zhu J, Jiang J, Xie H. Zebrafish cobll1a regulates lipid homeostasis via the RA signaling pathway. Front Cell Dev Biol 2024; 12:1381362. [PMID: 38699158 PMCID: PMC11063382 DOI: 10.3389/fcell.2024.1381362] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2024] [Accepted: 04/04/2024] [Indexed: 05/05/2024] Open
Abstract
Background The COBLL1 gene has been implicated in human central obesity, fasting insulin levels, type 2 diabetes, and blood lipid profiles. However, its molecular mechanisms remain largely unexplored. Methods In this study, we established cobll1a mutant lines using the CRISPR/Cas9-mediated gene knockout technique. To further dissect the molecular underpinnings of cobll1a during early development, transcriptome sequencing and bioinformatics analysis was employed. Results Our study showed that compared to the control, cobll1a -/- zebrafish embryos exhibited impaired development of digestive organs, including the liver, intestine, and pancreas, at 4 days post-fertilization (dpf). Transcriptome sequencing and bioinformatics analysis results showed that in cobll1a knockout group, the expression level of genes in the Retinoic Acid (RA) signaling pathway was affected, and the expression level of lipid metabolism-related genes (fasn, scd, elovl2, elovl6, dgat1a, srebf1 and srebf2) were significantly changed (p < 0.01), leading to increased lipid synthesis and decreased lipid catabolism. The expression level of apolipoprotein genes (apoa1a, apoa1b, apoa2, apoa4a, apoa4b, and apoea) genes were downregulated. Conclusion Our study suggest that the loss of cobll1a resulted in disrupted RA metabolism, reduced lipoprotein expression, and abnormal lipid transport, therefore contributing to lipid accumulation and deleterious effects on early liver development.
Collapse
Affiliation(s)
- Ting Zeng
- Hunan International Joint Laboratory of Animal Intestinal Ecology and Health, Laboratory of Animal Nutrition and Human Health, College of Life Sciences, Hunan Normal University, Changsha, Hunan, China
- Hunan Provincial Key Laboratory of Animal Intestinal Function and Regulation, Changsha, Hunan, China
| | - Jinrui Lv
- Hunan International Joint Laboratory of Animal Intestinal Ecology and Health, Laboratory of Animal Nutrition and Human Health, College of Life Sciences, Hunan Normal University, Changsha, Hunan, China
- Hunan Provincial Key Laboratory of Animal Intestinal Function and Regulation, Changsha, Hunan, China
| | - Jiaxin Liang
- Hunan International Joint Laboratory of Animal Intestinal Ecology and Health, Laboratory of Animal Nutrition and Human Health, College of Life Sciences, Hunan Normal University, Changsha, Hunan, China
- Hunan Provincial Key Laboratory of Animal Intestinal Function and Regulation, Changsha, Hunan, China
| | - Binling Xie
- Hunan International Joint Laboratory of Animal Intestinal Ecology and Health, Laboratory of Animal Nutrition and Human Health, College of Life Sciences, Hunan Normal University, Changsha, Hunan, China
- Hunan Provincial Key Laboratory of Animal Intestinal Function and Regulation, Changsha, Hunan, China
| | - Ling Liu
- Hunan International Joint Laboratory of Animal Intestinal Ecology and Health, Laboratory of Animal Nutrition and Human Health, College of Life Sciences, Hunan Normal University, Changsha, Hunan, China
- Hunan Provincial Key Laboratory of Animal Intestinal Function and Regulation, Changsha, Hunan, China
| | - Yuanyuan Tan
- Hunan International Joint Laboratory of Animal Intestinal Ecology and Health, Laboratory of Animal Nutrition and Human Health, College of Life Sciences, Hunan Normal University, Changsha, Hunan, China
- Hunan Provincial Key Laboratory of Animal Intestinal Function and Regulation, Changsha, Hunan, China
| | - Junwei Zhu
- Hunan International Joint Laboratory of Animal Intestinal Ecology and Health, Laboratory of Animal Nutrition and Human Health, College of Life Sciences, Hunan Normal University, Changsha, Hunan, China
- Hunan Provincial Key Laboratory of Animal Intestinal Function and Regulation, Changsha, Hunan, China
| | - Jifan Jiang
- Hunan International Joint Laboratory of Animal Intestinal Ecology and Health, Laboratory of Animal Nutrition and Human Health, College of Life Sciences, Hunan Normal University, Changsha, Hunan, China
- Hunan Provincial Key Laboratory of Animal Intestinal Function and Regulation, Changsha, Hunan, China
| | - Huaping Xie
- Hunan International Joint Laboratory of Animal Intestinal Ecology and Health, Laboratory of Animal Nutrition and Human Health, College of Life Sciences, Hunan Normal University, Changsha, Hunan, China
- Hunan Provincial Key Laboratory of Animal Intestinal Function and Regulation, Changsha, Hunan, China
| |
Collapse
|
14
|
Mahmoudi SK, Tarzemani S, Aghajanzadeh T, Kasravi M, Hatami B, Zali MR, Baghaei K. Exploring the role of genetic variations in NAFLD: implications for disease pathogenesis and precision medicine approaches. Eur J Med Res 2024; 29:190. [PMID: 38504356 PMCID: PMC10953212 DOI: 10.1186/s40001-024-01708-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2023] [Accepted: 02/01/2024] [Indexed: 03/21/2024] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) is one of the leading causes of chronic liver diseases, affecting more than one-quarter of people worldwide. Hepatic steatosis can progress to more severe forms of NAFLD, including NASH and cirrhosis. It also may develop secondary diseases such as diabetes and cardiovascular disease. Genetic and environmental factors regulate NAFLD incidence and progression, making it a complex disease. The contribution of various environmental risk factors, such as type 2 diabetes, obesity, hyperlipidemia, diet, and sedentary lifestyle, to the exacerbation of liver injury is highly understood. Nevertheless, the underlying mechanisms of genetic variations in the NAFLD occurrence or its deterioration still need to be clarified. Hence, understanding the genetic susceptibility to NAFLD is essential for controlling the course of the disease. The current review discusses genetics' role in the pathological pathways of NAFLD, including lipid and glucose metabolism, insulin resistance, cellular stresses, and immune responses. Additionally, it explains the role of the genetic components in the induction and progression of NAFLD in lean individuals. Finally, it highlights the utility of genetic knowledge in precision medicine for the early diagnosis and treatment of NAFLD patients.
Collapse
Affiliation(s)
- Seyedeh Kosar Mahmoudi
- Basic and Molecular Epidemiology of Gastrointestinal Disorders Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, 1985714711, Iran
| | - Shadi Tarzemani
- Basic and Molecular Epidemiology of Gastrointestinal Disorders Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, 1985714711, Iran
| | - Taha Aghajanzadeh
- Basic and Molecular Epidemiology of Gastrointestinal Disorders Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, 1985714711, Iran.
| | - Mohammadreza Kasravi
- Basic and Molecular Epidemiology of Gastrointestinal Disorders Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, 1985714711, Iran
| | - Behzad Hatami
- Gastroenterology and Liver Diseases Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, 1985714711, Iran
| | - Mohammad Reza Zali
- Gastroenterology and Liver Diseases Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, 1985714711, Iran
| | - Kaveh Baghaei
- Basic and Molecular Epidemiology of Gastrointestinal Disorders Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, 1985714711, Iran.
- Gastroenterology and Liver Diseases Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, 1985714711, Iran.
| |
Collapse
|
15
|
Tidwell J, Wu GY. Unique Genetic Features of Lean NAFLD: A Review of Mechanisms and Clinical Implications. J Clin Transl Hepatol 2024; 12:70-78. [PMID: 38250459 PMCID: PMC10794266 DOI: 10.14218/jcth.2023.00252] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/25/2023] [Revised: 07/11/2023] [Accepted: 08/04/2023] [Indexed: 01/23/2024] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) affects 25% of the global population. About 20% have a normal body mass index (BMI) and a variant known as lean NAFLD. Unlike typical NAFLD cases associated with obesity and diabetes, lean NAFLD causes liver disease by mechanisms not related to excess weight or insulin resistance. Genetic disorders are among the major factors in developing lean NAFLD, and genome-wide association studies have identified several genes associated with the condition. This review aims to increase awareness by describing the genetic markers linked to NAFLD and the defects involved in developing lean NAFLD.
Collapse
Affiliation(s)
- Jasmine Tidwell
- Department of Medicine, University of Connecticut Health Center, Farmington, CT, USA
| | - George Y. Wu
- Department of Medicine, University of Connecticut Health Center, Farmington, CT, USA
- Division of Gastroenterology-Hepatology, University of Connecticut Health Center, Farmington, CT, USA
| |
Collapse
|
16
|
van Zwol W, van de Sluis B, Ginsberg HN, Kuivenhoven JA. VLDL Biogenesis and Secretion: It Takes a Village. Circ Res 2024; 134:226-244. [PMID: 38236950 PMCID: PMC11284300 DOI: 10.1161/circresaha.123.323284] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/23/2023] [Accepted: 09/21/2023] [Indexed: 01/23/2024]
Abstract
The production and secretion of VLDLs (very-low-density lipoproteins) by hepatocytes has a direct impact on liver fat content, as well as the concentrations of cholesterol and triglycerides in the circulation and thus affects both liver and cardiovascular health, respectively. Importantly, insulin resistance, excess caloric intake, and lack of physical activity are associated with overproduction of VLDL, hepatic steatosis, and increased plasma levels of atherogenic lipoproteins. Cholesterol and triglycerides in remnant particles generated by VLDL lipolysis are risk factors for atherosclerotic cardiovascular disease and have garnered increasing attention over the last few decades. Presently, however, increased risk of atherosclerosis is not the only concern when considering today's cardiometabolic patients, as they often also experience hepatic steatosis, a prevalent disorder that can progress to steatohepatitis and cirrhosis. This duality of metabolic risk highlights the importance of understanding the molecular regulation of the biogenesis of VLDL, the lipoprotein that transports triglycerides and cholesterol out of the liver. Fortunately, there has been a resurgence of interest in the intracellular assembly, trafficking, degradation, and secretion of VLDL by hepatocytes, which has led to many exciting new molecular insights that are the topic of this review. Increasing our understanding of the biology of this pathway will aid to the identification of novel therapeutic targets to improve both the cardiovascular and the hepatic health of cardiometabolic patients. This review focuses, for the first time, on this duality.
Collapse
Affiliation(s)
- Willemien van Zwol
- Department of Paediatrics, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Bart van de Sluis
- Department of Paediatrics, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Henry. N. Ginsberg
- Department of Medicine, Vagelos College of Physicians and Surgeons, Columbia University, New York, New York, USA
| | - Jan Albert Kuivenhoven
- Department of Paediatrics, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| |
Collapse
|
17
|
Quaglia A, Roberts EA, Torbenson M. Developmental and Inherited Liver Disease. MACSWEEN'S PATHOLOGY OF THE LIVER 2024:122-294. [DOI: 10.1016/b978-0-7020-8228-3.00003-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/02/2025]
|
18
|
Henry Z, Janin A, Nony S, Marmontel O, Cariou B, Marrec M, Caussy C, Charrière S, Moulin P, Rieusset J, Perros F, Di Filippo M. Interest of minigene splicing reporter assay in familial hypobetalipoproteinemia genetic diagnosis: the example of the missense mutation APOB c.1468C>T. Clin Chem Lab Med 2023; 61:e259-e262. [PMID: 37309596 DOI: 10.1515/cclm-2023-0330] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Accepted: 06/02/2023] [Indexed: 06/14/2023]
Affiliation(s)
- Zoé Henry
- Service de Biochimie et Biologie Moléculaire, Laboratoire de Biologie Médicale MultiSites, Hospices Civils de Lyon, Bron, France
- Fédération d'endocrinologie, maladies métaboliques, diabète et nutrition, Hôpital Louis Pradel, Hospices Civils de Lyon, Bron, France
- CarMeN Laboratory, INSERM U1060, INRAE U1397, Université Claude Bernard Lyon 1, Pierre-Bénite, France
| | - Alexandre Janin
- Service de Biochimie et Biologie Moléculaire, Laboratoire de Biologie Médicale MultiSites, Hospices Civils de Lyon, Bron, France
- CNRS UMR5261, INSERM U1315, Institut NeuroMyoGène, Université Claude Bernard Lyon 1, Université de Lyon, Lyon, France
| | - Séverine Nony
- Service de Biochimie et Biologie Moléculaire, Laboratoire de Biologie Médicale MultiSites, Hospices Civils de Lyon, Bron, France
| | - Oriane Marmontel
- Service de Biochimie et Biologie Moléculaire, Laboratoire de Biologie Médicale MultiSites, Hospices Civils de Lyon, Bron, France
- CarMeN Laboratory, INSERM U1060, INRAE U1397, Université Claude Bernard Lyon 1, Pierre-Bénite, France
| | - Bertrand Cariou
- Nantes Université, CHU Nantes, CNRS, INSERM, L'institut du Thorax, Nantes, France
| | - Marie Marrec
- Nantes Université, CHU Nantes, CNRS, INSERM, L'institut du Thorax, Nantes, France
| | - Cyrielle Caussy
- CarMeN Laboratory, INSERM U1060, INRAE U1397, Université Claude Bernard Lyon 1, Pierre-Bénite, France
- Hôpital Lyon Sud, Département Endocrinologie, Diabète et Nutrition, Hospices Civils de Lyon, Pierre-Bénite, France
| | - Sybil Charrière
- Fédération d'endocrinologie, maladies métaboliques, diabète et nutrition, Hôpital Louis Pradel, Hospices Civils de Lyon, Bron, France
- CarMeN Laboratory, INSERM U1060, INRAE U1397, Université Claude Bernard Lyon 1, Pierre-Bénite, France
| | - Philippe Moulin
- Fédération d'endocrinologie, maladies métaboliques, diabète et nutrition, Hôpital Louis Pradel, Hospices Civils de Lyon, Bron, France
- CarMeN Laboratory, INSERM U1060, INRAE U1397, Université Claude Bernard Lyon 1, Pierre-Bénite, France
| | - Jennifer Rieusset
- CarMeN Laboratory, INSERM U1060, INRAE U1397, Université Claude Bernard Lyon 1, Pierre-Bénite, France
| | - Frédéric Perros
- CarMeN Laboratory, INSERM U1060, INRAE U1397, Université Claude Bernard Lyon 1, Pierre-Bénite, France
| | - Mathilde Di Filippo
- Service de Biochimie et Biologie Moléculaire, Laboratoire de Biologie Médicale MultiSites, Hospices Civils de Lyon, Bron, France
- CarMeN Laboratory, INSERM U1060, INRAE U1397, Université Claude Bernard Lyon 1, Pierre-Bénite, France
| |
Collapse
|
19
|
van Zwol W, Rimbert A, Wolters JC, Smit M, Bloks VW, Kloosterhuis NJ, Huijkman NCA, Koster MH, Tharehalli U, de Neck SM, Bournez C, Fuh MM, Kuipers J, Rajan S, de Bruin A, Ginsberg HN, van Westen GJP, Hussain MM, Scheja L, Heeren J, Zimmerman P, van de Sluis B, Kuivenhoven JA. Loss of hepatic SMLR1 causes hepatosteatosis and protects against atherosclerosis due to decreased hepatic VLDL secretion. Hepatology 2023; 78:1418-1432. [PMID: 36053190 PMCID: PMC10581432 DOI: 10.1002/hep.32709] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Revised: 08/02/2022] [Accepted: 08/03/2022] [Indexed: 12/08/2022]
Abstract
BACKGROUND AND AIMS The assembly and secretion of VLDL from the liver, a pathway that affects hepatic and plasma lipids, remains incompletely understood. We set out to identify players in the VLDL biogenesis pathway by identifying genes that are co-expressed with the MTTP gene that encodes for microsomal triglyceride transfer protein, key to the lipidation of apolipoprotein B, the core protein of VLDL. Using human and murine transcriptomic data sets, we identified small leucine-rich protein 1 ( SMLR1 ), encoding for small leucine-rich protein 1, a protein of unknown function that is exclusively expressed in liver and small intestine. APPROACH AND RESULTS To assess the role of SMLR1 in the liver, we used somatic CRISPR/CRISPR-associated protein 9 gene editing to silence murine Smlr1 in hepatocytes ( Smlr1 -LKO). When fed a chow diet, male and female mice show hepatic steatosis, reduced plasma apolipoprotein B and triglycerides, and reduced VLDL secretion without affecting microsomal triglyceride transfer protein activity. Immunofluorescence studies show that SMLR1 is in the endoplasmic reticulum and Cis-Golgi complex. The loss of hepatic SMLR1 in female mice protects against diet-induced hyperlipidemia and atherosclerosis but causes NASH. On a high-fat, high-cholesterol diet, insulin and glucose tolerance tests did not reveal differences in male Smlr1 -LKO mice versus controls. CONCLUSIONS We propose a role for SMLR1 in the trafficking of VLDL from the endoplasmic reticulum to the Cis-Golgi complex. While this study uncovers SMLR1 as a player in the VLDL assembly, trafficking, and secretion pathway, it also shows that NASH can occur with undisturbed glucose homeostasis and atheroprotection.
