51
|
Li F, Zhao X, Li H, Liu Y, Zhang Y, Huang X, Cao J, Du F, Wu D, Yu H. Hepatic lysosomal acid lipase drives the autophagy-lysosomal response and alleviates cholesterol metabolic disorder in ApoE deficient mice. Biochim Biophys Acta Mol Cell Biol Lipids 2021; 1866:159027. [PMID: 34416392 DOI: 10.1016/j.bbalip.2021.159027] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2021] [Revised: 07/22/2021] [Accepted: 08/13/2021] [Indexed: 02/07/2023]
Abstract
Lysosomal acid lipase (LAL)-dependent lipolysis degrades cholesteryl ester (CE) and triglyceride in the lysosome. LAL deficiency in human and mice leads to hypercholesterolemia, hepatic CE deposition, and atherosclerosis. Despite its hepatocyte-specific deficiency leads to CE accumulation, the regulation of LAL in cholesterol metabolic disease remains elusive. For the in vitro study, the target gene Lipa was transfected with recombinant shRNA or lentiviral vector in Hepa1-6 cells. It was found that LAL silencing in cells affected lysosomal function by reducing LAL activity and proteolytic activity, and altered the expression of genes related to cholesterol metabolism and autophagy, leading to cholesterol accumulation; whereas LAL overexpression improved the above effects. To explore the impacts of hepatic LAL on cholesterol metabolic disease in vivo, apolipoprotein E deficient (ApoE-/-) mice were intravenously injected with lentivirus to achieve hepatic LAL overexpression and fed a Western diet for 16 weeks. The results showed that hepatic LAL overexpression significantly reduced plasma lipid levels, alleviated inflammation and oxidative status in plasma and liver, and attenuated hepatic steatosis and fibrosis in ApoE-/- mice. Mechanically, hepatic LAL promoted cholesterol transport and biliary excretion by increasing liver X receptor alpha (LXRα) and its downstream genes, and modulated the compliance of the autophagy-lysosomal pathway. Our data provide the original evidence of the validity of hepatic LAL in controlling cholesterol metabolism and liver homeostasis, suggesting that targeting hepatic LAL may provide a promising approach to rescue cholesterol metabolic disorders, such as hypercholesterolemia and liver disease.
Collapse
Affiliation(s)
- Feifei Li
- Department of Biochemistry and Molecular Biology, Hubei Provincial Key Laboratory of Developmentally Originated Disease, Wuhan University School of Basic Medical Sciences, Wuhan, Hubei 430071, China
| | - Xiaojie Zhao
- Department of Biochemistry and Molecular Biology, Hubei Provincial Key Laboratory of Developmentally Originated Disease, Wuhan University School of Basic Medical Sciences, Wuhan, Hubei 430071, China
| | - Hao Li
- Department of Pharmacy, Zhongnan Hospital of Wuhan University, Wuhan, Hubei 430071, China
| | - Yu Liu
- Department of Biochemistry and Molecular Biology, Hubei Provincial Key Laboratory of Developmentally Originated Disease, Wuhan University School of Basic Medical Sciences, Wuhan, Hubei 430071, China
| | - Yu Zhang
- Department of Biochemistry and Molecular Biology, Hubei Provincial Key Laboratory of Developmentally Originated Disease, Wuhan University School of Basic Medical Sciences, Wuhan, Hubei 430071, China
| | - Xiaopeng Huang
- Department of Biochemistry and Molecular Biology, Hubei Provincial Key Laboratory of Developmentally Originated Disease, Wuhan University School of Basic Medical Sciences, Wuhan, Hubei 430071, China
| | - Jia Cao
- Department of Biochemistry and Molecular Biology, Hubei Provincial Key Laboratory of Developmentally Originated Disease, Wuhan University School of Basic Medical Sciences, Wuhan, Hubei 430071, China
| | - Fen Du
- Department of Biochemistry and Molecular Biology, Hubei Provincial Key Laboratory of Developmentally Originated Disease, Wuhan University School of Basic Medical Sciences, Wuhan, Hubei 430071, China
| | - Dongfang Wu
- Department of Pharmacy, Zhongnan Hospital of Wuhan University, Wuhan, Hubei 430071, China.
| | - Hong Yu
- Department of Biochemistry and Molecular Biology, Hubei Provincial Key Laboratory of Developmentally Originated Disease, Wuhan University School of Basic Medical Sciences, Wuhan, Hubei 430071, China.
| |
Collapse
|
52
|
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: 57] [Impact Index Per Article: 19.0] [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
|
53
|
Wolman's disease presenting with secondary hemophagocytic lymphohistiocytosis: a case report from Saudi Arabia and literature review. BMC Pediatr 2021; 21:72. [PMID: 33568092 PMCID: PMC7874635 DOI: 10.1186/s12887-021-02541-2] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/20/2020] [Accepted: 02/04/2021] [Indexed: 12/20/2022] Open
Abstract
Background Hemophagocytic lymphohistiocytosis (HLH) is a rare and potentially fatal syndrome that is characterized by strong activation of the immune system from hyperinflammatory cytokines. Symptoms of HLH patients include fever, hepatosplenomegaly, cytopenia, and hyperferritinemia. Inherited HLH is classified as primary, whereas secondary HLH (sHLH) occurs when acquired from non-inherited reasons that include severe infection, immune deficiency syndrome, autoimmune disorder, neoplasm, and metabolic disorder. Wolman’s disease (WD) is a rare and fatal infantile metabolic disorder caused by lysosomal acid lipase deficiency, that exhibits similar clinical signs and symptoms as HLH. This paper reports the case of an infant diagnosed with WD and who presented with sHLH. Case presentation A 4-month-old infant presenting with hepatosplenomegaly, failure to thrive, and other abnormalities. WD diagnosis was confirmed by the presence of the LIPA gene homozygous deletion c.(428 + 1_967-1)_(*1_?)del. The infant also met the HLH-2004 diagnostic criteria. Conclusions Metabolic disorder such as WD should be investigated in infants fulfilling the HLH criteria to diagnose the underlying condition. More studies are needed to understand the link between WD and sHLH and to identify appropriate therapies.
Collapse
|
54
|
Abstract
The organoid model represents a major breakthrough in cell biology that has revolutionised biomedical research. Organoids are 3D physiological in vitro structures that recapitulate morphological and functional features of in vivo tissues and offer significant advantages over traditional cell culture methods. Liver organoids are of particular interest because of the pleiotropy of functions exerted by the human liver, their utility to model different liver diseases, and their potential application as cell-based therapies in regenerative medicine. Moreover, because they can be derived from patient tissues, organoid models offer new perspectives in personalised medicine and drug discovery. In this review, we discuss the current liver organoid models for the study of liver disease.
Collapse
Key Words
- 3D cultures
- A1AT, alpha-1 antitrypsin
- ALD, alcohol-related liver disease
- CCA, cholangiocarcinoma
- CFTR, cystic fibrosis transmembrane conductance regulator
- CHC, combined hepato-cholangiocarcinoma
- CLD, chronic liver disease
- CTLN1, citrullinemia type 1
- Chol-orgs, cholangiocyte organoids
- Disease modelling
- EGF, epidermal growth factor
- ER, endoplasmic reticulum
- ESCs, embryonic stem cells
- FFAs, free fatty acids
- HCC, hepatocellular carcinoma
- HUVEC, human umbilical vein endothelial cells
- Hep-orgs, hepatocyte organoids
- IL-, interleukin-
- Liver disease
- MSC, mesenchymal stem cell
- NAFLD, non-alcoholic fatty liver disease
- Organoids
- PDO, patient-derived organoid
- PDX, patient-derived xenograft
- PHH, primary human hepatocyte
- PSC, primary sclerosing cholangitis
- Personalised medicine
- Preclinical models
- iPSC, induced pluripotent stem cell
Collapse
Affiliation(s)
- Sandro Nuciforo
- Department of Biomedicine, University Hospital Basel, University of Basel, CH-4031 Basel, Switzerland
| | - Markus H Heim
- Department of Biomedicine, University Hospital Basel, University of Basel, CH-4031 Basel, Switzerland.,Clarunis, University Center for Gastrointestinal and Liver Diseases, CH-4002 Basel, Switzerland
| |
Collapse
|
55
|
Carotti S, Aquilano K, Valentini F, Ruggiero S, Alletto F, Morini S, Picardi A, Antonelli-Incalzi R, Lettieri-Barbato D, Vespasiani-Gentilucci U. An overview of deregulated lipid metabolism in nonalcoholic fatty liver disease with special focus on lysosomal acid lipase. Am J Physiol Gastrointest Liver Physiol 2020; 319:G469-G480. [PMID: 32812776 DOI: 10.1152/ajpgi.00049.2020] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Obesity and type 2 diabetes are frequently complicated by excess fat accumulation in the liver, which is known as nonalcoholic fatty liver disease (NAFLD). In this context, liver steatosis develops as a result of the deregulation of pathways controlling de novo lipogenesis and fat catabolism. Recent evidences suggest the clinical relevance of a reduction in the activity of lysosomal acid lipase (LAL), which is a key enzyme for intracellular fat disposal, in patients with NAFLD. In this review, we provided a comprehensive overview of the critical steps in hepatic fat metabolism and alterations in these pathways in NAFLD, with a special focus on lipophagy and LAL activity. During NAFLD, hepatic fat metabolism is impaired at several levels, which is significantly contributed to by impaired lipophagy, in which reduced LAL activity may play an important role. For further research and intervention in NAFLD, targeting LAL activity may provide interesting perspectives.
Collapse
Affiliation(s)
- Simone Carotti
- Unit of Microscopic and Ultrastructural Anatomy, University Campus Bio-Medico, Rome, Italy
| | - Katia Aquilano
- Department of Biology, University of Rome, Tor Vergata, Rome, Italy
| | - Francesco Valentini
- Unit of Microscopic and Ultrastructural Anatomy, University Campus Bio-Medico, Rome, Italy
| | - Sergio Ruggiero
- Unit of Microscopic and Ultrastructural Anatomy, University Campus Bio-Medico, Rome, Italy
| | - Francesca Alletto
- Unit of Internal Medicine and Hepatology, University Campus Bio-Medico, Rome, Italy
| | - Sergio Morini
- Unit of Microscopic and Ultrastructural Anatomy, University Campus Bio-Medico, Rome, Italy
| | - Antonio Picardi
- Unit of Internal Medicine and Hepatology, University Campus Bio-Medico, Rome, Italy
| | | | - Daniele Lettieri-Barbato
- Department of Biology, University of Rome, Tor Vergata, Rome, Italy.,IRCCS Fondazione Santa Lucia, Rome, Italy
| | | |
Collapse
|
56
|
Kloska A, Węsierska M, Malinowska M, Gabig-Cimińska M, Jakóbkiewicz-Banecka J. Lipophagy and Lipolysis Status in Lipid Storage and Lipid Metabolism Diseases. Int J Mol Sci 2020; 21:E6113. [PMID: 32854299 PMCID: PMC7504288 DOI: 10.3390/ijms21176113] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2020] [Revised: 08/12/2020] [Accepted: 08/21/2020] [Indexed: 12/15/2022] Open
Abstract
This review discusses how lipophagy and cytosolic lipolysis degrade cellular lipids, as well as how these pathway ys communicate, how they affect lipid metabolism and energy homeostasis in cells and how their dysfunction affects the pathogenesis of lipid storage and lipid metabolism diseases. Answers to these questions will likely uncover novel strategies for the treatment of aforementioned human diseases, but, above all, will avoid destructive effects of high concentrations of lipids-referred to as lipotoxicity-resulting in cellular dysfunction and cell death.
Collapse
Affiliation(s)
- Anna Kloska
- Department of Medical Biology and Genetics, Faculty of Biology, University of Gdańsk, Wita Stwosza 59, 80-308 Gdańsk, Poland; (A.K.); (M.W.); (M.M.)
| | - Magdalena Węsierska
- Department of Medical Biology and Genetics, Faculty of Biology, University of Gdańsk, Wita Stwosza 59, 80-308 Gdańsk, Poland; (A.K.); (M.W.); (M.M.)
| | - Marcelina Malinowska
- Department of Medical Biology and Genetics, Faculty of Biology, University of Gdańsk, Wita Stwosza 59, 80-308 Gdańsk, Poland; (A.K.); (M.W.); (M.M.)
| | - Magdalena Gabig-Cimińska
- Department of Medical Biology and Genetics, Faculty of Biology, University of Gdańsk, Wita Stwosza 59, 80-308 Gdańsk, Poland; (A.K.); (M.W.); (M.M.)
- Laboratory of Molecular Biology, Institute of Biochemistry and Biophysics, Polish Academy of Sciences, Kładki 24, 80-822 Gdańsk, Poland
| | - Joanna Jakóbkiewicz-Banecka
- Department of Medical Biology and Genetics, Faculty of Biology, University of Gdańsk, Wita Stwosza 59, 80-308 Gdańsk, Poland; (A.K.); (M.W.); (M.M.)
| |
Collapse
|
57
|
Sookoian S, Pirola CJ, Valenti L, Davidson NO. Genetic Pathways in Nonalcoholic Fatty Liver Disease: Insights From Systems Biology. Hepatology 2020; 72:330-346. [PMID: 32170962 PMCID: PMC7363530 DOI: 10.1002/hep.31229] [Citation(s) in RCA: 78] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/16/2019] [Revised: 02/12/2020] [Accepted: 03/06/2020] [Indexed: 12/16/2022]
Abstract
Nonalcoholic fatty liver disease (NAFLD) represents a burgeoning worldwide epidemic whose etiology reflects multiple interactions between environmental and genetic factors. Here, we review the major pathways and dominant genetic modifiers known to be relevant players in human NAFLD and which may determine key components of the heritability of distinctive disease traits including steatosis and fibrosis. In addition, we have employed general assumptions which are based on known genetic factors in NAFLD to build a systems biology prediction model that includes functional enrichment. This prediction model highlights additional complementary pathways that represent plausible intersecting signaling networks that we define here as an NAFLD-Reactome. We review the evidence connecting variants in each of the major known genetic modifiers (variants in patatin-like phospholipase domain containing 3, transmembrane 6 superfamily member 2, membrane-bound O-acyltransferase domain containing 7, glucokinase regulator, and hydroxysteroid 17-beta dehydrogenase 13) to NAFLD and expand the associated underlying mechanisms using functional enrichment predictions, based on both preclinical and cell-based experimental findings. These major candidate gene variants function in distinct pathways, including substrate delivery for de novo lipogenesis; mitochondrial energy use; lipid droplet assembly, lipolytic catabolism, and fatty acid compartmentalization; and very low-density lipoprotein assembly and secretion. The NAFLD-Reactome model expands these pathways and allows for hypothesis testing, as well as serving as a discovery platform for druggable targets across multiple pathways that promote NAFLD development and influence several progressive outcomes. In conclusion, we summarize the strengths and weaknesses of studies implicating selected variants in the pathophysiology of NAFLD and highlight opportunities for future clinical research and pharmacologic intervention, as well as the implications for clinical practice.
Collapse
Affiliation(s)
- Silvia Sookoian
- University of Buenos Aires, School of Medicine, Institute of Medical Research ALanari, Ciudad Autónoma de Buenos Aires, Argentina.,National Scientific and Technical Research Council (CONICET)−University of Buenos Aires, Institute of Medical Research (IDIM), Department of Clinical and Molecular Hepatology, Ciudad Autónoma de Buenos Aires, Argentina
| | - Carlos J. Pirola
- University of Buenos Aires, School of Medicine, Institute of Medical Research ALanari, Ciudad Autónoma de Buenos Aires, Argentina.,National Scientific and Technical Research Council (CONICET)−University of Buenos Aires, Institute of Medical Research (IDIM), Department of Molecular Genetics and Biology of Complex Diseases, Ciudad Autónoma de Buenos Aires, Argentina
| | - Luca Valenti
- Translational Medicine, Department of Transfusion Medicine and Hematology, Fondazione IRCCS Ca Granda OspedalePoliclinico Milano, Department of Pathophysiology and Transplantation, Universita degli Studi di Milano, Milan, Italy
| | - Nicholas O. Davidson
- Departments of Medicine and Developmental Biology, Washington University School of Medicine, St. Louis, MO 63110, USA
| |
Collapse
|
58
|
Trépo E, Valenti L. Update on NAFLD genetics: From new variants to the clinic. J Hepatol 2020; 72:1196-1209. [PMID: 32145256 DOI: 10.1016/j.jhep.2020.02.020] [Citation(s) in RCA: 226] [Impact Index Per Article: 56.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/23/2019] [Revised: 02/04/2020] [Accepted: 02/13/2020] [Indexed: 02/07/2023]
Abstract
Non-alcoholic fatty liver disease (NAFLD) is the leading cause of liver diseases in high-income countries and the burden of NAFLD is increasing at an alarming rate. The risk of developing NAFLD and related complications is highly variable among individuals and is determined by environmental and genetic factors. Genome-wide association studies have uncovered robust and reproducible associations between variations in genes such as PNPLA3, TM6SF2, MBOAT7, GCKR, HSD17B13 and the natural history of NAFLD. These findings have provided compelling new insights into the biology of NAFLD and highlighted potentially attractive pharmaceutical targets. More recently the development of polygenic risk scores, which have shown promising results for the clinical risk prediction of other complex traits (such as cardiovascular disease and breast cancer), have provided new impetus for the clinical validation of genetic variants in NAFLD risk stratification. Herein, we review current knowledge on the genetic architecture of NAFLD, including gene-environment interactions, and discuss the implications for disease pathobiology, drug discovery and risk prediction. We particularly focus on the potential clinical translation of recent genetic advances, discussing methodological hurdles that must be overcome before these discoveries can be implemented in everyday practice.