Collapse
Affiliation(s)
- Willemien van Zwol
- Department of Pediatrics, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Antoine Rimbert
- Université de Nantes, CNRS, INSERM, l'institut du thorax, Nantes, France
| | - Justina C. Wolters
- Department of Pediatrics, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Marieke Smit
- Department of Pediatrics, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Vincent W. Bloks
- Department of Pediatrics, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Niels J. Kloosterhuis
- Department of Pediatrics, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Nicolette C. A. Huijkman
- Department of Pediatrics, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Mirjam H. Koster
- Department of Pediatrics, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Umesh Tharehalli
- Department of Pediatrics, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Simon M. de Neck
- Department of Biomolecular Health Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, the Netherlands
| | - Colin Bournez
- Division of Drug Discovery and Safety, Leiden Academic Center for Drug Research, Leiden University, Leiden, The Netherlands
| | - Marceline M. Fuh
- Department of Biochemistry and Molecular Cell Biology, University Medical Center Hamburg−Eppendorf, Hamburg, Germany
| | - Jeroen Kuipers
- Department of Biomedical Sciences of Cells and Systems, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Sujith Rajan
- Department of Foundations of Medicine, NYU Long Island School of Medicine, Mineola, New York, USA
| | - Alain de Bruin
- Department of Pediatrics, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
- Department of Biomolecular Health Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, the Netherlands
| | - Henry N. Ginsberg
- Department of Medicine, Columbia University, Vagelos College of Physicians and Surgeons, New York, New York, USA
| | - Gerard J. P. van Westen
- Division of Drug Discovery and Safety, Leiden Academic Center for Drug Research, Leiden University, Leiden, The Netherlands
| | - M. Mahmood Hussain
- Department of Foundations of Medicine, NYU Long Island School of Medicine, Mineola, New York, USA
| | - Ludger Scheja
- Department of Biochemistry and Molecular Cell Biology, University Medical Center Hamburg−Eppendorf, Hamburg, Germany
| | - Joerg Heeren
- Department of Biochemistry and Molecular Cell Biology, University Medical Center Hamburg−Eppendorf, Hamburg, Germany
| | | | - Bart van de Sluis
- Department of Pediatrics, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Jan Albert Kuivenhoven
- Department of Pediatrics, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| |
Collapse
|
20
|
Nasr P, Jönsson C, Ekstedt M, Kechagias S. Non-metabolic causes of steatotic liver disease. METABOLISM AND TARGET ORGAN DAMAGE 2023; 3. [DOI: 10.20517/mtod.2023.20] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
Abstract
Hepatic steatosis is caused by exaggerated hepatic lipid accumulation and is a common histological and radiological finding. Non-alcoholic fatty liver disease (NAFLD), or metabolic dysfunction associated steatotic liver disease (MASLD), is highly associated with metabolic syndrome and represents the most common cause of hepatic steatosis. However, since several comorbidities, lifestyle factors, and drugs can cause hepatic steatosis, MASLD is, to some extent, a diagnosis of exclusion. Nevertheless, initiatives have been taken to encompass positive (instead of negative) criteria for diagnosis - such as the presence of cardiometabolic risk factors together with hepatic steatosis. Nonetheless, before confirming a patient with MASLD, it is essential to map and evaluate other causes of fatty liver disease or steatotic liver disease. Several causes of hepatic steatosis have been identified in studies; however, the study cohorts are scarce and often anecdotal. Additionally, many studies have shown correlation without proving causation, and many are retrospective without reporting relevant patient characteristics and comorbidities - making it difficult to draw conclusions regarding the underlying etiology or present comorbidity of hepatic steatosis. In this narrative review, we aimed to identify and summarize present studies evaluating the impact of the most common and often suggested causes of hepatic steatosis.
Collapse
|
21
|
Yue C, Li D, Fan S, Tao F, Yu Y, Lu W, Chen Q, Yuan A, Wu J, Zhao G, Dong H, Hu Y. Long-term and liver-selected ginsenoside C-K nanoparticles retard NAFLD progression by restoring lipid homeostasis. Biomaterials 2023; 301:122291. [PMID: 37619263 DOI: 10.1016/j.biomaterials.2023.122291] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2023] [Revised: 07/25/2023] [Accepted: 08/19/2023] [Indexed: 08/26/2023]
Abstract
Non-alcoholic fatty liver disease (NAFLD) is the most prevalent hepatic disease characterized as lipid accumulation, yet without any approved drug. And development of therapeutic molecules is obstructed by low efficiency and organ toxicity. Herein, we develop a long-term, low-toxic and liver-selected nano candidate, nabCK, to alleviate NAFLD. NabCK is simply composed by natural compound ginsenoside compound K (CK) and albumin. As a major metabolite of ginseng, ginsenoside CK has excellently modulating functions for lipid metabolism, but accompanied by an extremely poor bioavailability <1%. Albumin is a key lipid carrier secreted and metabolized by livers. Thereby, it can improve solubility and liver-localization of CK. In adipocytes and hepatocytes, nabCK prevents lipid deposition and eliminates lipid droplets. Transcriptomic analysis reveals that nabCK rectifies various pathways that involved in steatosis development, including lipid absorption, lipid export, fatty acid biosynthesis, lipid storage and inflammation. All these pathways are modulated by mTOR, the pivotal feedback sensor that is hyperactive in NAFLD. NabCK suppresses mTOR activation to restores lipid homeostasis. In high-fat diet (HFD) induced NAFLD mice, nabCK retards development of steatosis and fibrosis, coupling a protective effect on cardiac tissues from lipotoxicity. Together, nabCK is a safe and potent candidate to offer benefits for NAFLD treatment.
Collapse
Affiliation(s)
- Chunyan Yue
- State Key Laboratory of Pharmaceutical Biotechnology, Medical School, School of Life Science, Nanjing University, Nanjing , 210093, China; Institute of Drug R&D, Medical School, Nanjing University, Nanjing, 210093, China
| | - Dandan Li
- State Key Laboratory of Pharmaceutical Biotechnology, Medical School, School of Life Science, Nanjing University, Nanjing , 210093, China; Institute of Drug R&D, Medical School, Nanjing University, Nanjing, 210093, China
| | - Shuxin Fan
- State Key Laboratory of Pharmaceutical Biotechnology, Medical School, School of Life Science, Nanjing University, Nanjing , 210093, China; Institute of Drug R&D, Medical School, Nanjing University, Nanjing, 210093, China
| | - Feng Tao
- State Key Laboratory of Pharmaceutical Biotechnology, Medical School, School of Life Science, Nanjing University, Nanjing , 210093, China; Institute of Drug R&D, Medical School, Nanjing University, Nanjing, 210093, China
| | - Yue Yu
- Affiliated Drum Tower Hospital, Medical School, Nanjing University, Nanjing, 210008, China
| | - Wenjing Lu
- State Key Laboratory of Pharmaceutical Biotechnology, Medical School, School of Life Science, Nanjing University, Nanjing , 210093, China; Institute of Drug R&D, Medical School, Nanjing University, Nanjing, 210093, China
| | - Qian Chen
- State Key Laboratory of Pharmaceutical Biotechnology, Medical School, School of Life Science, Nanjing University, Nanjing , 210093, China; Institute of Drug R&D, Medical School, Nanjing University, Nanjing, 210093, China
| | - Ahu Yuan
- State Key Laboratory of Pharmaceutical Biotechnology, Medical School, School of Life Science, Nanjing University, Nanjing , 210093, China; Institute of Drug R&D, Medical School, Nanjing University, Nanjing, 210093, China
| | - Jinhui Wu
- State Key Laboratory of Pharmaceutical Biotechnology, Medical School, School of Life Science, Nanjing University, Nanjing , 210093, China; Institute of Drug R&D, Medical School, Nanjing University, Nanjing, 210093, China
| | - Guoping Zhao
- Key Laboratory of Synthetic Biology, Institute of Plant Physiology and Ecology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, 200032, China
| | - Hong Dong
- State Key Laboratory of Pharmaceutical Biotechnology, Medical School, School of Life Science, Nanjing University, Nanjing , 210093, China; Institute of Drug R&D, Medical School, Nanjing University, Nanjing, 210093, China.
| | - Yiqiao Hu
- State Key Laboratory of Pharmaceutical Biotechnology, Medical School, School of Life Science, Nanjing University, Nanjing , 210093, China; Institute of Drug R&D, Medical School, Nanjing University, Nanjing, 210093, China.
| |
Collapse
|
22
|
Pais R, Cariou B, Noureddin M, Francque S, Schattenberg JM, Abdelmalek MF, Lalazar G, Varma S, Dietrich J, Miller V, Sanyal A, Ratziu V. A proposal from the liver forum for the management of comorbidities in non-alcoholic steatohepatitis therapeutic trials. J Hepatol 2023; 79:829-841. [PMID: 37001695 DOI: 10.1016/j.jhep.2023.03.014] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Revised: 02/08/2023] [Accepted: 03/13/2023] [Indexed: 06/19/2023]
Abstract
The current document has been developed by the Liver Forum who mandated the NAFLD-Associated Comorbidities Working Group - a multistakeholder group comprised of experts from academic medicine, industry and patient associations - to identify aspects of diverse comorbidities frequently associated with non-alcoholic steatohepatitis (NASH) that can interfere with the conduct of therapeutic trials and, in particular, impact efficacy and safety results. The objective of this paper is to propose guidance for the management of relevant comorbidities in both candidates and actual participants in NASH therapeutic trials. We relied on specific guidelines from scientific societies, when available, but adapted them to the particulars of NASH trials with the aim of addressing multiple interacting requirements such as maintaining patient safety, reaching holistic therapeutic objectives, minimising confounding effects on efficacy and safety of investigational agents and allowing for trial completion. We divided the field of action into: first, analysis and stabilisation of the patient's condition before inclusion in the trial and, second, management of comorbidities during trial conduct. For the former, we discussed the concept of acceptable vs. optimal control of comorbidities, defined metabolic and ponderal stability prior to randomisation and weighed the pros and cons of a run-in period. For the latter, we analysed non-hepatological comorbid conditions for changes or acute events possibly occurring during the trial, including changes in alcohol consumption, in order to detail when specific interventions are necessary and how best to manage concomitant drug intake in line with methodological constraints. These recommendations are intended to act as a guide for clinical trialists and are open to further refinement when additional data become available.