Collapse
Affiliation(s)
- Eric Trépo
- Department of Gastroenterology, Hepatopancreatology and Digestive Oncology, C.U.B. Hôpital Erasme, Université Libre de Bruxelles, Brussels, Belgium; Laboratory of Experimental Gastroenterology, Université Libre de Bruxelles, Brussels, Belgium.
| | - Luca Valenti
- Department of Pathophysiology and Transplantation, Università degli Studi di Milano, Milan, Italy; Translational Medicine - Department of Transfusion Medicine and Hematology, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy.
| |
Collapse
|
59
|
Rashu EB, Junker AE, Danielsen KV, Dahl E, Hamberg O, Borgwardt L, Christensen VB, Wewer Albrechtsen NJ, Gluud LL. Cholesteryl ester storage disease of clinical and genetic characterisation: A case report and review of literature. World J Clin Cases 2020; 8:1642-1650. [PMID: 32432142 PMCID: PMC7211528 DOI: 10.12998/wjcc.v8.i9.1642] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/15/2020] [Revised: 03/26/2020] [Accepted: 04/16/2020] [Indexed: 02/05/2023] Open
Abstract
BACKGROUND Cholesteryl ester storage disease (CESD) is a rare genetic disease. Its symptoms and severity are highly variable. CESD is a systemic disease that can lead to the accumulation of fat and inflammation in the liver, as well as gastrointestinal and cardiovascular disease. The majority of patients require liver transplantation due to decompensated cirrhosis. Enzyme replacement therapy has been approved based on a randomized trial. Our study aims to clinically and genetically evaluate two siblings with CESD who underwent liver transplantation, as well as their first-degree family members.
CASE SUMMARY The siblings were compound heterozygous for the missense variant in LIPA exon 8, c.894G>A, (p.Gln298Gln) and a single base pair deletion, c.482del (p.Asn161Ilefs*19). Analyses of single nucleotide polymorphisms showed variants with an increased risk of fatty liver disease and fibrosis for both patients. Clinically, both patients show signs of recurrence of CESD in the liver after transplantation and additional gastrointestinal and cardiovascular signs of CESD. Three family members who were LIPA heterozygous had a lysosomal acid lipase activity below the reference value. One of these carriers, a seven-year-old boy, was found to have severe dyslipidemia and was subsequently treated with statins.
CONCLUSION Our study underlines that CESD is a multi-organ disease, the progression of which may occur post-liver transplantation. Our findings underline the need for monitoring of complications and assessment of possible further treatment.
Collapse
Affiliation(s)
- Elias Badal Rashu
- Gastrounit, Copenhagen University Hospital Hvidovre, Hvidovre 2650, Denmark
| | | | | | - Emilie Dahl
- Department of Hepatology, Rigshospitalet, Copenhagen University, Copenhagen 2100, Denmark
| | - Ole Hamberg
- Department of Hepatology, Rigshospitalet, Copenhagen University, Copenhagen 2100, Denmark
| | - Line Borgwardt
- Centre of Genomic Medicine, Rigshospitalet, Copenhagen University, Copenhagen 2100, Denmark
| | - Vibeke Brix Christensen
- Department of Paediatrics and Adolescent Medicine, Rigshospitalet, Copenhagen University, Copenhagen 2100, Denmark
| | - Nicolai J Wewer Albrechtsen
- Novo Nordisk Foundation Center for Protein Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen 2200, Denmark
- Department for Clinical Biochemistry, Rigshospitalet, University of Copenhagen, Copenhagen 2200, Denmark
| | - Lise L Gluud
- Gastrounit, Copenhagen University Hospital Hvidovre, Hvidovre 2650, Denmark
| |
Collapse
|
60
|
Nascimbeni F, Dionisi Vici C, Vespasiani Gentilucci U, Angelico F, Nobili V, Petta S, Valenti L. AISF update on the diagnosis and management of adult-onset lysosomal storage diseases with hepatic involvement. Dig Liver Dis 2020; 52:359-367. [PMID: 31902560 DOI: 10.1016/j.dld.2019.12.005] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/19/2019] [Revised: 12/07/2019] [Accepted: 12/10/2019] [Indexed: 12/11/2022]
Abstract
Lysosomal storage diseases (LSDs) are a heterogeneous group of inherited disorders caused by loss-of-function mutations in genes encoding for lysosomal enzymes/proteins. The consequence is a progressive accumulation of substrates in these intracellular organelles, resulting in cellular and tissue damage. The overall incidence is about 1/8000 live births, but is likely underestimated. LSDs are chronic progressive multi-systemic disorders, generally presenting with visceromegaly, and involvement of the central nervous system, eyes, the skeleton, and the respiratory and cardiovascular systems. The age at onset and phenotypic expression are highly variable, according to the specific enzymatic defect and tissues involved, the residual activity, and the disease-causing genotype. Enzyme-replacement therapies and substrate-reduction therapies have recently become available, leading to the improvement in symptoms, disease progression and quality of life of affected individuals. Liver involvement and hepatosplenomegaly are frequent features of LSDs and a hallmark of adult-onset forms, frequently leading to medical attention. LSDs should therefore be considered in the differential diagnosis of liver disease with organomegaly. The present document will provide a short overview of adult-onset LSDs with hepatic involvement, highlighting the specificities and systemic manifestations of the ones most frequently encountered in clinical practice, which may hint at the correct diagnosis and the appropriate treatment.
Collapse
Affiliation(s)
- Fabio Nascimbeni
- Regional Referral Centre for Lysosomal Storage Diseases, Division of Internal Medicine and Metabolism, Civil Hospital, AOU of Modena, University of Modena and Reggio Emilia, Modena, Italy
| | - Carlo Dionisi Vici
- Division of Metabolism, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | | | - Francesco Angelico
- Department of Public Health and Infective Diseases, Università Sapienza, Roma, Italy
| | - Valerio Nobili
- Division of Hepatology and Gastroenterology, Bambino Gesù Children's Hospital IRCCS, Rome, Italy
| | - Salvatore Petta
- Gastroenterology and Hepatology, PROMISE, Palermo University, Italy
| | - Luca Valenti
- Department of Pathophysiology and Transplantation, Università degli Studi di Milano, and Translational Medicine, Department of Transfusion Medicine and Hematology, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy.
| |
Collapse
|
61
|
Meng Y, Heybrock S, Neculai D, Saftig P. Cholesterol Handling in Lysosomes and Beyond. Trends Cell Biol 2020; 30:452-466. [PMID: 32413315 DOI: 10.1016/j.tcb.2020.02.007] [Citation(s) in RCA: 90] [Impact Index Per Article: 22.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2020] [Revised: 02/14/2020] [Accepted: 02/21/2020] [Indexed: 01/06/2023]
Abstract
Lysosomes are of major importance for the regulation of cellular cholesterol homeostasis. Food-derived cholesterol and cholesterol esters contained within lipoproteins are delivered to lysosomes by endocytosis. From the lysosomal lumen, cholesterol is transported to the inner surface of the lysosomal membrane through the glycocalyx; this shuttling requires Niemann-Pick C (NPC) 1 and NPC2 proteins. The lysosomal membrane proteins lysosomal-associated membrane protein (LAMP)-2 and lysosomal integral membrane protein (LIMP)-2/SCARB2 also bind cholesterol. LAMP-2 may serve as a cholesterol reservoir, whereas LIMP-2, like NPC1, is able to transport cholesterol through a transglycocalyx tunnel. Contact sites and fusion events between lysosomes and other organelles mediate the distribution of cholesterol. Lysosomal cholesterol content is sensed thereby regulating mammalian target of rapamycin complex (mTORC)-dependent signaling. This review summarizes our understanding of the major steps in cholesterol handling from the moment it enters the lysosome until it leaves this compartment.
Collapse
Affiliation(s)
- Ying Meng
- Department of Cell Biology, School of Basic Medical Sciences, Zhejiang University, Hangzhou, Zhejiang, China
| | - Saskia Heybrock
- Biochemisches Institut, Christian-Albrechts-Universität Kiel, Kiel, Germany
| | - Dante Neculai
- Department of Cell Biology, School of Basic Medical Sciences, Zhejiang University, Hangzhou, Zhejiang, China
| | - Paul Saftig
- Biochemisches Institut, Christian-Albrechts-Universität Kiel, Kiel, Germany.
| |
Collapse
|
62
|
Yıldız Y, Sivri HS. Inborn errors of metabolism in the differential diagnosis of fatty liver disease. TURKISH JOURNAL OF GASTROENTEROLOGY 2020; 31:3-16. [PMID: 32009609 DOI: 10.5152/tjg.2019.19367] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Nonalcoholic fatty liver disease (NAFLD) has become the most common chronic liver disease across all age groups. Obesity, diabetes, and metabolic syndrome, are the primary causes that are closely linked with the development of NAFLD. However, in young children, rare inborn errors of metabolism are predominant secondary causes of NAFLD. Furthermore, inborn errors of metabolism causing hepatosteatosis are often misdiagnosed as NAFLD in adolescents and adults. Many inborn errors of metabolism are treatable disorders and therefore require special consideration. This review aims to summarize the basic characteristics and diagnostic clues of inborn errors of metabolism associated with fatty liver disease. A suggested clinical and laboratory diagnostic approach is also discussed.
Collapse
Affiliation(s)
- Yılmaz Yıldız
- Pediatric Metabolic Diseases Unit, Dr. Sami Ulus Training and Research Hospital for Maternity and Children's Health and Diseases, Ankara, Turkey
| | - Hatice Serap Sivri
- Division of Metabolic Diseases, Department of Pediatrics, Hacettepe University School of Medicine, Ankara, Turkey
| |
Collapse
|
63
|
Pastores GM, Hughes DA. Lysosomal Acid Lipase Deficiency: Therapeutic Options. Drug Des Devel Ther 2020; 14:591-601. [PMID: 32103901 PMCID: PMC7023879 DOI: 10.2147/dddt.s149264] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2019] [Accepted: 01/16/2020] [Indexed: 12/23/2022] Open
Abstract
Lysosomal acid lipase (LAL) deficiency is a metabolic (storage) disorder, encompassing a severe (Wolman disease) and attenuated (Cholesterol ester storage disease) subtype; both inherited as autosomal recessive traits. Cardinal clinical features include the combination of hepatic dysfunction and dyslipidemia, as a consequence of cholesteryl esters and triglyceride accumulation, predominately in the liver and vascular and reticuloendothelial system. Significant morbidity can arise, due to liver failure and/or atherosclerosis; in part related to the severity of the underlying gene defect and corresponding enzyme deficiency. Diagnosis is based on demonstration of decreased LAL enzyme activity, complemented by analysis of the cognate gene defects. Therapeutic options include dietary manipulation and the use of lipid-lowering drugs. Sebelipase alfa, a recombinant enzyme replacement therapy, has garnered regulatory approval, following demonstration of improvements in disease-relevant markers and clinical benefit in clinical trials, which included increased survival in the most severe cases.
Collapse
Affiliation(s)
- Gregory M Pastores
- Department of Medicine (Genetics)/National Centre for Inherited Metabolic Disorders, Mater Misericordiae University Hospital and University College Dublin, Dublin, Ireland
| | - Derralynn A Hughes
- Royal Free London NHS Foundation Trust, University College London, LondonNW3 2QG, UK
| |
Collapse
|
64
|
Cebola I. Liver gene regulatory networks: Contributing factors to nonalcoholic fatty liver disease. WILEY INTERDISCIPLINARY REVIEWS-SYSTEMS BIOLOGY AND MEDICINE 2020; 12:e1480. [PMID: 32020788 DOI: 10.1002/wsbm.1480] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/29/2019] [Revised: 01/02/2020] [Accepted: 01/03/2020] [Indexed: 12/17/2022]
Abstract
Metabolic diseases such as nonalcoholic fatty liver disease (NAFLD) result from complex interactions between intrinsic and extrinsic factors, including genetics and exposure to obesogenic environments. These risk factors converge in aberrant gene expression patterns in the liver, which are underlined by altered cis-regulatory networks. In homeostasis and in disease states, liver cis-regulatory networks are established by coordinated action of liver-enriched transcription factors (TFs), which define enhancer landscapes, activating broad gene programs with spatiotemporal resolution. Recent advances in DNA sequencing have dramatically expanded our ability to map active transcripts, enhancers and TF cistromes, and to define the 3D chromatin topology that contains these elements. Deployment of these technologies has allowed investigation of the molecular processes that regulate liver development and metabolic homeostasis. Moreover, genomic studies of NAFLD patients and NAFLD models have demonstrated that the liver undergoes pervasive regulatory rewiring in NAFLD, which is reflected by aberrant gene expression profiles. We have therefore achieved an unprecedented level of detail in the understanding of liver cis-regulatory networks, particularly in physiological conditions. Future studies should aim to map active regulatory elements with added levels of resolution, addressing how the chromatin landscapes of different cell lineages contribute to and are altered in NAFLD and NAFLD-associated metabolic states. Such efforts would provide additional clues into the molecular factors that trigger this disease. This article is categorized under: Biological Mechanisms > Metabolism Biological Mechanisms > Regulatory Biology Laboratory Methods and Technologies > Genetic/Genomic Methods.
Collapse
Affiliation(s)
- Inês Cebola
- Department of Metabolism, Digestion and Reproduction, Section of Genetics and Genomics, Imperial College London, London, UK
| |
Collapse
|
65
|
The Puzzling Conservation and Diversification of Lipid Droplets from Bacteria to Eukaryotes. Results Probl Cell Differ 2020; 69:281-334. [PMID: 33263877 DOI: 10.1007/978-3-030-51849-3_11] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
Membrane compartments are amongst the most fascinating markers of cell evolution from prokaryotes to eukaryotes, some being conserved and the others having emerged via a series of primary and secondary endosymbiosis events. Membrane compartments comprise the system limiting cells (one or two membranes in bacteria, a unique plasma membrane in eukaryotes) and a variety of internal vesicular, subspherical, tubular, or reticulated organelles. In eukaryotes, the internal membranes comprise on the one hand the general endomembrane system, a dynamic network including organelles like the endoplasmic reticulum, the Golgi apparatus, the nuclear envelope, etc. and also the plasma membrane, which are linked via direct lateral connectivity (e.g. between the endoplasmic reticulum and the nuclear outer envelope membrane) or indirectly via vesicular trafficking. On the other hand, semi-autonomous organelles, i.e. mitochondria and chloroplasts, are disconnected from the endomembrane system and request vertical transmission following cell division. Membranes are organized as lipid bilayers in which proteins are embedded. The budding of some of these membranes, leading to the formation of the so-called lipid droplets (LDs) loaded with hydrophobic molecules, most notably triacylglycerol, is conserved in all clades. The evolution of eukaryotes is marked by the acquisition of mitochondria and simple plastids from Gram-positive bacteria by primary endosymbiosis events and the emergence of extremely complex plastids, collectively called secondary plastids, bounded by three to four membranes, following multiple and independent secondary endosymbiosis events. There is currently no consensus view of the evolution of LDs in the Tree of Life. Some features are conserved; others show a striking level of diversification. Here, we summarize the current knowledge on the architecture, dynamics, and multitude of functions of the lipid droplets in prokaryotes and in eukaryotes deriving from primary and secondary endosymbiosis events.
Collapse
|
66
|
Sulaiman RA. Inherited metabolic disorders and dyslipidaemia. J Clin Pathol 2019; 73:384-390. [PMID: 31757783 DOI: 10.1136/jclinpath-2019-205910] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2019] [Revised: 09/23/2019] [Accepted: 10/07/2019] [Indexed: 11/04/2022]
Abstract
Monogenic dyslipidaemia is a diverse group of multisystem disorders. Patients may present to various specialities from early childhood to late in adult life, and it usually takes longer before the diagnosis is established. Increased awareness of these disorders among clinicians is imperative for early diagnosis. This best practice review provides an overview of primary dyslipidaemias, highlighting their clinical presentation, relevant biochemical and molecular tests. It also addresses the emerging role of genetics in the early diagnosis and prevention of these disorders.
Collapse
Affiliation(s)
- Raashda A Sulaiman
- Department of Medical Genetics, King Faisal Specialist Hospital and Research Center, Riyadh, Saudi Arabia
| |
Collapse
|
67
|
Aivazidis S, Jain A, Rauniyar AK, Anderson CC, Marentette JO, Orlicky DJ, Fritz KS, Harris PS, Siegel D, Maclean KN, Roede JR. SNARE proteins rescue impaired autophagic flux in Down syndrome. PLoS One 2019; 14:e0223254. [PMID: 31714914 PMCID: PMC6850524 DOI: 10.1371/journal.pone.0223254] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2019] [Accepted: 09/17/2019] [Indexed: 01/20/2023] Open
Abstract
Down syndrome (DS) is a chromosomal disorder caused by trisomy of chromosome 21 (Ts21). Unbalanced karyotypes can lead to dysfunction of the proteostasis network (PN) and disrupted proteostasis is mechanistically associated with multiple DS comorbidities. Autophagy is a critical component of the PN that has not previously been investigated in DS. Based on our previous observations of PN disruption in DS, we investigated possible dysfunction of the autophagic machinery in human DS fibroblasts and other DS cell models. Following induction of autophagy by serum starvation, DS fibroblasts displayed impaired autophagic flux indicated by autophagolysosome accumulation and elevated p62, NBR1, and LC3-II abundance, compared to age- and sex-matched, euploid (CTL) fibroblasts. While lysosomal physiology was unaffected in both groups after serum starvation, we observed decreased basal abundance of the Soluble N-ethylmaleimide-sensitive-factor Attachment protein Receptor (SNARE) family members syntaxin 17 (STX17) and Vesicle Associated Membrane Protein 8 (VAMP8) indicating that decreased autophagic flux in DS is due at least in part to a possible impairment of autophagosome-lysosome fusion. This conclusion was further supported by the observation that over-expression of either STX17 or VAMP8 in DS fibroblasts restored autophagic degradation and reversed p62 accumulation. Collectively, our results indicate that impaired autophagic clearance is a characteristic of DS cells that can be reversed by enhancement of SNARE protein expression and provides further evidence that PN disruption represents a candidate mechanism for multiple aspects of pathogenesis in DS and a possible future target for therapeutic intervention.