Collapse
Affiliation(s)
- Raluca Pais
- Sorbonne Université, Assistance Publique-Hôpitaux de Paris, Hôpital Pitié-Salpêtrière, Institute of Cardiometabolism and Nutrition, France; Centre de Recherche Saint Antoine, INSERM UMRS_938 Paris, France
| | - Bertrand Cariou
- Nantes Université, CHU Nantes, CNRS, INSERM, l'institut du Thorax, F-44000 Nantes, France
| | | | - Sven Francque
- Department of Gastroenterology Hepatology, Antwerp University Hospital, Drie Eikenstraat 655, B-2650 Edegem, Belgium; InflaMed Centre of Excellence, Laboratory for Experimental Medicine and Paediatrics, Translational Sciences in Inflammation and Immunology, Faculty of Medicine and Health Sciences, University of Antwerp, Universiteitsplein 1, B-2610 Wilrijk, Belgium European Reference Network on Hepatological Diseases (ERN RARE-LIVER), Belgium
| | - Jörn M Schattenberg
- Metabolic Liver Research Program, I. Department of Medicine, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - Manal F Abdelmalek
- Division of Gastroenterology and Hepatology, Duke University, Durham, NC, USA
| | - Gadi Lalazar
- Liver Unit, Digestive Disease Institute, Shaare Zedek Medical Center, Jerusalem, Israel
| | - Sharat Varma
- Novo Nordisk A/S, Vandtårnsvej 108-110, 2860 Søborg Denmark
| | - Julie Dietrich
- GENFIT, Parc Eurasanté 885, Avenue Eugène Avinée, 59120, Loos, France
| | - Veronica Miller
- Forum for Collaborative Research, University of California Berkeley School of Public Health, Washington D.C., USA
| | - Arun Sanyal
- Virginia Commonwealth University School of Medicine, Richmond, Virginia, USA
| | - Vlad Ratziu
- Sorbonne Université, Assistance Publique-Hôpitaux de Paris, Hôpital Pitié-Salpêtrière, Institute of Cardiometabolism and Nutrition, France; INSERM UMRS 1138 CRC, Paris, France.
| |
Collapse
|
23
|
McHenry S, Awad A, Kozlitina J, Stitziel NO, Davidson NO. Low LDL Cholesterol Is Not an Independent Risk Factor for Hepatic Steatosis. Dig Dis Sci 2023; 68:3451-3457. [PMID: 37291473 DOI: 10.1007/s10620-023-07980-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/04/2022] [Accepted: 05/11/2023] [Indexed: 06/10/2023]
Abstract
BACKGROUND Genetic mutations causing defective VLDL secretion and low LDL cholesterol are associated with hepatic steatosis and nonalcoholic fatty liver disease (NAFLD). AIMS Determine if low LDL cholesterol (< 5th percentile) was an independent predictor of hepatic steatosis. METHODS Secondary data analysis of the Dallas Heart study (an urban, multiethnic, probability-based sample), we defined hepatic steatosis utilizing intrahepatic triglyceride (IHTG) analyzed using magnetic resonance spectroscopy in conjunction and available demographic, serological and genetic information. We exclude patients on lipid lowering medications. RESULTS Of the 2094 subjects that met our exclusion criteria, 86 had a low LDL cholesterol, of whom 19 (22%) exhibited hepatic steatosis. After matching for age, sex, BMI, and alcohol consumption, low LDL cholesterol was not a risk factor for hepatic steatosis compared to those with normal (50-180 mg/dL) or high (> 180 mg/dL) LDL. When analyzed as a continuous variable, we observed lower IHTG in the low LDL group compared to the normal or high LDL groups (2.2%, 3.5%, 4.6%; all pairwise comparisons p < 0.001). Subjects with both hepatic steatosis and low LDL cholesterol exhibited a more favorable lipid profile but similar insulin resistance and hepatic fibrosis risk compared to other subjects with hepatic steatosis. The distribution of variant alleles associated with NAFLD, including PNPLA3, GCKR, and MTTP was indistinguishable between subjects with hepatic steatosis and low versus high LDL cholesterol. CONCLUSION These findings suggest that low serum LDL levels are not a useful predictor of hepatic steatosis and NAFLD. Moreover, subjects with low LDL exhibit a more favorable lipid profile and lower IHTG.
Collapse
Affiliation(s)
- Scott McHenry
- Division of Gastroenterology, Department of Medicine, Washington University in Saint Louis, St. Louis, MO, 53110, USA.
| | - Ameen Awad
- Division of Gastroenterology, Department of Medicine, Washington University in Saint Louis, St. Louis, MO, 53110, USA
| | - Julia Kozlitina
- Eugene McDermott Center for Human Growth and Development, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Nathan O Stitziel
- Division of Gastroenterology, Department of Medicine, Washington University in Saint Louis, St. Louis, MO, 53110, USA
| | - Nicholas O Davidson
- Division of Gastroenterology, Department of Medicine, Washington University in Saint Louis, St. Louis, MO, 53110, USA
| |
Collapse
|
24
|
Grove JI, Lo PC, Shrine N, Barwell J, Wain LV, Tobin MD, Salter AM, Borkar AN, Cuevas-Ocaña S, Bennett N, John C, Ntalla I, Jones GE, Neal CP, Thomas MG, Kuht H, Gupta P, Vemala VM, Grant A, Adewoye AB, Shenoy KT, Balakumaran LK, Hollox EJ, Hannan NR, Aithal GP. Identification and characterisation of a rare MTTP variant underlying hereditary non-alcoholic fatty liver disease. JHEP Rep 2023; 5:100764. [PMID: 37484212 PMCID: PMC10362796 DOI: 10.1016/j.jhepr.2023.100764] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 03/28/2023] [Accepted: 04/11/2023] [Indexed: 07/25/2023] Open
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.
Collapse
Affiliation(s)
- Jane I. Grove
- National Institute of Health Research (NIHR) Nottingham Biomedical Research Centre, Nottingham University Hospitals NHS Trust & University of Nottingham, Nottingham, UK
- Nottingham Digestive Diseases Centre, Translational Medical Sciences, School of Medicine, University of Nottingham, Nottingham, UK
| | - Peggy C.K. Lo
- Translational Medical Sciences, School of Medicine, University of Nottingham, Nottingham, UK
- University of Nottingham Biodiscovery Institute, University of Nottingham, Nottingham, UK
| | - Nick Shrine
- Genetic Epidemiology Group, Department of Population Health Sciences, University of Leicester, Leicester, UK
| | - Julian Barwell
- Clinical Genetics Department, University Hospitals Leicester NHS Trust, Leicester, UK
| | - Louise V. Wain
- Genetic Epidemiology Group, Department of Population Health Sciences, University of Leicester, Leicester, UK
- NIHR Leicester Respiratory Biomedical Research Centre, Glenfield Hospital, Leicester, UK
| | - Martin D. Tobin
- Genetic Epidemiology Group, Department of Population Health Sciences, University of Leicester, Leicester, UK
- NIHR Leicester Respiratory Biomedical Research Centre, Glenfield Hospital, Leicester, UK
| | | | - Aditi N. Borkar
- School of Veterinary Medicine and Science, University of Nottingham, Nottingham, UK
| | - Sara Cuevas-Ocaña
- Translational Medical Sciences, School of Medicine, University of Nottingham, Nottingham, UK
- University of Nottingham Biodiscovery Institute, University of Nottingham, Nottingham, UK
| | - Neil Bennett
- Genetic Epidemiology Group, Department of Population Health Sciences, University of Leicester, Leicester, UK
| | - Catherine John
- Genetic Epidemiology Group, Department of Population Health Sciences, University of Leicester, Leicester, UK
| | - Ioanna Ntalla
- Clinical Genetics Department, University Hospitals Leicester NHS Trust, Leicester, UK
| | - Gabriela E. Jones
- Clinical Genetics Department, University Hospitals Leicester NHS Trust, Leicester, UK
| | | | - Mervyn G. Thomas
- Ulverscroft Eye Unit, Department of Neuroscience, Psychology and Behaviour, University of Leicester, Leicester, UK
| | - Helen Kuht
- Ulverscroft Eye Unit, Department of Neuroscience, Psychology and Behaviour, University of Leicester, Leicester, UK
| | - Pankaj Gupta
- Department of Chemical Pathology and Metabolic Diseases, University Hospitals of Leicester NHS Trust, Leicester, UK
- Department of Cardiovascular Sciences, University of Leicester, Leicester, UK
| | - Vishwaraj M. Vemala
- Department of Gastroenterology, University Hospitals of Leicester NHS Trust, Leicester, UK
| | - Allister Grant
- Department of Gastroenterology, University Hospitals of Leicester NHS Trust, Leicester, UK
| | - Adeolu B. Adewoye
- Department of Genetics and Genome Biology, University of Leicester, Leicester, UK
| | | | | | - Edward J. Hollox
- Department of Genetics and Genome Biology, University of Leicester, Leicester, UK
| | - Nicholas R.F. Hannan
- Translational Medical Sciences, School of Medicine, University of Nottingham, Nottingham, UK
- University of Nottingham Biodiscovery Institute, University of Nottingham, Nottingham, UK
| | - Guruprasad P. Aithal
- National Institute of Health Research (NIHR) Nottingham Biomedical Research Centre, Nottingham University Hospitals NHS Trust & University of Nottingham, Nottingham, UK
- Nottingham Digestive Diseases Centre, Translational Medical Sciences, School of Medicine, University of Nottingham, Nottingham, UK
| |
Collapse
|
25
|
Molk N, Bitenc M, Urlep D, Zerjav Tansek M, Bertok S, Trebusak Podkrajsek K, Sustar U, Kovac J, Battelino T, Debeljak M, Groselj U. Non-alcoholic fatty liver disease in a pediatric patient with heterozygous familial hypobetalipoproteinemia due to a novel APOB variant: a case report and systematic literature review. Front Med (Lausanne) 2023; 10:1106441. [PMID: 37384046 PMCID: PMC10293746 DOI: 10.3389/fmed.2023.1106441] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2022] [Accepted: 05/09/2023] [Indexed: 06/30/2023] Open
Abstract
Background Familial hypobetalipoproteinemia (FHBL) is an autosomal semi-dominant disorder usually caused by variants in the APOB gene that frequently interferes with protein length. Clinical manifestations include malabsorption, non-alcoholic fatty liver disease, low levels of lipid-soluble vitamins, and neurological, endocrine, and hematological dysfunction. Methods Genomic DNA was isolated from the blood samples of the pediatric patient with hypocholesterolemia and his parents and brother. Next-generation sequencing (NGS) was performed, and an expanded dyslipidemia panel was employed for genetic analysis. In addition, a systematic review of the literature on FHBL heterozygous patients was performed. Case report Genetic investigation revealed the presence of a heterozygous variant in the APOB (NM_000384.3) gene c.6624dup[=], which changes the open reading frame and leads to early termination of translation into the p.Leu2209IlefsTer5 protein (NP_000375.3). The identified variant was not previously reported. Familial segregation analysis confirmed the variant in the mother of the subject, who also has a low level of low-density lipoprotein and non-alcoholic fatty liver disease. We have introduced therapy that includes limiting fats in the diet and adding lipid-soluble vitamins E, A, K, and D and calcium carbonate. We reported 35 individuals with APOB gene variations linked to FHBL in the systematic review. Conclusion We have identified a novel pathogenic variant in the APOB gene causing FHBL in pediatric patients with hypocholesterolemia and fatty liver disease. This case illustrates the importance of genetic testing for dyslipidemias in patients with significant decreases in plasma cholesterol as we can avoid damaging neurological and ophthalmological effects by sufficient vitamin supplementation and regular follow-ups.
Collapse
Affiliation(s)
- Neza Molk
- Department of Pediatric Endocrinology, Diabetes and Metabolism, University Medical Center-University Children's Hospital, Ljubljana, Slovenia
| | - Mojca Bitenc
- Department of Pediatric Endocrinology, Diabetes and Metabolism, University Medical Center-University Children's Hospital, Ljubljana, Slovenia
| | - Darja Urlep
- Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
- Department of Gastroenterology, Hepatology and Nutrition, University Children's Hospital Ljubljana, University Medical Center, Ljubljana, Slovenia
| | - Mojca Zerjav Tansek
- Department of Pediatric Endocrinology, Diabetes and Metabolism, University Medical Center-University Children's Hospital, Ljubljana, Slovenia
- Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| | - Sara Bertok
- Department of Pediatric Endocrinology, Diabetes and Metabolism, University Medical Center-University Children's Hospital, Ljubljana, Slovenia
- Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| | - Katarina Trebusak Podkrajsek
- Department of Gastroenterology, Hepatology and Nutrition, University Children's Hospital Ljubljana, University Medical Center, Ljubljana, Slovenia
- Clinical Institute for Special Laboratory Diagnostics, University Children's Hospital, University Medical Center Ljubljana, Ljubljana, Slovenia
| | - Ursa Sustar
- Department of Pediatric Endocrinology, Diabetes and Metabolism, University Medical Center-University Children's Hospital, Ljubljana, Slovenia
- Clinical Institute for Special Laboratory Diagnostics, University Children's Hospital, University Medical Center Ljubljana, Ljubljana, Slovenia
| | - Jernej Kovac
- Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
- Clinical Institute for Special Laboratory Diagnostics, University Children's Hospital, University Medical Center Ljubljana, Ljubljana, Slovenia
| | - Tadej Battelino
- Department of Pediatric Endocrinology, Diabetes and Metabolism, University Medical Center-University Children's Hospital, Ljubljana, Slovenia
- Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| | - Marusa Debeljak
- Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
- Clinical Institute for Special Laboratory Diagnostics, University Children's Hospital, University Medical Center Ljubljana, Ljubljana, Slovenia
| | - Urh Groselj
- Department of Pediatric Endocrinology, Diabetes and Metabolism, University Medical Center-University Children's Hospital, Ljubljana, Slovenia
- Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| |
Collapse
|
26
|
Gill PK, Hegele RA. Low cholesterol states: clinical implications and management. Expert Rev Endocrinol Metab 2023; 18:241-253. [PMID: 37089071 DOI: 10.1080/17446651.2023.2204932] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/04/2023] [Accepted: 04/17/2023] [Indexed: 04/25/2023]
Abstract
INTRODUCTION Hypocholesterolemia results from genetic - both monogenic and polygenic - and non-genetic causes and can sometimes be a source of clinical concern. We review etiologies and sequelae of hypocholesterolemia and therapeutics inspired from genetic hypocholesterolemia. AREAS COVERED Monogenic hypocholesterolemia disorders caused by the complete absence of apolipoprotein (apo) B-containing lipoproteins (abetalipoproteinemia and homozygous hypobetalipoproteinemia) or an isolated absence of apo B-48 lipoproteinemia (chylomicron retention disease) lead to clinical sequelae. These include gastrointestinal disturbances and severe vitamin deficiencies that affect multiple body systems, i.e. neurological, musculoskeletal, ophthalmological, and hematological. Monogenic hypocholesterolemia disorders with reduced but not absent levels of apo B lipoproteins have a milder clinical presentation and patients are protected against atherosclerotic cardiovascular disease. Patients with heterozygous hypobetalipoproteinemia have somewhat increased risk of hepatic disease, while patients with PCSK9 deficiency, ANGPTL3 deficiency, and polygenic hypocholesterolemia typically have anunremarkable clinical presentation. EXPERT OPINION In patients with severe monogenic hypocholesterolemia, early initiation of high-dose vitamin therapy and a low-fat diet are essential for optimal prognosis. The molecular basis of monogenic hypocholesterolemia has inspired novel therapeutics to help patients with the opposite phenotype - i.e. elevated apo B-containing lipoproteins. In particular, inhibitors of PCSK9 and ANGPTL3 show important clinical impact.