Collapse
Affiliation(s)
- Stefanos Aivazidis
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado, Aurora, CO, United States of America
| | - Abhilasha Jain
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado, Aurora, CO, United States of America
| | - Abhishek K. Rauniyar
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado, Aurora, CO, United States of America
| | - Colin C. Anderson
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado, Aurora, CO, United States of America
| | - John O. Marentette
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado, Aurora, CO, United States of America
| | - David J. Orlicky
- Department of Pathology, University of Colorado School of Medicine, Aurora, CO, United States of America
| | - Kristofer S. Fritz
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado, Aurora, CO, United States of America
| | - Peter S. Harris
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado, Aurora, CO, United States of America
| | - David Siegel
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado, Aurora, CO, United States of America
| | - Kenneth N. Maclean
- Department of Pediatrics, University of Colorado School of Medicine, Aurora, CO, United States of America
- The Linda Crnic Institute for Down Syndrome, University of Colorado, Aurora, CO, United States of America
| | - James R. Roede
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado, Aurora, CO, United States of America
- The Linda Crnic Institute for Down Syndrome, University of Colorado, Aurora, CO, United States of America
- * E-mail:
| |
Collapse
|
68
|
Soll D, Spira D, Hollstein T, Haberbosch L, Demuth I, Steinhagen-Thiessen E, Bobbert T, Spranger J, Kassner U. Clinical outcome of a patient with lysosomal acid lipase deficiency and first results after initiation of treatment with Sebelipase alfa: A case report. Mol Genet Metab Rep 2019; 20:100479. [PMID: 31249784 PMCID: PMC6587018 DOI: 10.1016/j.ymgmr.2019.100479] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2019] [Revised: 05/24/2019] [Accepted: 05/25/2019] [Indexed: 01/28/2023] Open
Abstract
We report on a case of very rare autosomal recessive cholesteryl ester storage disease due to lysosomal acid lipase deficiency (LALD). LALD is caused by mutations in the lysosomal acid lipase A (LIPA) gene resulting in cholesteryl ester accumulation in liver, spleen, and macrophages. It can lead to liver failure, accelerated atherosclerosis and premature death. Until recently, treatment options were limited to lipid-lowering medications to control dyslipidemia. Presently, a long-term enzyme replacement therapy with Sebelipase alfa, a recombinant human lysosomal acid lipase, is available for patients with LALD. Our patient's condition became conspicuous at the age of two due to a xanthogranuloma of the chin together with increased lipid levels, elevated liver enzymes and hepatomegaly. It took another five years until our patient was diagnosed with LALD after genetic testing. A bi-weekly therapy with intravenous Sebelipase alfa was started at the age of 26 years. It led to normalization of lipid levels, reduction of liver enzymes and beginning regression of hepatomegaly in the absence of adverse drug reactions after 46 infusions. Since LALD can take a fatal course even in patients with a long-term stable condition, it is essential to identify affected patients early and to treat them appropriately by enzyme replacement therapy. LALD should be suspected in patients with low high-density lipoprotein cholesterol (HDL-C) and high low-density lipoprotein cholesterol (LDL-C) in conjunction with elevated liver enzymes or hepatomegaly. A registry for LALD patients shall help to advance our understanding of the disease as well as improve patient care (NCT01633489).
Collapse
Affiliation(s)
- Dominik Soll
- Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health, Lipid Clinic, Interdisciplinary Metabolism Center, Germany
| | - Dominik Spira
- Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health, Lipid Clinic, Interdisciplinary Metabolism Center, Germany
| | - Tim Hollstein
- Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health, Lipid Clinic, Interdisciplinary Metabolism Center, Germany
| | - Linus Haberbosch
- Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health, Lipid Clinic, Interdisciplinary Metabolism Center, Germany
| | - Ilja Demuth
- Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health, Lipid Clinic, Interdisciplinary Metabolism Center, Germany.,Berlin-Brandenburg Center for Regenerative Medicine (BCRT), Charité University Medicine Berlin, Germany
| | - Elisabeth Steinhagen-Thiessen
- Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health, Lipid Clinic, Interdisciplinary Metabolism Center, Germany
| | - Thomas Bobbert
- Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health, Lipid Clinic, Interdisciplinary Metabolism Center, Germany
| | - Joachim Spranger
- Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health, Lipid Clinic, Interdisciplinary Metabolism Center, Germany
| | - Ursula Kassner
- Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health, Lipid Clinic, Interdisciplinary Metabolism Center, Germany
| |
Collapse
|
69
|
Lysosomal acid lipase deficiency in Brazilian children: a case series. JORNAL DE PEDIATRIA (VERSÃO EM PORTUGUÊS) 2019. [DOI: 10.1016/j.jpedp.2018.08.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
|
70
|
Benevides GN, Miura IK, Person NC, Pugliese RPS, Danesi VLB, Lima FR, Porta G. Lysosomal acid lipase deficiency in Brazilian children: a case series. J Pediatr (Rio J) 2019; 95:552-558. [PMID: 31340901 DOI: 10.1016/j.jped.2018.05.016] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/18/2017] [Revised: 04/27/2018] [Accepted: 04/27/2018] [Indexed: 11/18/2022] Open
Abstract
OBJECTIVE To describe the demographic, clinical, laboratory and molecular characteristics of patients with lysosomal acid lipase deficiency. METHODS A retrospective review of the medical records of children with the disease. RESULTS Seven children with lysosomal acid lipase deficiency (5 male; 2 female); 6 were mixed race, and 1 was black. The mean ages at the first onset of symptoms and at diagnosis were 5.0 years (4 months to 9 years) and 6.9 years (3-10 years), respectively. Symptom manifestations at onset were: 3 patients had abdominal pain, one had bone/joint pain due to rickets, and 1 had chronic diarrhea and respiratory insufficiency due to interstitial pneumonitis. One was asymptomatic, and clinical suspicion arose due to hepatomegaly. Six patients had hepatomegaly, and none had splenomegaly. Two patients were siblings. Enzymatic assay and molecular analysis confirmed the diagnoses. Genetic analysis revealed a rare pathogenic variant (p.L89P) in three patients, described only once in medical literature and never described in Brazil. None of those patients were related to each other. Lysosomal acid lipase deficiency was previously described as an autosomal recessive disease, but three patients were heterozygous and undoubtedly had the disease (low enzyme activity, suggestive lab findings and clinical symptoms). CONCLUSION This case series supports that lysosomal acid lipase deficiency can present with highly heterogeneous signs and symptoms among patients, but it should be considered in children presenting with gastrointestinal symptoms associated with dyslipidemia. We describe a rare variant in three non-related patients that may suggest a Brazilian genotype for lysosomal acid lipase deficiency.
Collapse
Affiliation(s)
- Gabriel Nuncio Benevides
- Instituto da Criança, Hospital das Clínicas, Unidade de Gastroenterologia, Hepatologia e Nutrologia Pediátrica, São Paulo, SP, Brazil.
| | - Irene Kazue Miura
- Hospital Menino Jesus, Departamento de Hepatologia Pediátrica, São Paulo, SP, Brazil
| | - Natalia Canale Person
- Instituto da Criança, Hospital das Clínicas, Unidade de Gastroenterologia, Hepatologia e Nutrologia Pediátrica, São Paulo, SP, Brazil
| | | | | | | | - Gilda Porta
- Hospital Menino Jesus, Departamento de Hepatologia Pediátrica, São Paulo, SP, Brazil
| |
Collapse
|
71
|
NAFLD in children: new genes, new diagnostic modalities and new drugs. Nat Rev Gastroenterol Hepatol 2019; 16:517-530. [PMID: 31278377 DOI: 10.1038/s41575-019-0169-z] [Citation(s) in RCA: 195] [Impact Index Per Article: 39.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 06/04/2019] [Indexed: 12/13/2022]
Abstract
Nonalcoholic fatty liver disease (NAFLD) has rapidly become the most common form of chronic liver disease in children and adolescents. Over the past 5 years, developments have revolutionized our understanding of the genetic factors, natural history, diagnostic modalities and therapeutic targets for this disease. New polymorphisms, such as those in PNPLA3, TM6SF2, MBOAT7 and GCKR, have been identified and used to predict the development and severity of NAFLD in both adults and children, and their interaction with environmental factors has been elucidated. Studies have demonstrated the true burden of paediatric NAFLD and its progression to end-stage liver disease in adulthood. In particular, nonalcoholic steatohepatitis can progress to advanced fibrosis and cirrhosis, emphasizing the importance of early diagnosis. Non-invasive imaging tests, such as transient elastography, will probably replace liver biopsy for the diagnosis of nonalcoholic steatohepatitis and the assessment of fibrosis severity in the near future. The therapeutic landscape is also expanding rapidly with the development of drugs that can modify liver steatosis, inflammation and fibrosis, indicating that pharmacotherapy for NAFLD will become available in the future. In this Review, we summarize current knowledge and new advances related to the pathogenesis and management of paediatric NAFLD.