Collapse
Affiliation(s)
- Praneet K Gill
- Department of Medicine, Schulich School of Medicine and Dentistry, Western University, London, Canada
| | - Robert A Hegele
- Department of Medicine, Schulich School of Medicine and Dentistry, Western University, London, Canada
| |
Collapse
|
27
|
Zheng M, Huang DQ, Konkwo C, Agrawal S, Khera AV, Loomba R, Vilarinho S, Ajmera V. Genomic analysis of lean individuals with NAFLD identifies monogenic disorders in a prospective cohort study. JHEP Rep 2023; 5:100692. [PMID: 36937991 PMCID: PMC10017416 DOI: 10.1016/j.jhepr.2023.100692] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Revised: 01/13/2023] [Accepted: 01/17/2023] [Indexed: 02/05/2023] Open
Abstract
Background & Aims Lean patients with non-alcoholic fatty liver disease (NAFLD) represent 10-20% of the affected population and may have heterogeneous drivers of disease. We have recently proposed the evaluation of patients with lean NAFLD without visceral adiposity for rare monogenic drivers of disease. Here, we aimed to validate this framework in a well-characterised cohort of patients with biopsy-proven NAFLD by performing whole exome sequencing. Methods This prospective study included 124 patients with biopsy-proven NAFLD and paired liver biopsies who underwent standardised research visits including advanced magnetic resonance imaging (MRI) assessment of liver fat and stiffness. Results Six patients with lean NAFLD were identified and underwent whole exome sequencing. Two lean patients (33%) were identified to have monogenic disorders. The lean patients with monogenic disorders had similar age, and anthropometric and MRI characteristics to lean patients without a monogenic disorder. Patient 1 harbours a rare homozygous pathogenic mutation in ALDOB (aldolase B) and was diagnosed with hereditary fructose intolerance. Patient 2 harbours a rare heterozygous mutation in apolipoprotein B (APOB). The pathogenicity of this APOB variant (p.Val1856CysfsTer2) was further validated in the UK Biobank and associated with lower circulating APOB levels (beta = -0.51 g/L, 95% CI -0.65 to -0.36 g/L, p = 1.4 × 10-11) and higher liver fat on MRI (beta = +10.4%, 95% CI 4.3-16.5%, p = 8.8 × 10-4). Hence, patient 2 was diagnosed with heterozygous familial hypobetalipoproteinaemia. Conclusions In this cohort of well-characterised patients with lean NAFLD without visceral adiposity, 33% (2/6) had rare monogenic drivers of disease, highlighting the importance of genomic analysis in this NAFLD subtype. Impact and Implications Although most people with non-alcoholic fatty liver disease (NAFLD) are overweight or obese, a subset are lean and may have unique genetic mutations that cause their fatty liver disease. We show that 33% of study participants with NAFLD who were lean harboured unique mutations that cause their fatty liver, and that these mutations had effects beyond the liver. This study demonstrates the value of genetic assessment of NAFLD in lean individuals to identify distinct subtypes of disease.
Collapse
Key Words
- ALT, alanine aminotransferase
- APOB, apolipoprotein B
- FHBL, familial hypobetalipoproteinaemia
- LOFHC, high-confidence predicted loss-of-function
- MRE, magnetic resonance elastography
- MRI, magnetic resonance imaging
- MRI-PDFF, magnetic resonance imaging proton density fat fraction
- NAFLD
- NAFLD, non-alcoholic fatty liver disease
- Non-obese
- Rare genetic variants
- UCSD, University of California San Diego
- WES, whole exome sequencing
- Whole exome sequencing
Collapse
Affiliation(s)
- Melanie Zheng
- Departments of Internal Medicine, Section of Digestive Diseases, and of Pathology, Yale School of Medicine, New Haven, CT, USA
| | - Daniel Q. Huang
- NAFLD Research Center, Division of Gastroenterology, University of California at San Diego, La Jolla, CA, USA
- Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Chigoziri Konkwo
- Departments of Internal Medicine, Section of Digestive Diseases, and of Pathology, Yale School of Medicine, New Haven, CT, USA
| | - Saaket Agrawal
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Amit V. Khera
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Verve Therapeutics, Cambridge, MA, USA
| | - Rohit Loomba
- NAFLD Research Center, Division of Gastroenterology, University of California at San Diego, La Jolla, CA, USA
| | - Sílvia Vilarinho
- Departments of Internal Medicine, Section of Digestive Diseases, and of Pathology, Yale School of Medicine, New Haven, CT, USA
- Department of Pathology, Yale School of Medicine, New Haven, CT, USA
| | - Veeral Ajmera
- NAFLD Research Center, Division of Gastroenterology, University of California at San Diego, La Jolla, CA, USA
| |
Collapse
|
28
|
Li X, Liu Q, Pan Y, Chen S, Zhao Y, Hu Y. New insights into the role of dietary triglyceride absorption in obesity and metabolic diseases. Front Pharmacol 2023; 14:1097835. [PMID: 36817150 PMCID: PMC9932209 DOI: 10.3389/fphar.2023.1097835] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2022] [Accepted: 01/20/2023] [Indexed: 02/05/2023] Open
Abstract
The incidence of obesity and associated metabolic diseases is increasing globally, adversely affecting human health. Dietary fats, especially triglycerides, are an important source of energy for the body, and the intestine absorbs lipids through a series of orderly and complex steps. A long-term high-fat diet leads to intestinal dysfunction, inducing obesity and metabolic disorders. Therefore, regulating dietary triglycerides absorption is a promising therapeutic strategy. In this review, we will discuss diverse aspects of the dietary triglycerides hydrolysis, fatty acid uptake, triglycerides resynthesis, chylomicron assembly, trafficking, and secretion processes in intestinal epithelial cells, as well as potential targets in this process that may influence dietary fat-induced obesity and metabolic diseases. We also mention the possible shortcomings and deficiencies in modulating dietary lipid absorption targets to provide a better understanding of their administrability as drugs in obesity and related metabolic disorders.
Collapse
Affiliation(s)
- Xiaojing Li
- Key Laboratory of Liver and Kidney Diseases (Ministry of Education), Institute of Liver Diseases, Shanghai Key Laboratory of Traditional Chinese Clinical Medicine, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Qiaohong Liu
- Institute of Clinical Pharmacology, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yuqing Pan
- Key Laboratory of Liver and Kidney Diseases (Ministry of Education), Institute of Liver Diseases, Shanghai Key Laboratory of Traditional Chinese Clinical Medicine, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Si Chen
- Key Laboratory of Liver and Kidney Diseases (Ministry of Education), Institute of Liver Diseases, Shanghai Key Laboratory of Traditional Chinese Clinical Medicine, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yu Zhao
- Key Laboratory of Liver and Kidney Diseases (Ministry of Education), Institute of Liver Diseases, Shanghai Key Laboratory of Traditional Chinese Clinical Medicine, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China,*Correspondence: Yu Zhao, ; Yiyang Hu,
| | - Yiyang Hu
- Key Laboratory of Liver and Kidney Diseases (Ministry of Education), Institute of Liver Diseases, Shanghai Key Laboratory of Traditional Chinese Clinical Medicine, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China,Institute of Clinical Pharmacology, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China,*Correspondence: Yu Zhao, ; Yiyang Hu,
| |
Collapse
|
29
|
Han SK, Baik SK, Kim MY. Non-alcoholic fatty liver disease: Definition and subtypes. Clin Mol Hepatol 2023; 29:S5-S16. [PMID: 36577427 PMCID: PMC10029964 DOI: 10.3350/cmh.2022.0424] [Citation(s) in RCA: 102] [Impact Index Per Article: 51.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/28/2022] [Revised: 12/21/2022] [Accepted: 12/24/2022] [Indexed: 12/30/2022] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) is one of the most common liver diseases worldwide, with a global prevalence of approximately 30%. However, the prevalence of NAFLD has been variously reported depending on the comorbidities. The rising prevalence of obesity in both the adult and pediatric populations is projected to consequently continue increasing NAFLD prevalence. It is a major cause of chronic liver disease worldwide, including cirrhosis and hepatocellular carcinoma (HCC). NAFLD has a variety of clinical phenotypes and heterogeneity due to the complexity of pathogenesis and clinical conditions of its occurrence, resulting in various clinical prognoses. In this article, we briefly described the basic definition of NAFLD and classified the subtypes based on current knowledge in this field.
Collapse
Affiliation(s)
- Seul Ki Han
- Department of Internal Medicine, Yonsei University Wonju College of Medicine, Wonju, Korea
- Regenerative Medicine Research Center, Yonsei University Wonju College of Medicine, Wonju, Korea
- Cell Therapy and Tissue Engineering Center, Yonsei University Wonju College of Medicine, Wonju, Korea
| | - Soon Koo Baik
- Department of Internal Medicine, Yonsei University Wonju College of Medicine, Wonju, Korea
- Regenerative Medicine Research Center, Yonsei University Wonju College of Medicine, Wonju, Korea
- Cell Therapy and Tissue Engineering Center, Yonsei University Wonju College of Medicine, Wonju, Korea
| | - Moon Young Kim
- Department of Internal Medicine, Yonsei University Wonju College of Medicine, Wonju, Korea
- Regenerative Medicine Research Center, Yonsei University Wonju College of Medicine, Wonju, Korea
- Cell Therapy and Tissue Engineering Center, Yonsei University Wonju College of Medicine, Wonju, Korea
| |
Collapse
|
30
|
Liu R, Jin Y, Liu B, Zhang Q, Li X, Cai D, Tian L, Jiang X, Zhang W, Sun J, Bai W. Untargeted Lipidomics Revealed the Protective Effects of Cyanidin-3- O-glucoside on Bisphenol A-Induced Liver Lipid Metabolism Disorder in Rats. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2023; 71:1077-1090. [PMID: 36597173 DOI: 10.1021/acs.jafc.2c06849] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/17/2023]
Abstract
Bisphenol A (BPA) is an estrogenic endocrine disruptor that induces metabolic disorders. Cyanidin-3-O-glucoside (C3G) has multiple functional activities and is the most abundant anthocyanin belonging to the flavonoid subgroup. This study aimed to investigate the protective effect of C3G on BPA-induced liver lipid metabolism disorder and explore its mechanism via lipidomics analysis. The results showed that C3G supplementation significantly ameliorated the serum levels of low-density lipoprotein cholesterol, high-density lipoprotein cholesterol, total cholesterol, triacylglycerols (TG), and alanine and aspartate aminotransferase (ALT and AST). Furthermore, liver lipidomics indicated that C3G effectively facilitated the recovery of differential lipid metabolites, including TGs, phosphatidylethanolamines, phosphatidylcholines, lysophosphatidylcholines, phosphatidylinositol, cholesteryl esters, and phosphatidylserine, and reversed the levels of hepatic lipid synthesis-related genes. Our results suggest that C3G has an effective regulatory effect on BPA-induced disorders of lipid metabolism.
Collapse
Affiliation(s)
- Ruijing Liu
- Department of Food Science and Engineering, Institute of Food Safety and Nutrition, Guangdong Engineering Technology Center of Food Safety Molecular Rapid Detection, Jinan University, Guangzhou 510632, P. R. China
- Key Laboratory for Bio-Based Materials and Energy of Ministry of Education, College of Materials and Energy, South China Agricultural University, Guangzhou 510642, P. R. China
| | - Yulong Jin
- Key Laboratory for Bio-Based Materials and Energy of Ministry of Education, College of Materials and Energy, South China Agricultural University, Guangzhou 510642, P. R. China
| | - Boping Liu
- Key Laboratory for Bio-Based Materials and Energy of Ministry of Education, College of Materials and Energy, South China Agricultural University, Guangzhou 510642, P. R. China
| | - Qing Zhang
- Department of Food Science and Engineering, Institute of Food Safety and Nutrition, Guangdong Engineering Technology Center of Food Safety Molecular Rapid Detection, Jinan University, Guangzhou 510632, P. R. China
| | - Xusheng Li
- Department of Food Science and Engineering, Institute of Food Safety and Nutrition, Guangdong Engineering Technology Center of Food Safety Molecular Rapid Detection, Jinan University, Guangzhou 510632, P. R. China
| | - Dongbao Cai
- Department of Food Science and Engineering, Institute of Food Safety and Nutrition, Guangdong Engineering Technology Center of Food Safety Molecular Rapid Detection, Jinan University, Guangzhou 510632, P. R. China
| | - Lingmin Tian
- Department of Food Science and Engineering, Institute of Food Safety and Nutrition, Guangdong Engineering Technology Center of Food Safety Molecular Rapid Detection, Jinan University, Guangzhou 510632, P. R. China
| | - Xinwei Jiang
- Department of Food Science and Engineering, Institute of Food Safety and Nutrition, Guangdong Engineering Technology Center of Food Safety Molecular Rapid Detection, Jinan University, Guangzhou 510632, P. R. China
| | - Wenbao Zhang
- Department of Food Science and Engineering, Institute of Food Safety and Nutrition, Guangdong Engineering Technology Center of Food Safety Molecular Rapid Detection, Jinan University, Guangzhou 510632, P. R. China
| | - Jianxia Sun
- School of Chemical Engineering and Light Industry, Guangdong University of Technology, Guangzhou 510006, P. R. China
| | - Weibin Bai
- Department of Food Science and Engineering, Institute of Food Safety and Nutrition, Guangdong Engineering Technology Center of Food Safety Molecular Rapid Detection, Jinan University, Guangzhou 510632, P. R. China
| |
Collapse
|
31
|
Gao L, He Z, Wu Y. Advances in Anti-metabolic Disease Treatments Targeting CD47. Curr Pharm Des 2022; 28:3720-3728. [PMID: 36201266 DOI: 10.2174/1381612828666221006123144] [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: 05/26/2022] [Revised: 08/15/2022] [Accepted: 08/21/2022] [Indexed: 01/28/2023]
Abstract
Metabolic disorders include a cluster of conditions that result from hyperglycemia, hyperlipidemia, insulin resistance, obesity, and hepatic steatosis, which cause the dysfunction of immune cells and innate cells, such as macrophages, natural killer cells, vascular endothelial cells, hepatocytes, and human kidney tubular epithelial cells. Besides targeting the derangements in lipid metabolism, therapeutic modulations to regulate abnormal responses in the immune system and innate cell dysfunctions may prove to be promising strategies in the management of metabolic diseases. In recent years, several targets have been explored for the CD47 molecule (CD47), a glycosylated protein, which was originally reported to transmit an anti-phagocytic signal known as "don't eat me" in the atherosclerotic environment, hindering the efferocytosis of immune cells and promoting arterial plaque accumulation. Subsequently, the role of CD47 has been explored in obesity, fatty liver, and lipotoxic nephropathy, and its utility as a therapeutic target has been investigated using anti-CD47 antibodies or inhibitors of the THBS1/CD47 axis and the CD47/SIRPα signaling pathway. This review summarizes the mechanisms of action of CD47 in different cell types during metabolic diseases and the clinical research progress to date, providing a reference for the comprehensive targeting of CD47 to treat metabolic diseases and the devising of potential improvements to possible side effects.
Collapse
Affiliation(s)
- Li Gao
- Department of Nephropathy, The First Affiliated Hospital of Anhui Medical University, Hefei 230022, China.,Center for Scientific Research of Anhui Medical University, Hefei 230022, China
| | - Zhe He
- Department of Nephropathy, The First Affiliated Hospital of Anhui Medical University, Hefei 230022, China
| | - Yonggui Wu
- Department of Nephropathy, The First Affiliated Hospital of Anhui Medical University, Hefei 230022, China.,Center for Scientific Research of Anhui Medical University, Hefei 230022, China
| |
Collapse
|
32
|
Sakuma T, Nakamura M, Chiba T, Iwanaga T, Kan M, Kojima R, Ao J, Ma Y, Unozawa H, Fujita N, Kanayama K, Kanzaki H, Koroki K, Kobayashi K, Nakagawa R, Kanogawa N, Kiyono S, Kondo T, Saito T, Ogasawara S, Nakamoto S, Muroyama R, Kato J, Kishimoto T, Kato N. A diet-induced murine model for non-alcoholic fatty liver disease with obesity and insulin resistance that rapidly develops steatohepatitis and fibrosis. J Transl Med 2022; 102:1150-1157. [PMID: 35643859 DOI: 10.1038/s41374-022-00807-6] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Revised: 05/08/2022] [Accepted: 05/09/2022] [Indexed: 11/09/2022] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) has become the leading cause of chronic liver disease worldwide. Patients with NAFLD often suffer steatohepatitis, which can progress to cirrhosis and hepatocellular carcinoma. The presence of visceral obesity or type 2 diabetes mellitus (T2DM) is a major risk factor and potential therapeutic target for NAFLD. The establishment of animal models with these metabolic comorbidities and with the rapid progression of the disease is needed for developing treatments for NAFLD but remains to be archived. In the present study, KK-Ay mice, widely used as T2DM models, or C57BL6 mice were fed a high-fat, high-fructose, and high-cholesterol diet supplemented with cholic acid (NAFLD diet). The KK-Ay mice fed a NAFLD diet exhibited remarkable obesity and insulin resistance. A prominent accumulation of triglycerides and cholesterol in the liver was observed at 4 weeks. These mice developed steatohepatitis at 4 weeks and fibrosis at 12 weeks. In contrast, C57BL6 mice fed a NAFLD diet remained lean, although they still developed steatohepatitis and fibrosis. In summary, we established a diet-induced murine NAFLD model with the rapid development of steatohepatitis and fibrosis, bearing obesity and insulin resistance. This model could be useful as preclinical models for drug development of NAFLD.
Collapse
Affiliation(s)
- Takafumi Sakuma
- Department of Gastroenterology, Chiba University, Graduate School of Medicine, Chiba, 260-8677, Japan
| | - Masato Nakamura
- Department of Gastroenterology, Chiba University, Graduate School of Medicine, Chiba, 260-8677, Japan.
| | - Tetsuhiro Chiba
- Department of Gastroenterology, Chiba University, Graduate School of Medicine, Chiba, 260-8677, Japan
| | - Terunao Iwanaga
- Department of Gastroenterology, Chiba University, Graduate School of Medicine, Chiba, 260-8677, Japan
| | - Motoyasu Kan
- Department of Gastroenterology, Chiba University, Graduate School of Medicine, Chiba, 260-8677, Japan
| | - Ryuta Kojima
- Department of Gastroenterology, Chiba University, Graduate School of Medicine, Chiba, 260-8677, Japan
| | - Junjie Ao
- Department of Gastroenterology, Chiba University, Graduate School of Medicine, Chiba, 260-8677, Japan
| | - Yaojia Ma
- Department of Gastroenterology, Chiba University, Graduate School of Medicine, Chiba, 260-8677, Japan
| | - Hidemi Unozawa
- Department of Gastroenterology, Chiba University, Graduate School of Medicine, Chiba, 260-8677, Japan
| | - Naoto Fujita
- Department of Gastroenterology, Chiba University, Graduate School of Medicine, Chiba, 260-8677, Japan
| | - Kengo Kanayama
- Department of Gastroenterology, Chiba University, Graduate School of Medicine, Chiba, 260-8677, Japan
| | - Hiroaki Kanzaki
- Department of Gastroenterology, Chiba University, Graduate School of Medicine, Chiba, 260-8677, Japan
| | - Keisuke Koroki
- Department of Gastroenterology, Chiba University, Graduate School of Medicine, Chiba, 260-8677, Japan
| | - Kazufumi Kobayashi
- Department of Gastroenterology, Chiba University, Graduate School of Medicine, Chiba, 260-8677, Japan.,Translational Research and Development Center, Chiba University Hospital, Chiba, 260-8677, Japan
| | - Ryo Nakagawa
- Department of Gastroenterology, Chiba University, Graduate School of Medicine, Chiba, 260-8677, Japan
| | - Naoya Kanogawa
- Department of Gastroenterology, Chiba University, Graduate School of Medicine, Chiba, 260-8677, Japan
| | - Soichiro Kiyono
- Department of Gastroenterology, Chiba University, Graduate School of Medicine, Chiba, 260-8677, Japan
| | - Takayuki Kondo
- Department of Gastroenterology, Chiba University, Graduate School of Medicine, Chiba, 260-8677, Japan
| | - Tomoko Saito
- Department of Gastroenterology, Chiba University, Graduate School of Medicine, Chiba, 260-8677, Japan
| | - Sadahisa Ogasawara
- Department of Gastroenterology, Chiba University, Graduate School of Medicine, Chiba, 260-8677, Japan.,Translational Research and Development Center, Chiba University Hospital, Chiba, 260-8677, Japan
| | - Shingo Nakamoto
- Department of Gastroenterology, Chiba University, Graduate School of Medicine, Chiba, 260-8677, Japan
| | - Ryosuke Muroyama
- Department of Molecular Virology, Chiba University, Graduate School of Medicine, Chiba, 260-8677, Japan
| | - Jun Kato
- Department of Gastroenterology, Chiba University, Graduate School of Medicine, Chiba, 260-8677, Japan
| | - Takashi Kishimoto
- Department of Molecular Pathology, Chiba University, Graduate School of Medicine, Chiba, 260-8677, Japan
| | - Naoya Kato
- Department of Gastroenterology, Chiba University, Graduate School of Medicine, Chiba, 260-8677, Japan
| |
Collapse
|
33
|
Martínez‐Arranz I, Bruzzone C, Noureddin M, Gil‐Redondo R, Mincholé I, Bizkarguenaga M, Arretxe E, Iruarrizaga‐Lejarreta M, Fernández‐Ramos D, Lopitz‐Otsoa F, Mayo R, Embade N, Newberry E, Mittendorf B, Izquierdo‐Sánchez L, Smid V, Arnold J, Iruzubieta P, Pérez Castaño Y, Krawczyk M, Marigorta UM, Morrison MC, Kleemann R, Martín‐Duce A, Hayardeny L, Vitek L, Bruha R, Aller de la Fuente R, Crespo J, Romero‐Gomez M, Banales JM, Arrese M, Cusi K, Bugianesi E, Klein S, Lu SC, Anstee QM, Millet O, Davidson NO, Alonso C, Mato JM. Metabolic subtypes of patients with NAFLD exhibit distinctive cardiovascular risk profiles. Hepatology 2022; 76:1121-1134. [PMID: 35220605 PMCID: PMC9790568 DOI: 10.1002/hep.32427] [Citation(s) in RCA: 39] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/11/2021] [Revised: 01/18/2022] [Accepted: 02/15/2022] [Indexed: 12/31/2022]
Abstract
BACKGROUND AND AIMS We previously identified subsets of patients with NAFLD with different metabolic phenotypes. Here we align metabolomic signatures with cardiovascular disease (CVD) and genetic risk factors. APPROACH AND RESULTS We analyzed serum metabolome from 1154 individuals with biopsy-proven NAFLD, and from four mouse models of NAFLD with impaired VLDL-triglyceride (TG) secretion, and one with normal VLDL-TG secretion. We identified three metabolic subtypes: A (47%), B (27%), and C (26%). Subtype A phenocopied the metabolome of mice with impaired VLDL-TG secretion; subtype C phenocopied the metabolome of mice with normal VLDL-TG; and subtype B showed an intermediate signature. The percent of patients with NASH and fibrosis was comparable among subtypes, although subtypes B and C exhibited higher liver enzymes. Serum VLDL-TG levels and secretion rate were lower among subtype A compared with subtypes B and C. Subtype A VLDL-TG and VLDL-apolipoprotein B concentrations were independent of steatosis, whereas subtypes B and C showed an association with these parameters. Serum TG, cholesterol, VLDL, small dense LDL5,6 , and remnant lipoprotein cholesterol were lower among subtype A compared with subtypes B and C. The 10-year high risk of CVD, measured with the Framingham risk score, and the frequency of patatin-like phospholipase domain-containing protein 3 NAFLD risk allele were lower in subtype A. CONCLUSIONS Metabolomic signatures identify three NAFLD subgroups, independent of histological disease severity. These signatures align with known CVD and genetic risk factors, with subtype A exhibiting a lower CVD risk profile. This may account for the variation in hepatic versus cardiovascular outcomes, offering clinically relevant risk stratification.
Collapse
Affiliation(s)
| | | | - Mazen Noureddin
- Karsh Division of Gastroenterology and HepatologyCedars‐Sinai Medical CenterLos AngelesCaliforniaUSA
| | | | | | | | | | | | | | | | | | | | - Elizabeth Newberry
- Department of MedicineWashington University School of MedicineSt. LouisMissouriUSA
| | - Bettina Mittendorf
- Center for Human NutritionWashington University School of MedicineSt. LouisMissouriUSA
| | - Laura Izquierdo‐Sánchez
- Department of Liver and Gastrointestinal DiseasesBiodonostia Research InstituteDonostia University HospitalDonostiaSpain
| | - Vaclav Smid
- First Faculty of MedicineCharles UniversityPragueCzech Republic
| | - Jorge Arnold
- Departamento de GastroenterologiaEscuela de MedicinaPontificia Universidad Católica de ChileSantiago de ChileChile
| | - Paula Iruzubieta
- Marqués de Valdecilla University HospitalCantabria UniversitySantanderSpain
| | - Ylenia Pérez Castaño
- Department of Digestive SystemOsakidetza Basque Health ServiceDonostia University HospitalSan SebastianSpain
| | - Marcin Krawczyk
- Department of Medicine IISaarland University Medical CenterHomburgGermany
- Laboratory of Metabolic Liver DiseasesCenter for Preclinical ResearchDepartment of General, Transplant and Liver SurgeryMedical University of WarsawWarsawPoland
| | | | - Martine C. Morrison
- Department of Metabolic Health ResearchNetherlands Organization for Applied Scientific ResearchLeidenThe Netherlands
| | - Robert Kleemann
- Department of Metabolic Health ResearchNetherlands Organization for Applied Scientific ResearchLeidenThe Netherlands
| | - Antonio Martín‐Duce
- Alcalá University School of Medicine and Health SciencesUniversity Hospital Prıncipe de AsturiasMadridSpain
| | | | - Libor Vitek
- First Faculty of MedicineCharles UniversityPragueCzech Republic
| | - Radan Bruha
- First Faculty of MedicineCharles UniversityPragueCzech Republic
| | - Rocío Aller de la Fuente
- Department of Digestive DiseaseClinic University HospitalUniversity Hospital of ValladolidValladolidSpain
| | - Javier Crespo
- Marqués de Valdecilla University HospitalCantabria UniversitySantanderSpain
| | | | - Jesus M Banales
- Department of Liver and Gastrointestinal DiseasesBiodonostia Research InstituteDonostia University HospitalDonostiaSpain
- University of the Basque CountryCIBERehdIKERBASQUEDonostiaSpain
| | - Marco Arrese
- Departamento de GastroenterologiaEscuela de MedicinaPontificia Universidad Católica de ChileSantiago de ChileChile
- Centro de Envejecimiento y RegeneraciónSantiagoChile
| | - Kenneth Cusi
- Division of Endocrinology, Diabetes and MetabolismUniversity of Florida and Malcom Randall VAMCGainesvilleFloridaUSA
| | | | - Samuel Klein
- Center for Human NutritionWashington University School of MedicineSt. LouisMissouriUSA
| | - Shelly C. Lu
- Karsh Division of Gastroenterology and HepatologyCedars‐Sinai Medical CenterLos AngelesCaliforniaUSA
| | - Quentin M. Anstee
- Translational & Clinical Research InstituteFaculty of Medical SciencesNewcastle UniversityNewcastle Upon TyneUK
- Newcastle NIHR Biomedical Research CenterNewcastle Upon Tyne Hospitals NHS TrustNewcastle Upon TyneUK
| | | | - Nicholas O. Davidson
- Department of MedicineWashington University School of MedicineSt. LouisMissouriUSA
| | | | | |
Collapse
|
34
|
Guidance for the diagnosis and treatment of hypolipidemia disorders. J Clin Lipidol 2022; 16:797-812. [PMID: 36243606 DOI: 10.1016/j.jacl.2022.08.009] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2022] [Accepted: 08/31/2022] [Indexed: 11/15/2022]
|
35
|
Lin H, Wang L, Liu Z, Long K, Kong M, Ye D, Chen X, Wang K, Wu KKL, Fan M, Song E, Wang C, Hoo RLC, Hui X, Hallenborg P, Piao H, Xu A, Cheng KKY. Hepatic MDM2 Causes Metabolic Associated Fatty Liver Disease by Blocking Triglyceride-VLDL Secretion via ApoB Degradation. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2022; 9:e2200742. [PMID: 35524581 PMCID: PMC9284139 DOI: 10.1002/advs.202200742] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Revised: 03/15/2022] [Indexed: 05/06/2023]
Abstract
Dysfunctional triglyceride-very low-density lipoprotein (TG-VLDL) metabolism is linked to metabolic-associated fatty liver disease (MAFLD); however, the underlying cause remains unclear. The study shows that hepatic E3 ubiquitin ligase murine double minute 2 (MDM2) controls MAFLD by blocking TG-VLDL secretion. A remarkable upregulation of MDM2 is observed in the livers of human and mouse models with different levels of severity of MAFLD. Hepatocyte-specific deletion of MDM2 protects against high-fat high-cholesterol diet-induced hepatic steatosis and inflammation, accompanied by a significant elevation in TG-VLDL secretion. As an E3 ubiquitin ligase, MDM2 targets apolipoprotein B (ApoB) for proteasomal degradation through direct protein-protein interaction, which leads to reduced TG-VLDL secretion in hepatocytes. Pharmacological blockage of the MDM2-ApoB interaction alleviates dietary-induced hepatic steatohepatitis and fibrosis by inducing hepatic ApoB expression and subsequent TG-VLDL secretion. The effect of MDM2 on VLDL metabolism is p53-independent. Collectively, these findings suggest that MDM2 acts as a negative regulator of hepatic ApoB levels and TG-VLDL secretion in MAFLD. Inhibition of the MDM2-ApoB interaction may represent a potential therapeutic approach for MAFLD treatment.