Collapse
|
72
|
Cohen JL, Burfield J, Valdez-Gonzalez K, Samuels A, Stefanatos AK, Yudkoff M, Pedro H, Ficicioglu C. Early diagnosis of infantile-onset lysosomal acid lipase deficiency in the advent of available enzyme replacement therapy. Orphanet J Rare Dis 2019; 14:198. [PMID: 31412917 PMCID: PMC6692931 DOI: 10.1186/s13023-019-1129-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2019] [Accepted: 06/12/2019] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND Lysosomal acid lipase deficiency (LAL-D) is an autosomal recessive disorder that can present as a severe, infantile form also known as Wolman disease. We sought to determine the outcomes and clinical needs of infants diagnosed with LAL-D, treated with enzyme replacement therapy (ERT). METHODS A chart review was conducted on two infantile-onset LAL-D patients to determine clinical outcomes based on laboratory results, abdominal imaging, growth and dietary records, cardiology, endocrinology, ophthalmology, hematology, and neurocognitive evaluations. RESULTS Two patients, both diagnosed and treated before 6 months old, demonstrated clinical improvement following weekly ERT. They required dosage increases to optimize growth and symptomatology. Both received a formula low in long chain triglycerides and high in medium chain triglycerides, an intervention that allowed significant catch-up growth. Patient 1 required treatment for partial adrenal insufficiency and hypothyroidism. Both patients demonstrated reduction in liver and spleen size and varying degrees of improved liver function. Neither experienced serious adverse reactions to ERT. CONCLUSION ERT has led to longer and healthier survival of affected infants. It is imperative that dietary interventions and systemic clinical care become integral to the management. Continued evidence of survival and clinical improvement in this population, coupled with available mass spectrometry enzyme assay from dried blood spots, raises the question of this rare and possibly underdiagnosed disorder's candidacy for newborn screening.
Collapse
Affiliation(s)
- Jennifer L. Cohen
- Division of Human Genetics, Department of Pediatrics, The Children’s Hospital of Philadelphia, 3401 Civic Center Blvd, Philadelphia, PA 19104 USA
- Perelman School of Medicine, University of Pennsylvania, 3400 Civic Center Blvd, Philadelphia, PA 19104 USA
| | - Jessica Burfield
- Division of Human Genetics, Department of Pediatrics, The Children’s Hospital of Philadelphia, 3401 Civic Center Blvd, Philadelphia, PA 19104 USA
| | | | - Angela Samuels
- Hackensack University Medical Center, 30 Prospect Ave, Hackensack, NJ 07601 USA
| | - Arianna K. Stefanatos
- Division of Human Genetics, Department of Pediatrics, The Children’s Hospital of Philadelphia, 3401 Civic Center Blvd, Philadelphia, PA 19104 USA
| | - Marc Yudkoff
- Division of Human Genetics, Department of Pediatrics, The Children’s Hospital of Philadelphia, 3401 Civic Center Blvd, Philadelphia, PA 19104 USA
- Perelman School of Medicine, University of Pennsylvania, 3400 Civic Center Blvd, Philadelphia, PA 19104 USA
| | - Helio Pedro
- Hackensack University Medical Center, 30 Prospect Ave, Hackensack, NJ 07601 USA
| | - Can Ficicioglu
- Division of Human Genetics, Department of Pediatrics, The Children’s Hospital of Philadelphia, 3401 Civic Center Blvd, Philadelphia, PA 19104 USA
- Perelman School of Medicine, University of Pennsylvania, 3400 Civic Center Blvd, Philadelphia, PA 19104 USA
- Division of Metabolism (Biochemical Genetics), The Children’s Hospital of Philadelphia, 3501 Civic center Blvd #9054, Philadelphia, PA 19104 USA
| |
Collapse
|
73
|
Taliento AE, Dallio M, Federico A, Prati D, Valenti L. Novel Insights into the Genetic Landscape of Nonalcoholic Fatty Liver Disease. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2019; 16:E2755. [PMID: 31375010 PMCID: PMC6695718 DOI: 10.3390/ijerph16152755] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/02/2019] [Revised: 07/27/2019] [Accepted: 07/30/2019] [Indexed: 12/19/2022]
Abstract
Nonalcoholic fatty liver disease (NAFLD), the most common liver disorder worldwide, is epidemiologically associated with overweight, insulin resistance features and type 2 diabetes, and can progress to advanced liver fibrosis and hepatocellular carcinoma. Genetic factors play an important role in the development of NAFLD, which is a multifactorial disease. Several common naturally occurring variants modulating lipid and retinol metabolism in hepatocytes predispose to NAFLD development and progression, in particular those in PNPLA3, TM6SF2, MBOAT7, and HSD17B13. In addition, genetic variants that protect hepatic cells from oxidative stress modulate the susceptibility to progressive NAFLD. Although the molecular mechanisms linking these genetic variants with liver disease are not yet fully understood, hepatic fat has emerged as a major driver of the disease, while altered retinol metabolism and mitochondrial oxidative stress play a role in determining the development of advanced NAFLD.
Collapse
Affiliation(s)
- Alice Emma Taliento
- Translational Medicine, Department of Transfusion Medicine and Hematology, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico IRCCS, 20122 Milan, Italy
| | - Marcello Dallio
- Department of Precision Medicine, University of Campania "Luigi Vanvitelli", 80131 Naples, Italy.
| | - Alessandro Federico
- Department of Precision Medicine, University of Campania "Luigi Vanvitelli", 80131 Naples, Italy
| | - Daniele Prati
- Translational Medicine, Department of Transfusion Medicine and Hematology, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico IRCCS, 20122 Milan, Italy
| | - Luca Valenti
- Translational Medicine, Department of Transfusion Medicine and Hematology, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico IRCCS, 20122 Milan, Italy.
- Department of Pathophysiology and Transplantation, University of Milan, 20122 Milano, Italy.
| |
Collapse
|
74
|
Association between Lysosomal Dysfunction and Obesity-Related Pathology: A Key Knowledge to Prevent Metabolic Syndrome. Int J Mol Sci 2019; 20:ijms20153688. [PMID: 31357643 PMCID: PMC6696452 DOI: 10.3390/ijms20153688] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2019] [Revised: 07/25/2019] [Accepted: 07/26/2019] [Indexed: 02/06/2023] Open
Abstract
Obesity causes various health problems, such as type 2 diabetes, non-alcoholic fatty liver disease, and cardio- and cerebrovascular diseases. Metabolic organs, particularly white adipose tissue (WAT) and liver, are deeply involved in obesity. WAT contains many adipocytes with energy storage capacity and secretes adipokines depending on the obesity state, while liver plays pivotal roles in glucose and lipid metabolism. This review outlines and underscores the relationship between obesity and lysosomal functions, including lysosome biogenesis, maturation and activity of lysosomal proteases in WAT and liver. It has been revealed that obesity-induced abnormalities of lysosomal proteases contribute to inflammation and cellular senescence in adipocytes. Previous reports have demonstrated obesity-induced ectopic lipid accumulation in liver is associated with abnormality of lysosomal proteases as well as other lysosomal enzymes. These studies demonstrate that lysosomal dysfunction in WAT and liver underlies part of the obesity-related pathology, raising the possibility that strategies to modulate lysosomal function may be effective in preventing or treating the metabolic syndrome.