Collapse
Affiliation(s)
- Huige Lin
- Department of Health Technology and InformaticsThe Hong Kong Polytechnic UniversityHung HomKowloonHong Kong
| | - Lin Wang
- Department of Health Technology and InformaticsThe Hong Kong Polytechnic UniversityHung HomKowloonHong Kong
- The State Key Laboratory of Pharmaceutical BiotechnologyThe University of Hong KongPokfulamHong Kong
- Department of MedicineThe University of Hong KongPokfulamHong Kong
| | - Zhuohao Liu
- The State Key Laboratory of Pharmaceutical BiotechnologyThe University of Hong KongPokfulamHong Kong
- Department of MedicineThe University of Hong KongPokfulamHong Kong
- Department of NeurosurgeryShenzhen HospitalSouthern Medical UniversityShenzhen518000P. R. China
| | - Kekao Long
- Department of Health Technology and InformaticsThe Hong Kong Polytechnic UniversityHung HomKowloonHong Kong
| | - Mengjie Kong
- Department of Health Technology and InformaticsThe Hong Kong Polytechnic UniversityHung HomKowloonHong Kong
| | - Dewei Ye
- Key Laboratory of Glucolipid Metabolic Diseases of the Ministry of EducationGuangdong Pharmaceutical UniversityGuangzhou510000P. R. China
| | - Xi Chen
- Department of Health Technology and InformaticsThe Hong Kong Polytechnic UniversityHung HomKowloonHong Kong
| | - Kai Wang
- Department of Health Technology and InformaticsThe Hong Kong Polytechnic UniversityHung HomKowloonHong Kong
| | - Kelvin KL Wu
- Department of Health Technology and InformaticsThe Hong Kong Polytechnic UniversityHung HomKowloonHong Kong
| | - Mengqi Fan
- Key Laboratory of Glucolipid Metabolic Diseases of the Ministry of EducationGuangdong Pharmaceutical UniversityGuangzhou510000P. R. China
| | - Erfei Song
- Department of Metabolic and Bariatric SurgeryThe First Affiliated Hospital of Jinan UniversityGuangzhou510000P. R. China
| | - Cunchuan Wang
- Department of Metabolic and Bariatric SurgeryThe First Affiliated Hospital of Jinan UniversityGuangzhou510000P. R. China
| | - Ruby LC Hoo
- The State Key Laboratory of Pharmaceutical BiotechnologyThe University of Hong KongPokfulamHong Kong
- Department of Pharmacology and PharmacyThe University of Hong KongPokfulamHong Kong
| | - Xiaoyan Hui
- The State Key Laboratory of Pharmaceutical BiotechnologyThe University of Hong KongPokfulamHong Kong
- Department of MedicineThe University of Hong KongPokfulamHong Kong
| | - Philip Hallenborg
- Department of Biochemistry and Molecular BiologyUniversity of Southern DenmarkSouthern Denmark5230Denmark
| | - Hailong Piao
- Dalian Institute of Chemical PhysicsChinese Academy of SciencesDalian116000P. R. China
| | - Aimin Xu
- The State Key Laboratory of Pharmaceutical BiotechnologyThe University of Hong KongPokfulamHong Kong
- Department of MedicineThe University of Hong KongPokfulamHong Kong
- Department of Pharmacology and PharmacyThe University of Hong KongPokfulamHong Kong
| | - Kenneth KY Cheng
- Department of Health Technology and InformaticsThe Hong Kong Polytechnic UniversityHung HomKowloonHong Kong
| |
Collapse
|
36
|
Kudo T, Sasaki K, Tada H. Familial hypobetalipoproteinemia caused by homozygous loss-of-function mutations in PCSK9: A case report. J Clin Lipidol 2022; 16:596-600. [DOI: 10.1016/j.jacl.2022.07.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2022] [Revised: 07/07/2022] [Accepted: 07/20/2022] [Indexed: 11/25/2022]
|
37
|
Vanhoye X, Janin A, Caillaud A, Rimbert A, Venet F, Gossez M, Dijk W, Marmontel O, Nony S, Chatelain C, Durand C, Lindenbaum P, Rieusset J, Cariou B, Moulin P, Di Filippo M. APOB CRISPR-Cas9 Engineering in Hypobetalipoproteinemia: A Promising Tool for Functional Studies of Novel Variants. Int J Mol Sci 2022; 23:4281. [PMID: 35457099 PMCID: PMC9030618 DOI: 10.3390/ijms23084281] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2022] [Revised: 04/04/2022] [Accepted: 04/11/2022] [Indexed: 02/01/2023] Open
Abstract
Hypobetalipoproteinemia is characterized by LDL-cholesterol and apolipoprotein B (apoB) plasma levels below the fifth percentile for age and sex. Familial hypobetalipoproteinemia (FHBL) is mostly caused by premature termination codons in the APOB gene, a condition associated with fatty liver and steatohepatitis. Nevertheless, many families with a FHBL phenotype carry APOB missense variants of uncertain significance (VUS). We here aimed to develop a proof-of-principle experiment to assess the pathogenicity of VUS using the genome editing of human liver cells. We identified a novel heterozygous APOB-VUS (p.Leu351Arg), in a FHBL family. We generated APOB knock-out (KO) and APOB-p.Leu351Arg knock-in Huh7 cells using CRISPR-Cas9 technology and studied the APOB expression, synthesis and secretion by digital droplet PCR and ELISA quantification. The APOB expression was decreased by 70% in the heterozygous APOB-KO cells and almost abolished in the homozygous-KO cells, with a consistent decrease in apoB production and secretion. The APOB-p.Leu351Arg homozygous cells presented with a 40% decreased APOB expression and undetectable apoB levels in cellular extracts and supernatant. Thus, the p.Leu351Arg affected the apoB secretion, which led us to classify this new variant as likely pathogenic and to set up a hepatic follow-up in this family. Therefore, the functional assessment of APOB-missense variants, using gene-editing technologies, will lead to improvements in the molecular diagnosis of FHBL and the personalized follow-up of these patients.
Collapse
Affiliation(s)
- Xavier Vanhoye
- Service de Biochimie et de Biologie Moléculaire, Laboratoire de Biologie Médicale MultiSites, Hospices Civils de Lyon, F-69677 Bron, France; (X.V.); (A.J.); (O.M.); (S.N.); (C.C.)
| | - Alexandre Janin
- Service de Biochimie et de Biologie Moléculaire, Laboratoire de Biologie Médicale MultiSites, Hospices Civils de Lyon, F-69677 Bron, France; (X.V.); (A.J.); (O.M.); (S.N.); (C.C.)
- Institut NeuroMyoGène, CNRS UMR5310, INSERM U1217, Université Claude Bernard Lyon 1, Université de Lyon, F-69008 Lyon, France
| | - Amandine Caillaud
- Institut du Thorax, Nantes Université, CHU Nantes, CNRS, INSERM, F-44000 Nantes, France; (A.C.); (B.C.)
| | - Antoine Rimbert
- Institut du Thorax, Nantes Université, CNRS, INSERM, F-44000 Nantes, France; (A.R.); (W.D.); (P.L.)
| | - Fabienne Venet
- Laboratoire d’Immunologie, Edouard Herriot Hospital, Hospices Civils de Lyon, F-69437 Lyon, France; (F.V.); (M.G.)
- Centre International de Recherche en Infectiologie (CIRI), INSERM U1111, CNRS, UMR5308, Ecole Normale Supérieure de Lyon, Université Claude Bernard-Lyon 1, F-69364 Lyon, France
| | - Morgane Gossez
- Laboratoire d’Immunologie, Edouard Herriot Hospital, Hospices Civils de Lyon, F-69437 Lyon, France; (F.V.); (M.G.)
- Centre International de Recherche en Infectiologie (CIRI), INSERM U1111, CNRS, UMR5308, Ecole Normale Supérieure de Lyon, Université Claude Bernard-Lyon 1, F-69364 Lyon, France
| | - Wieneke Dijk
- Institut du Thorax, Nantes Université, CNRS, INSERM, F-44000 Nantes, France; (A.R.); (W.D.); (P.L.)
| | - Oriane Marmontel
- Service de Biochimie et de Biologie Moléculaire, Laboratoire de Biologie Médicale MultiSites, Hospices Civils de Lyon, F-69677 Bron, France; (X.V.); (A.J.); (O.M.); (S.N.); (C.C.)
- CarMen Laboratory, INSERM, INRA, INSA Lyon, Université Claude Bernard Lyon 1, Pierre-Bénite, F-69364 Lyon, France; (C.D.); (J.R.); (P.M.)
| | - Séverine Nony
- Service de Biochimie et de Biologie Moléculaire, Laboratoire de Biologie Médicale MultiSites, Hospices Civils de Lyon, F-69677 Bron, France; (X.V.); (A.J.); (O.M.); (S.N.); (C.C.)
| | - Charlotte Chatelain
- Service de Biochimie et de Biologie Moléculaire, Laboratoire de Biologie Médicale MultiSites, Hospices Civils de Lyon, F-69677 Bron, France; (X.V.); (A.J.); (O.M.); (S.N.); (C.C.)
| | - Christine Durand
- CarMen Laboratory, INSERM, INRA, INSA Lyon, Université Claude Bernard Lyon 1, Pierre-Bénite, F-69364 Lyon, France; (C.D.); (J.R.); (P.M.)
| | - Pierre Lindenbaum
- Institut du Thorax, Nantes Université, CNRS, INSERM, F-44000 Nantes, France; (A.R.); (W.D.); (P.L.)
| | - Jennifer Rieusset
- CarMen Laboratory, INSERM, INRA, INSA Lyon, Université Claude Bernard Lyon 1, Pierre-Bénite, F-69364 Lyon, France; (C.D.); (J.R.); (P.M.)
| | - Bertrand Cariou
- Institut du Thorax, Nantes Université, CHU Nantes, CNRS, INSERM, F-44000 Nantes, France; (A.C.); (B.C.)
| | - Philippe Moulin
- CarMen Laboratory, INSERM, INRA, INSA Lyon, Université Claude Bernard Lyon 1, Pierre-Bénite, F-69364 Lyon, France; (C.D.); (J.R.); (P.M.)
- Fédération d’Endocrinologie, Maladies Métaboliques, Diabète et Nutrition, Hôpital Louis Pradel, Hospices Civils de Lyon, F-69677 Bron, France
| | - Mathilde Di Filippo
- Service de Biochimie et de Biologie Moléculaire, Laboratoire de Biologie Médicale MultiSites, Hospices Civils de Lyon, F-69677 Bron, France; (X.V.); (A.J.); (O.M.); (S.N.); (C.C.)
- CarMen Laboratory, INSERM, INRA, INSA Lyon, Université Claude Bernard Lyon 1, Pierre-Bénite, F-69364 Lyon, France; (C.D.); (J.R.); (P.M.)
| |
Collapse
|
38
|
Welty FK. Familial hypobetalipoproteinemia and abetalipoproteinemia. CHOLESTEROL 2022:465-480. [DOI: 10.1016/b978-0-323-85857-1.00026-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
39
|
Haas ME, Pirruccello JP, Friedman SN, Wang M, Emdin CA, Ajmera VH, Simon TG, Homburger JR, Guo X, Budoff M, Corey KE, Zhou AY, Philippakis A, Ellinor PT, Loomba R, Batra P, Khera AV. Machine learning enables new insights into genetic contributions to liver fat accumulation. CELL GENOMICS 2021; 1:100066. [PMID: 34957434 PMCID: PMC8699145 DOI: 10.1016/j.xgen.2021.100066] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/12/2021] [Revised: 07/13/2021] [Accepted: 09/09/2021] [Indexed: 12/14/2022]
Abstract
Excess liver fat, called hepatic steatosis, is a leading risk factor for end-stage liver disease and cardiometabolic diseases but often remains undiagnosed in clinical practice because of the need for direct imaging assessments. We developed an abdominal MRI-based machine-learning algorithm to accurately estimate liver fat (correlation coefficients, 0.97-0.99) from a truth dataset of 4,511 middle-aged UK Biobank participants, enabling quantification in 32,192 additional individuals. 17% of participants had predicted liver fat levels indicative of steatosis, and liver fat could not have been reliably estimated based on clinical factors such as BMI. A genome-wide association study of common genetic variants and liver fat replicated three known associations and identified five newly associated variants in or near the MTARC1, ADH1B, TRIB1, GPAM, and MAST3 genes (p < 3 × 10-8). A polygenic score integrating these eight genetic variants was strongly associated with future risk of chronic liver disease (hazard ratio > 1.32 per SD score, p < 9 × 10-17). Rare inactivating variants in the APOB or MTTP genes were identified in 0.8% of individuals with steatosis and conferred more than 6-fold risk (p < 2 × 10-5), highlighting a molecular subtype of hepatic steatosis characterized by defective secretion of apolipoprotein B-containing lipoproteins. We demonstrate that our imaging-based machine-learning model accurately estimates liver fat and may be useful in epidemiological and genetic studies of hepatic steatosis.
Collapse
Affiliation(s)
- Mary E. Haas
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
- Department of Molecular Biology, Department of Medicine, Massachusetts General Hospital, Boston, MA 02114, USA
| | - James P. Pirruccello
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
- Department of Medicine, Harvard Medical School, Boston, MA 02114, USA
- Cardiology Division, Department of Medicine, Massachusetts General Hospital, Boston, MA 02114, USA
- Machine Learning for Health, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Samuel N. Friedman
- Machine Learning for Health, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Minxian Wang
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Connor A. Emdin
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
- Department of Medicine, Harvard Medical School, Boston, MA 02114, USA
| | - Veeral H. Ajmera
- NAFLD Research Center, Department of Medicine, University of California San Diego, La Jolla, CA 92103, USA
| | - Tracey G. Simon
- Department of Medicine, Harvard Medical School, Boston, MA 02114, USA
- Liver Center, Division of Gastroenterology, Department of Medicine, Massachusetts General Hospital, Boston, MA 02114, USA
| | | | - Xiuqing Guo
- The Lundquist Institute for Biomedical Innovation and Department of Pediatrics, Harbor-UCLA Medical Center, Torrance, CA 90502, USA
| | - Matthew Budoff
- The Lundquist Institute for Biomedical Innovation and Department of Pediatrics, Harbor-UCLA Medical Center, Torrance, CA 90502, USA
| | - Kathleen E. Corey
- Department of Medicine, Harvard Medical School, Boston, MA 02114, USA
- Liver Center, Division of Gastroenterology, Department of Medicine, Massachusetts General Hospital, Boston, MA 02114, USA
| | | | - Anthony Philippakis
- Machine Learning for Health, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
- Eric and Wendy Schmidt Center, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Patrick T. Ellinor
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
- Department of Medicine, Harvard Medical School, Boston, MA 02114, USA
- Cardiology Division, Department of Medicine, Massachusetts General Hospital, Boston, MA 02114, USA
- Machine Learning for Health, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Rohit Loomba
- NAFLD Research Center, Department of Medicine, University of California San Diego, La Jolla, CA 92103, USA
| | - Puneet Batra
- Machine Learning for Health, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Amit V. Khera
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
- Center for Genomic Medicine, Department of Medicine, Massachusetts General Hospital, Boston, MA 02114, USA
- Department of Medicine, Harvard Medical School, Boston, MA 02114, USA
- Cardiology Division, Department of Medicine, Massachusetts General Hospital, Boston, MA 02114, USA
- Machine Learning for Health, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| |
Collapse
|
40
|
Dongiovanni P, Meroni M, Longo M, Fargion S, Fracanzani AL. Genetics, Immunity and Nutrition Boost the Switching from NASH to HCC. Biomedicines 2021; 9:1524. [PMID: 34829753 PMCID: PMC8614742 DOI: 10.3390/biomedicines9111524] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Revised: 10/20/2021] [Accepted: 10/22/2021] [Indexed: 12/12/2022] Open
Abstract
Nonalcoholic fatty liver disease (NAFLD) is the leading contributor to the global burden of chronic liver diseases. The phenotypic umbrella of NAFLD spans from simple and reversible steatosis to nonalcoholic steatohepatitis (NASH), which may worsen into cirrhosis and hepatocellular carcinoma (HCC). Notwithstanding, HCC may develop also in the absence of advanced fibrosis, causing a delayed time in diagnosis as a consequence of the lack of HCC screening in these patients. The precise event cascade that may precipitate NASH into HCC is intricate and it entails diverse triggers, encompassing exaggerated immune response, endoplasmic reticulum (ER) and oxidative stress, organelle derangement and DNA aberrancies. All these events may be accelerated by both genetic and environmental factors. On one side, common and rare inherited variations that affect hepatic lipid remodeling, immune microenvironment and cell survival may boost the switching from steatohepatitis to liver cancer, on the other, diet-induced dysbiosis as well as nutritional and behavioral habits may furtherly precipitate tumor onset. Therefore, dietary and lifestyle interventions aimed to restore patients' health contribute to counteract NASH progression towards HCC. Even more, the combination of therapeutic strategies with dietary advice may maximize benefits, with the pursuit to improve liver function and prolong survival.
Collapse
Affiliation(s)
- Paola Dongiovanni
- General Medicine and Metabolic Diseases, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, Pad. Granelli, 20122 Milan, Italy; (M.M.); (M.L.); (S.F.); (A.L.F.)
| | - Marica Meroni
- General Medicine and Metabolic Diseases, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, Pad. Granelli, 20122 Milan, Italy; (M.M.); (M.L.); (S.F.); (A.L.F.)
| | - Miriam Longo
- General Medicine and Metabolic Diseases, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, Pad. Granelli, 20122 Milan, Italy; (M.M.); (M.L.); (S.F.); (A.L.F.)
- Department of Clinical Sciences and Community Health, Università degli Studi di Milano, 20122 Milan, Italy
| | - Silvia Fargion
- General Medicine and Metabolic Diseases, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, Pad. Granelli, 20122 Milan, Italy; (M.M.); (M.L.); (S.F.); (A.L.F.)
| | - Anna Ludovica Fracanzani
- General Medicine and Metabolic Diseases, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, Pad. Granelli, 20122 Milan, Italy; (M.M.); (M.L.); (S.F.); (A.L.F.)
- Department of Pathophysiology and Transplantation, Università degli Studi di Milano, 20122 Milan, Italy
| |
Collapse
|
41
|
Meroni M, Longo M, Tria G, Dongiovanni P. Genetics Is of the Essence to Face NAFLD. Biomedicines 2021; 9:1359. [PMID: 34680476 PMCID: PMC8533437 DOI: 10.3390/biomedicines9101359] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2021] [Accepted: 09/27/2021] [Indexed: 02/07/2023] Open
Abstract
Nonalcoholic fatty liver disease (NAFLD) is the commonest cause of chronic liver disease worldwide. It is closely related to obesity, insulin resistance (IR) and dyslipidemia so much so it is considered the hepatic manifestation of the Metabolic Syndrome. The NAFLD spectrum extends from simple steatosis to nonalcoholic steatohepatitis (NASH), a clinical condition which may progress up to fibrosis, cirrhosis and hepatocellular carcinoma (HCC). NAFLD is a complex disease whose pathogenesis is shaped by both environmental and genetic factors. In the last two decades, several heritable modifications in genes influencing hepatic lipid remodeling, and mitochondrial oxidative status have been emerged as predictors of progressive hepatic damage. Among them, the patatin-like phospholipase domain-containing 3 (PNPLA3) p.I148M, the Transmembrane 6 superfamily member 2 (TM6SF2) p.E167K and the rs641738 membrane bound-o-acyltransferase domain-containing 7 (MBOAT7) polymorphisms are considered the most robust modifiers of NAFLD. However, a forefront frontier in the study of NAFLD heritability is to postulate score-based strategy, building polygenic risk scores (PRS), which aggregate the most relevant genetic determinants of NAFLD and biochemical parameters, with the purpose to foresee patients with greater risk of severe NAFLD, guaranteeing the most highly predictive value, the best diagnostic accuracy and the more precise individualized therapy.
Collapse
Affiliation(s)
- Marica Meroni
- General Medicine and Metabolic Diseases, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, Pad. Granelli, Via F Sforza 35, 20122 Milan, Italy; (M.M.); (M.L.); (G.T.)
| | - Miriam Longo
- General Medicine and Metabolic Diseases, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, Pad. Granelli, Via F Sforza 35, 20122 Milan, Italy; (M.M.); (M.L.); (G.T.)
- Department of Clinical Sciences and Community Health, Università Degli Studi di Milano, 20122 Milano, Italy
| | - Giada Tria
- General Medicine and Metabolic Diseases, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, Pad. Granelli, Via F Sforza 35, 20122 Milan, Italy; (M.M.); (M.L.); (G.T.)
| | - Paola Dongiovanni
- General Medicine and Metabolic Diseases, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, Pad. Granelli, Via F Sforza 35, 20122 Milan, Italy; (M.M.); (M.L.); (G.T.)
| |
Collapse
|
42
|
Newberry EP, Hall Z, Xie Y, Molitor EA, Bayguinov PO, Strout GW, Fitzpatrick JA, Brunt EM, Griffin JL, Davidson NO. Liver-Specific Deletion of Mouse Tm6sf2 Promotes Steatosis, Fibrosis, and Hepatocellular Cancer. Hepatology 2021; 74:1203-1219. [PMID: 33638902 PMCID: PMC8390580 DOI: 10.1002/hep.31771] [Citation(s) in RCA: 69] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Revised: 12/17/2020] [Accepted: 01/13/2021] [Indexed: 12/11/2022]
Abstract
BACKGROUND AND AIMS Human transmembrane 6 superfamily 2 (TM6SF2) variant rs58542926 is associated with NAFLD and HCC. However, conflicting reports in germline Tm6sf2 knockout mice suggest no change or decreased very low density lipoprotein (VLDL) secretion and either unchanged or increased hepatic steatosis, with no increased fibrosis. We generated liver-specific Tm6Sf2 knockout mice (Tm6 LKO) to study VLDL secretion and the impact on development and progression of NAFLD. APPROACH AND RESULTS Two independent lines of Tm6 LKO mice exhibited spontaneous hepatic steatosis. Targeted lipidomic analyses showed increased triglyceride species whose distribution and abundance phenocopied findings in mice with liver-specific deletion of microsomal triglyceride transfer protein. The VLDL triglyceride secretion was reduced with small, underlipidated particles and unchanged or increased apolipoprotein B. Liver-specific adeno-associated viral, serotype 8 (AAV8) rescue using either wild-type or mutant E167K-Tm6 reduced hepatic steatosis and improved VLDL secretion. The Tm6 LKO mice fed a high milk-fat diet for 3 weeks exhibited increased steatosis and fibrosis, and those phenotypes were further exacerbated when mice were fed fibrogenic, high fat/fructose diets for 20 weeks. In two models of HCC, either neonatal mice injected with streptozotocin (NASH/STAM) and high-fat fed or with diethylnitrosamine injection plus fibrogenic diet feeding, Tm6 LKO mice exhibited increased steatosis, greater tumor burden, and increased tumor area versus Tm6 flox controls. Additionally, diethylnitrosamine-injected and fibrogenic diet-fed Tm6 LKO mice administered wild-type Tm6 or E167K-mutant Tm6 AAV8 revealed significant tumor attenuation, with tumor burden inversely correlated with Tm6 protein levels. CONCLUSIONS Liver-specific Tm6sf2 deletion impairs VLDL secretion, promoting hepatic steatosis, fibrosis, and accelerated development of HCC, which was mitigated with AAV8- mediated rescue.
Collapse
Affiliation(s)
- Elizabeth P. Newberry
- Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110
| | - Zoe Hall
- Department of Biochemistry and Cambridge Systems Biology Centre, University of Cambridge, Cambridge, United Kingdom
- Biomolecular Medicine, Division of Systems Medicine, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, United Kingdom
| | - Yan Xie
- Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110
| | - Elizabeth A. Molitor
- Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110
| | - Peter O. Bayguinov
- Washington University Center for Cellular Imaging, Washington University in Saint Louis, St. Louis, MO 63130
| | - Gregory W. Strout
- Washington University Center for Cellular Imaging, Washington University in Saint Louis, St. Louis, MO 63130
| | - James A.J. Fitzpatrick
- Washington University Center for Cellular Imaging, Washington University in Saint Louis, St. Louis, MO 63130
- Departments of Cell Biology & Physiology and Neuroscience, Washington University School of Medicine, Louis, St. Louis, MO 63130
- Department of Biomedical Engineering, Washington University in Saint Louis, St. Louis, MO 63130
| | - Elizabeth M. Brunt
- Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110
| | - Julian L. Griffin
- Department of Biochemistry and Cambridge Systems Biology Centre, University of Cambridge, Cambridge, United Kingdom
- Biomolecular Medicine, Division of Systems Medicine, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, United Kingdom
| | - Nicholas O. Davidson
- Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110
| |
Collapse
|
43
|
Strong A, Ganetzky R, Rader DJ. Hepatic Manifestations of Mendelian Disorders of Cholesterol Biosynthesis and Cellular Metabolism. Clin Liver Dis (Hoboken) 2021; 18:266-273. [PMID: 34976370 PMCID: PMC8688899 DOI: 10.1002/cld.1154] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/10/2021] [Revised: 06/16/2021] [Accepted: 07/11/2021] [Indexed: 02/04/2023] Open
Abstract
Content available: Audio Recording.
Collapse
Affiliation(s)
- Alanna Strong
- Division of Human GeneticsChildren's Hospital of PhiladelphiaPhiladelphiaPA,The Center for Applied GenomicsChildren's Hospital of PhiladelphiaPhiladelphiaPA
| | - Rebecca Ganetzky
- Division of Human GeneticsChildren's Hospital of PhiladelphiaPhiladelphiaPA,Department of MedicinePerelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPA
| | - Daniel J. Rader
- Division of Human GeneticsChildren's Hospital of PhiladelphiaPhiladelphiaPA,Department of GeneticsPerelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPA,Department of MedicinePerelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPA,Institute for Translational Medicine and TherapeuticsPerelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPA
| |
Collapse
|
44
|
Templehof H, Moshe N, Avraham-Davidi I, Yaniv K. Zebrafish mutants provide insights into Apolipoprotein B functions during embryonic development and pathological conditions. JCI Insight 2021; 6:e130399. [PMID: 34236046 PMCID: PMC8410079 DOI: 10.1172/jci.insight.130399] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2019] [Accepted: 06/02/2021] [Indexed: 01/01/2023] Open
Abstract
Apolipoprotein B (ApoB) is the primary protein of chylomicrons, VLDLs, and LDLs and is essential for their production. Defects in ApoB synthesis and secretion result in several human diseases, including abetalipoproteinemia and familial hypobetalipoproteinemia (FHBL1). In addition, ApoB-related dyslipidemia is linked to nonalcoholic fatty liver disease (NAFLD), a silent pandemic affecting billions globally. Due to the crucial role of APOB in supplying nutrients to the developing embryo, ApoB deletion in mammals is embryonic lethal. Thus, a clear understanding of the roles of this protein during development is lacking. Here, we established zebrafish mutants for 2 apoB genes: apoBa and apoBb.1. Double-mutant embryos displayed hepatic steatosis, a common hallmark of FHBL1 and NAFLD, as well as abnormal liver laterality, decreased numbers of goblet cells in the gut, and impaired angiogenesis. We further used these mutants to identify the domains within ApoB responsible for its functions. By assessing the ability of different truncated forms of human APOB to rescue the mutant phenotypes, we demonstrate the benefits of this model for prospective therapeutic screens. Overall, these zebrafish models uncover what are likely previously undescribed functions of ApoB in organ development and morphogenesis and shed light on the mechanisms underlying hypolipidemia-related diseases.