Collapse
|
75
|
Pavanello C, Baragetti A, Branchi A, Grigore L, Castelnuovo S, Giorgio E, Catapano AL, Calabresi L, Gomaraschi M. Treatment with fibrates is associated with higher LAL activity in dyslipidemic patients. Pharmacol Res 2019; 147:104362. [PMID: 31330253 DOI: 10.1016/j.phrs.2019.104362] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/23/2019] [Revised: 07/01/2019] [Accepted: 07/18/2019] [Indexed: 01/01/2023]
Abstract
Lysosomal acid lipase (LAL) is responsible for the hydrolysis of cholesteryl esters (CE) and triglycerides (TG) within the lysosomes; generated cholesterol and free fatty acids (FFA) are released in the cytosol where they can regulate their own synthesis and metabolism. When LAL is not active, as in case of genetic mutations, CE and TG accumulate in the lysosomal compartment, while the lack of release of cholesterol and FFA in the cytosol leads to an upregulation of their synthesis. Thus, LAL plays a central role in the intracellular homeostasis of lipids. Since there are no indications about the effect of different lipid-lowering agents on LAL activity, aim of the study was to address the relationship between LAL activity and the type of lipid-lowering therapy in a cohort of dyslipidemic patients. LAL activity was measured on dried blood spot from 120 patients with hypercholesterolemia or mixed dyslipidemia and was negatively correlated to LDL-cholesterol levels. Among enrolled patients, ninety-one were taking one or more lipid-lowering drugs, as statins, fibrates, ezetimibe and omega-3 polyunsaturated fatty acids. When patients were stratified according to the type of lipid-lowering treatment, i.e. untreated, taking statins or taking fibrates, LAL activity was significantly higher in those with fibrates, even after adjustment for sex, age, BMI, lipid parameters, liver function, metabolic syndrome, diabetes and statin use. In a subset of patients tested after 3 months of treatment with micronized fenofibrate, LAL activity raised by 21%; the increase was negatively correlated with baseline LAL activity. Thus, the use of fibrates is independently associated with higher LAL activity in dyslipidemic patients, suggesting that the positive effects of PPAR-α activation on cellular and systemic lipid homeostasis can also include an improved LAL activity.
Collapse
Affiliation(s)
- Chiara Pavanello
- Centro E. Grossi Paoletti, Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, Milan, Italy
| | - Andrea Baragetti
- Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, Milan, Italy; S.I.S.A. Center for the Study of Atherosclerosis, Bassini Hospital, Cinisello Balsamo, Italy
| | - Adriana Branchi
- Centro per lo Studio e la Prevenzione dell'Aterosclerosi, Fondazione IRCCS Cà Granda Ospedale Maggiore Policlinico, Dipartimento di Scienze Cliniche e di Comunità, Università degli Studi di Milano, Milan, Italy
| | - Liliana Grigore
- S.I.S.A. Center for the Study of Atherosclerosis, Bassini Hospital, Cinisello Balsamo, Italy
| | - Samuela Castelnuovo
- Centro Dislipidemie, Dipartimento Cardiotoracovascolare, ASST Grande Ospedale Metropolitano Niguarda, Milan, Italy
| | - Eleonora Giorgio
- Centro E. Grossi Paoletti, Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, Milan, Italy
| | - Alberico L Catapano
- Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, Milan, Italy; IRCCS Multimedica Hospital, Milan, Italy
| | - Laura Calabresi
- Centro E. Grossi Paoletti, Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, Milan, Italy
| | - Monica Gomaraschi
- Centro E. Grossi Paoletti, Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, Milan, Italy.
| |
Collapse
|
76
|
Consuelo-Sánchez A, Vázquez-Frias R, Reyes-De La Rosa A, Acosta-Rodríguez-Bueno CP, Ortal-Vite MP, Cebolla JJ. Mutations identified in a cohort of Mexican patients with lysosomal acid lipase deficiency. Ann Hepatol 2019; 18:646-650. [PMID: 31182375 DOI: 10.1016/j.aohep.2018.07.005] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/08/2018] [Revised: 07/16/2018] [Accepted: 07/30/2018] [Indexed: 02/04/2023]
Abstract
INTRODUCTION AND OBJECTIVES Lysosomal acid lipase deficiency (LAL-D) is an autosomal recessive disease caused by mutations in the LIPA gene, located on the long arm of chromosome 10 (10q23.31). Up until now, more than 59 mutations have been described and which are the cause of a very wide clinical spectrum. The goal of this study was to identify the mutations present in Mexican pediatric patients with a diagnosis of LAL-D. MATERIALS AND METHODS A cross-sectional study was carried out which included all the pediatric patients with LAL-D treated in a tertiary hospital in Mexico from January 2000 to June 2017. RESULTS Sixteen patients with LAL-D were identified with a disease phenotype marked by the accumulation of cholesteryl esters. Eight distinct variants in the LIPA gene sequence were found, four pathogenic variants and four probably pathogenic. In six individuals, the variants were found in the homozygous state and ten were compound heterozygous. The eight variants were inverted, with five found on exon 4 and the others on exons 2, 8 and 10. The variant c.386A>G;p.His129Arg was the most common, being found in six of the 16 individuals (37.5%), making it much more frequent than what had previously been reported in the literature in proportion to the rest of the variants. The mutation known as E8SJM, which has been the mostly frequently found at the international level, was not the most common among this group of Mexican patients. In conclusion, Mexican patients present a different frequency of mutations associated with LAL-D in comparison to European populations.
Collapse
Affiliation(s)
- Alejandra Consuelo-Sánchez
- Department of Gastroenterology and Nutrition, Hospital Infantil de México Federico Gómez, Mexico City, Mexico.
| | - Rodrigo Vázquez-Frias
- Department of Gastroenterology and Nutrition, Hospital Infantil de México Federico Gómez, Mexico City, Mexico
| | | | | | - María P Ortal-Vite
- Department of Gastroenterology and Nutrition, Hospital Infantil de México Federico Gómez, Mexico City, Mexico
| | - Jorge J Cebolla
- Group of Haematological and Metabolic Diseases, Translational Research Unit, Miguel Servet University Hospital, Zaragoza, Spain
| |
Collapse
|
77
|
Mayanskiy N, Brzhozovskaya E, Pushkov A, Strokova T, Vlasov N, Surkov A, Gundobina O, Savostianov K. A kinetic assay of total lipase activity for detecting lysosomal acid lipase deficiency (LAL-D) and the molecular characterization of 18 LAL-D patients from Russia. JIMD Rep 2019; 48:75-82. [PMID: 31392116 PMCID: PMC6606977 DOI: 10.1002/jmd2.12050] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/20/2019] [Revised: 05/08/2019] [Accepted: 05/14/2019] [Indexed: 11/17/2022] Open
Abstract
Laboratory diagnostics of lysosomal acid lipase deficiency (LAL-D), a rare disorder associated with LIPA alterations, are based on the evaluation of LAL activity. In dry blood spots (DBS) submitted for LAL-D diagnostics (the screening cohort) over a two-year period or obtained from a cohort of retrospective LAL-D patients, we measured: (1) LAL activity using a two-reaction assay with 4-methylumbelliferone palmitate (4-MU-Palm) and Lalistat-2, a specific LAL inactivator; (2) total lipase (TL) activity by a 1-hour kinetic 4-MU-Palm cleavage reaction (no Lalistat-2). The TL activity was expressed as the area under the kinetic curve after 1 hour (TL-AUC1h) of the reaction and presented as the median (min-max). LAL activity was reduced in 30/537 individuals from the screening cohort, among which LIPA sequencing revealed six patients and one carrier. Overall, 16 (89%) individuals among six novel and 12 retrospective LAL-D patients carried at least one c.894G>A mutation (six were homozygous). The TL-AUC1h in nonLAL-D specimens with normal LAL activity (n = 90) was unambiguously higher (9471 [4015-23 585] RFU*h/punch) compared to LAL-D patients, including six new and nine retrospective patients (1810 [357-2608] RFU*h/punch). Importantly, in 13/15 examined nonLAL-D specimens with reduced LAL activity the TL-AUC1h was above a threshold of 2652 RFU*h/punch. Applying this threshold, the TL-AUC1h index discriminated all LAL-D patients (100% sensitivity) and 103/105 nonLAL-D specimens (98% specificity). Given that there is no need for Lalistat-2 and two parallel enzymatic reactions in conjunction with high sensitivity and specificity, the kinetic assay seems to be practical for LAL-D screening. SYNOPSIS Lysosomal acid lipase deficiency responsible for Wolman disease and cholesterol ester storage disease could be reliably detected using a kinetic assay of total lipase activity with a fluorogenic substrate.