Collapse
|
45
|
Tang LJ, Rios RS, Zhang H, Byrne CD, Targher G, Zheng MH. Telomerase: a key player in the pathogenesis of non-alcoholic fatty liver disease? Expert Rev Gastroenterol Hepatol 2021; 15:811-819. [PMID: 33709875 DOI: 10.1080/17474124.2021.1903318] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/17/2020] [Accepted: 03/10/2021] [Indexed: 10/21/2022]
Abstract
Introduction: Telomerase is a basic nuclear protein reverse transcriptase, which plays a key role in maintaining telomere stability, genome integrity, long-term cell activity, and potential continued proliferation.Area covered: This narrative review discusses key research advances involving telomerase in the development and progression of nonalcoholic fatty liver disease (NAFLD). The review evaluates 9a) whether the assessment of telomerase can be used as a noninvasive diagnostic tool; and (b) whether modification of telomerase function might be a useful potential therapeutic target for treatment of NAFLD. Furthermore, the relationship between telomerase and other chronic metabolic diseases is evaluated.Expert opinion: Several experimental and preclinical studies have suggested that telomerase plays an important role in the development of NAFLD. However, further mechanistic studies are needed to prove a causal relationship and to better elucidate whether the measurement of telomerase has utility as a diagnostic tool or whether pharmacological manipulation of telomerase has therapeutic potential in NAFLD treatment.
Collapse
Affiliation(s)
- Liang-Jie Tang
- NAFLD Research Center, Department of Hepatology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Rafael S Rios
- NAFLD Research Center, Department of Hepatology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Huai Zhang
- NAFLD Research Center, Department of Hepatology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Christopher D Byrne
- Southampton National Institute for Health Research Biomedical Research Centre, University Hospital Southampton, Southampton General Hospital, Southampton, UK
| | - Giovanni Targher
- Section of Endocrinology, Diabetes and Metabolism, Department of Medicine, University and Azienda Ospedaliera Universitaria Integrata of Verona, Verona, Italy
| | - Ming-Hua Zheng
- NAFLD Research Center, Department of Hepatology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
- Institute of Hepatology, Wenzhou Medical University, Wenzhou, China
- Key Laboratory of Diagnosis and Treatment for the Development of Chronic Liver Disease in Zhejiang Province, Wenzhou, China
| |
Collapse
|
46
|
Diagnosis and management of secondary causes of steatohepatitis. J Hepatol 2021; 74:1455-1471. [PMID: 33577920 DOI: 10.1016/j.jhep.2021.01.045] [Citation(s) in RCA: 63] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/13/2020] [Revised: 01/09/2021] [Accepted: 01/25/2021] [Indexed: 02/06/2023]
Abstract
The term non-alcoholic fatty liver disease (NAFLD) was originally coined to describe hepatic fat deposition as part of the metabolic syndrome. However, a variety of rare hereditary liver and metabolic diseases, intestinal diseases, endocrine disorders and drugs may underlie, mimic, or aggravate NAFLD. In contrast to primary NAFLD, therapeutic interventions are available for many secondary causes of NAFLD. Accordingly, secondary causes of fatty liver disease should be considered during the diagnostic workup of patients with fatty liver disease, and treatment of the underlying disease should be started to halt disease progression. Common genetic variants in several genes involved in lipid handling and metabolism modulate the risk of progression from steatosis to fibrosis, cirrhosis and hepatocellular carcinoma development in NAFLD, alcohol-related liver disease and viral hepatitis. Hence, we speculate that genotyping of common risk variants for liver disease progression may be equally useful to gauge the likelihood of developing advanced liver disease in patients with secondary fatty liver disease.
Collapse
|
47
|
Takahashi M, Okazaki H, Ohashi K, Ogura M, Ishibashi S, Okazaki S, Hirayama S, Hori M, Matsuki K, Yokoyama S, Harada-Shiba M. Current Diagnosis and Management of Abetalipoproteinemia. J Atheroscler Thromb 2021; 28:1009-1019. [PMID: 33994405 PMCID: PMC8560840 DOI: 10.5551/jat.rv17056] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Abetalipoproteinemia (ABL) is a rare autosomal recessive disorder caused by biallelic pathogenic mutations in the
MTTP
gene. Deficiency of microsomal triglyceride transfer protein (MTTP) abrogates the assembly of apolipoprotein (apo) B-containing lipoprotein in the intestine and liver, resulting in malabsorption of fat and fat-soluble vitamins and severe hypolipidemia. Patients with ABL typically manifest steatorrhea, vomiting, and failure to thrive in infancy. The deficiency of fat-soluble vitamins progressively develops into a variety of symptoms later in life, including hematological (acanthocytosis, anemia, bleeding tendency, etc.), neuromuscular (spinocerebellar ataxia, peripheral neuropathy, myopathy, etc.), and ophthalmological symptoms (e.g., retinitis pigmentosa). If left untreated, the disease can be debilitating and even lethal by the third decade of life due to the development of severe complications, such as blindness, neuromyopathy, and respiratory failure. High dose vitamin supplementation is the mainstay for treatment and may prevent, delay, or alleviate the complications and improve the prognosis, enabling some patients to live to the eighth decade of life. However, it cannot fully prevent or restore impaired function. Novel therapeutic modalities that improve quality of life and prognosis are awaited. The aim of this review is to 1) summarize the pathogenesis, clinical signs and symptoms, diagnosis, and management of ABL, and 2) propose diagnostic criteria that define eligibility to receive financial support from the Japanese government for patients with ABL as a rare and intractable disease. In addition, our diagnostic criteria and the entry criterion of low-density lipoprotein cholesterol (LDL-C) <15 mg/dL and apoB <15 mg/dL can be useful in universal or opportunistic screening for the disease. Registry research on ABL is currently ongoing to better understand the disease burden and unmet needs of this life-threatening disease with few therapeutic options.
Collapse
Affiliation(s)
- Manabu Takahashi
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Jichi Medical University
| | - Hiroaki Okazaki
- Department of Diabetes and Metabolic Diseases, Graduate School of Medicine, The University of Tokyo
| | - Ken Ohashi
- Department of General Internal Medicine, National Cancer Center Hospital
| | - Masatsune Ogura
- Department of Molecular Innovation in Lipidology, National Cerebral and Cardiovascular Center Research Institute
| | - Shun Ishibashi
- Division of Endocrinology and Metabolism, Department of Internal Medicine, School of Medicine, Jichi Medical University
| | - Sachiko Okazaki
- Division for Health Service Promotion, The University of Tokyo
| | - Satoshi Hirayama
- Department of Clinical Laboratory Medicine, Juntendo University Graduate School of Medicine
| | - Mika Hori
- Department of Endocrinology, Research Institute of Environmental Medicine, Nagoya University
| | - Kota Matsuki
- Department of Endocrinology and Metabolism, Hirosaki University Graduate School of Medicine
| | | | - Mariko Harada-Shiba
- Department of Molecular Pathogenesis, National Cerebral and Cardiovascular Center Research Institute
| | | |
Collapse
|
48
|
Bredefeld C, Peretti N, Hussain MM. New Classification and Management of Abetalipoproteinemia and Related Disorders. Gastroenterology 2021; 160:1912-1916. [PMID: 33275938 DOI: 10.1053/j.gastro.2020.11.040] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/17/2020] [Accepted: 11/19/2020] [Indexed: 02/03/2023]
Affiliation(s)
- Cindy Bredefeld
- Department of Medicine, New York University Long Island School of Medicine, NYU Langone Hospital - Long Island, Mineola, New York
| | - Noel Peretti
- Department of Pediatric Gastroenterology-Hepatology and Nutrition, Laboratory INSERM 1060 Cardiovascular Metabolism Endocrinology and Nutrition CarMEN, Lyon, France
| | - M Mahmood Hussain
- Department of Medicine, New York University Long Island School of Medicine, NYU Langone Hospital - Long Island, Mineola, New York
| | | |
Collapse
|
49
|
Montoro-Huguet MA, Belloc B, Domínguez-Cajal M. Small and Large Intestine (I): Malabsorption of Nutrients. Nutrients 2021; 13:1254. [PMID: 33920345 PMCID: PMC8070135 DOI: 10.3390/nu13041254] [Citation(s) in RCA: 51] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2021] [Revised: 04/06/2021] [Accepted: 04/07/2021] [Indexed: 02/06/2023] Open
Abstract
Numerous disorders can alter the physiological mechanisms that guarantee proper digestion and absorption of nutrients (macro- and micronutrients), leading to a wide variety of symptoms and nutritional consequences. Malabsorption can be caused by many diseases of the small intestine, as well as by diseases of the pancreas, liver, biliary tract, and stomach. This article provides an overview of pathophysiologic mechanisms that lead to symptoms or complications of maldigestion (defined as the defective intraluminal hydrolysis of nutrients) or malabsorption (defined as defective mucosal absorption), as well as its clinical consequences, including both gastrointestinal symptoms and extraintestinal manifestations and/or laboratory abnormalities. The normal uptake of nutrients, vitamins, and minerals by the gastrointestinal tract (GI) requires several steps, each of which can be compromised in disease. This article will first describe the mechanisms that lead to poor assimilation of nutrients, and secondly discuss the symptoms and nutritional consequences of each specific disorder. The clinician must be aware that many malabsorptive disorders are manifested by subtle disorders, even without gastrointestinal symptoms (for example, anemia, osteoporosis, or infertility in celiac disease), so the index of suspicion must be high to recognize the underlying diseases in time.
Collapse
Affiliation(s)
- Miguel A. Montoro-Huguet
- Departamento de Medicina, Psiquiatría y Dermatología, Facultad de Ciencias de la Salud y del Deporte, University of Zaragoza, 50009 Zaragoza, Spain
- Unidad de Gastroenterología, Hepatología y Nutrición, Hospital Universitario San Jorge de Huesca, 22004 Huesca, Spain; (B.B.); (M.D.-C.)
- Aragonese Institute of Health Sciences (IACS), 50009 Zaragoza, Spain
| | - Blanca Belloc
- Unidad de Gastroenterología, Hepatología y Nutrición, Hospital Universitario San Jorge de Huesca, 22004 Huesca, Spain; (B.B.); (M.D.-C.)
- Aragonese Institute of Health Sciences (IACS), 50009 Zaragoza, Spain
| | - Manuel Domínguez-Cajal
- Unidad de Gastroenterología, Hepatología y Nutrición, Hospital Universitario San Jorge de Huesca, 22004 Huesca, Spain; (B.B.); (M.D.-C.)
- Aragonese Institute of Health Sciences (IACS), 50009 Zaragoza, Spain
| |
Collapse
|
50
|
Rodríguez Gutiérrez PG, González García JR, Castillo De León YA, Zárate Guerrero JR, Magaña Torres MT. A novel p.Gly417Valfs*12 mutation in the MTTP gene causing abetalipoproteinemia: Presentation of the first patient in Mexico and analysis of the previously reported cases. J Clin Lab Anal 2021; 35:e23672. [PMID: 33258201 PMCID: PMC7957982 DOI: 10.1002/jcla.23672] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2020] [Revised: 11/10/2020] [Accepted: 11/12/2020] [Indexed: 12/26/2022] Open
Abstract
BACKGROUND Our aims were to describe the first Mexican patient with abetalipoproteinemia and to perform a comparative analysis of biochemical, clinical, and genetic characteristics of 100 cases reported in the literature. METHODS We performed biochemical and molecular screenings in a Mexican girl with extremely low lipid levels and in her family. Further, we integrated and evaluated the characteristics of the cases with abetalipoproteinemia described in the literature. RESULTS Our patient is a six-year-old girl who presented vomiting, chronic diarrhea, failure to thrive, malabsorption, acanthocytosis, anemia, transaminases elevation, and extremely low lipid levels. MTTP gene sequencing revealed homozygosity for a novel mutation p.Gly417Valfs*12 (G deletion c.1250). With the analysis of the reported cases, 60 clinical features (14 classical and 46 non-classical) were observed, being the most common acanthocytosis (57.5%), malabsorption (43.7%), and diarrhea (42.5%); 48.8% of the patients presented only classic clinical features, while the remaining 51.2% developed secondary effects due to a fat-soluble vitamin deficiency. An odds ratio analysis disclosed that patients diagnosed after 10 years of age have an increased risk for presenting clinical complications (OR = 18.0; 95% CI 6.0-54.1, p < 0.0001). A great diversity of mutations in MTTP has been observed (n = 76, being the most common p.G865X and p.N139_E140) and some of them with possible residual activity. CONCLUSION The first Mexican patient with abetalipoproteinemia presents a novel MTTP mutation p.Gly417Valfs*12. Three factors that could modulate the phenotype in abetalipoproteinemia were identified: age at diagnosis, treatment, and the causal mutation.
Collapse
Affiliation(s)
- Perla Graciela Rodríguez Gutiérrez
- División de GenéticaCentro de Investigación Biomédica de OccidenteInstituto Mexicano del Seguro SocialGuadalajaraMéxico
- Doctorado en Genética HumanaCentro Universitario de Ciencias de la SaludUniversidad de GuadalajaraGuadalajaraMéxico
| | - Juan Ramón González García
- División de GenéticaCentro de Investigación Biomédica de OccidenteInstituto Mexicano del Seguro SocialGuadalajaraMéxico
| | | | | | - María Teresa Magaña Torres
- División de GenéticaCentro de Investigación Biomédica de OccidenteInstituto Mexicano del Seguro SocialGuadalajaraMéxico
| |
Collapse
|