Collapse
Affiliation(s)
- Nikolay Mayanskiy
- Russian Children Clinical HospitalPirogov Russian National Research Medical UniversityMoscowRussia
| | | | - Alexander Pushkov
- Laboratory for molecular genetics and cellular biologyNational Medical Research Center for Children's HealthMoscowRussia
| | - Tatiana Strokova
- Department for pediatric gastroenterology, hepatology and nutritionFederal Research Center for Nutrition and BiotechnologyMoscowRussia
| | - Nikolay Vlasov
- Department for pediatricsFilatov's Children Hospital, No.5Saint‐PetersburgRussia
- Gastroenterology ChairState Pediatric Medical UniversitySaint‐PetersburgRussia
| | - Andrej Surkov
- Department for gastroenterologyNational Medical Research Center for Children's HealthMoscowRussia
| | - Olga Gundobina
- Department for gastroenterologyNational Medical Research Center for Children's HealthMoscowRussia
| | - Kirill Savostianov
- Laboratory for molecular genetics and cellular biologyNational Medical Research Center for Children's HealthMoscowRussia
| |
Collapse
|
78
|
Kyosen SO, Geocze S, Yamamoto MH, Martins AM. Endoscopic Findings in Lysosomal Acid Lipase Deficiency. J Pediatr Gastroenterol Nutr 2019; 68:e105. [PMID: 29762191 DOI: 10.1097/mpg.0000000000002030] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/10/2022]
Affiliation(s)
- Sandra Obikawa Kyosen
- Department of Pediatrics, Centro de Referência em Erros Inatos do Metabolismo (CREIM)
| | - Stephan Geocze
- Centro de Endoscopia Digestiva e Respiratória (CEDIR), Hospital São Paulo, Universidade Federal de São Paulo, São Paulo, Brazil
| | | | - Ana Maria Martins
- Department of Pediatrics, Centro de Referência em Erros Inatos do Metabolismo (CREIM)
| |
Collapse
|
79
|
Pajed L, Wagner C, Taschler U, Schreiber R, Kolleritsch S, Fawzy N, Pototschnig I, Schoiswohl G, Pusch LM, Wieser BI, Vesely P, Hoefler G, Eichmann TO, Zimmermann R, Lass A. Hepatocyte-specific deletion of lysosomal acid lipase leads to cholesteryl ester but not triglyceride or retinyl ester accumulation. J Biol Chem 2019; 294:9118-9133. [PMID: 31023823 PMCID: PMC6556574 DOI: 10.1074/jbc.ra118.007201] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2018] [Revised: 04/23/2019] [Indexed: 12/22/2022] Open
Abstract
Lysosomal acid lipase (LAL) hydrolyzes cholesteryl ester (CE) and retinyl ester (RE) and triglyceride (TG). Mice globally lacking LAL accumulate CE most prominently in the liver. The severity of the CE accumulation phenotype progresses with age and is accompanied by hepatomegaly and hepatic cholesterol crystal deposition. In contrast, hepatic TG accumulation is much less pronounced in these mice, and hepatic RE levels are even decreased. To dissect the functional role of LAL for neutral lipid ester mobilization in the liver, we generated mice specifically lacking LAL in hepatocytes (hep-LAL-ko). On a standard chow diet, hep-LAL-ko mice exhibited increased hepatic CE accumulation but unaltered TG and RE levels. Feeding the hep-LAL-ko mice a vitamin A excess/high-fat diet (VitA/HFD) further increased hepatic cholesterol levels, but hepatic TG and RE levels in these mice were lower than in control mice. Performing in vitro activity assays with lysosome-enriched fractions from livers of mice globally lacking LAL, we detected residual acid hydrolytic activities against TG and RE. Interestingly, this non-LAL acid TG hydrolytic activity was elevated in lysosome-enriched fractions from livers of hep-LAL-ko mice upon VitA/HFD feeding. In conclusion, the neutral lipid ester phenotype in livers from hep-LAL-ko mice indicates that LAL is limiting for CE turnover, but not for TG and RE turnovers. Furthermore, in vitro hydrolase activity assays revealed the existence of non-LAL acid hydrolytic activities for TG and RE. The corresponding acid lipase(s) catalyzing these reactions remains to be identified.
Collapse
Affiliation(s)
- Laura Pajed
- From the Institute of Molecular Biosciences, NAWI Graz, University of Graz, Heinrichstrasse 31/II
| | - Carina Wagner
- From the Institute of Molecular Biosciences, NAWI Graz, University of Graz, Heinrichstrasse 31/II
| | - Ulrike Taschler
- From the Institute of Molecular Biosciences, NAWI Graz, University of Graz, Heinrichstrasse 31/II
| | - Renate Schreiber
- From the Institute of Molecular Biosciences, NAWI Graz, University of Graz, Heinrichstrasse 31/II
| | - Stephanie Kolleritsch
- From the Institute of Molecular Biosciences, NAWI Graz, University of Graz, Heinrichstrasse 31/II
| | - Nermeen Fawzy
- From the Institute of Molecular Biosciences, NAWI Graz, University of Graz, Heinrichstrasse 31/II
| | - Isabella Pototschnig
- From the Institute of Molecular Biosciences, NAWI Graz, University of Graz, Heinrichstrasse 31/II
| | - Gabriele Schoiswohl
- From the Institute of Molecular Biosciences, NAWI Graz, University of Graz, Heinrichstrasse 31/II
| | - Lisa-Maria Pusch
- From the Institute of Molecular Biosciences, NAWI Graz, University of Graz, Heinrichstrasse 31/II
| | - Beatrix I Wieser
- the Diagnostic and Research Center for Molecular BioMedicine, Institute of Pathology, Medical University of Graz
| | - Paul Vesely
- the Diagnostic and Research Center for Molecular BioMedicine, Institute of Pathology, Medical University of Graz
| | - Gerald Hoefler
- the Diagnostic and Research Center for Molecular BioMedicine, Institute of Pathology, Medical University of Graz.,BioTechMed-Graz, 8010 Graz, Austria
| | - Thomas O Eichmann
- From the Institute of Molecular Biosciences, NAWI Graz, University of Graz, Heinrichstrasse 31/II.,the Center for Explorative Lipidomics, BioTechMed-Graz, and
| | - Robert Zimmermann
- From the Institute of Molecular Biosciences, NAWI Graz, University of Graz, Heinrichstrasse 31/II.,BioTechMed-Graz, 8010 Graz, Austria
| | - Achim Lass
- From the Institute of Molecular Biosciences, NAWI Graz, University of Graz, Heinrichstrasse 31/II, .,BioTechMed-Graz, 8010 Graz, Austria
| |
Collapse
|
80
|
Tanaka N, Kimura T, Fujimori N, Nagaya T, Komatsu M, Tanaka E. Current status, problems, and perspectives of non-alcoholic fatty liver disease research. World J Gastroenterol 2019; 25:163-177. [PMID: 30670907 PMCID: PMC6337019 DOI: 10.3748/wjg.v25.i2.163] [Citation(s) in RCA: 90] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/05/2018] [Revised: 12/24/2018] [Accepted: 12/27/2018] [Indexed: 02/06/2023] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) is a major chronic liver disease that can lead to liver cirrhosis, liver cancer, and ultimately death. NAFLD is pathologically classified as non-alcoholic fatty liver (NAFL) or non-alcoholic steatohepatitis (NASH) based on the existence of ballooned hepatocytes, although the states have been known to transform into each other. Moreover, since the detection of ballooned hepatocytes may be difficult with limited biopsied specimens, its clinical significance needs reconsideration. Repeated liver biopsy to assess histological NAFLD activity for therapeutic response is also impractical, creating the need for body fluid biomarkers and less invasive imaging modalities. Recent longitudinal observational studies have emphasized the importance of advanced fibrosis as a determinant of NAFLD outcome. Thus, identifying predictors of fibrosis progression and developing better screening methods will enable clinicians to isolate high-risk NAFLD patients requiring early intensive intervention. Despite the considerable heterogeneity of NAFLD with regard to underlying disease, patient age, and fibrosis stage, several clinical trials are underway to develop a first-in-class drug. In this review, we summarize the present status and future direction of NAFLD/NASH research towards solving unmet medical needs.
Collapse
Affiliation(s)
- Naoki Tanaka
- Department of Metabolic Regulation, Shinshu University School of Medicine, Matsumoto 390-8621, Japan
- International Research Center for Agricultural Food Industry, Shinshu University, Matsumoto 390-8621, Japan
| | - Takefumi Kimura
- Department of Internal Medicine, Shinshu University School of Medicine, Matsumoto 390-8621, Japan
| | - Naoyuki Fujimori
- Department of Internal Medicine, Shinshu University School of Medicine, Matsumoto 390-8621, Japan
| | - Tadanobu Nagaya
- Department of Internal Medicine, Shinshu University School of Medicine, Matsumoto 390-8621, Japan
| | - Michiharu Komatsu
- Department of Internal Medicine, Shinshu University School of Medicine, Matsumoto 390-8621, Japan
| | - Eiji Tanaka
- Department of Internal Medicine, Shinshu University School of Medicine, Matsumoto 390-8621, Japan
| |
Collapse
|
81
|
van der Lienden MJC, Gaspar P, Boot R, Aerts JMFG, van Eijk M. Glycoprotein Non-Metastatic Protein B: An Emerging Biomarker for Lysosomal Dysfunction in Macrophages. Int J Mol Sci 2018; 20:E66. [PMID: 30586924 PMCID: PMC6337583 DOI: 10.3390/ijms20010066] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2018] [Revised: 12/20/2018] [Accepted: 12/21/2018] [Indexed: 12/18/2022] Open
Abstract
Several diseases are caused by inherited defects in lysosomes, the so-called lysosomal storage disorders (LSDs). In some of these LSDs, tissue macrophages transform into prominent storage cells, as is the case in Gaucher disease. Here, macrophages become the characteristic Gaucher cells filled with lysosomes laden with glucosylceramide, because of their impaired enzymatic degradation. Biomarkers of Gaucher cells were actively searched, particularly after the development of costly therapies based on enzyme supplementation and substrate reduction. Proteins selectively expressed by storage macrophages and secreted into the circulation were identified, among which glycoprotein non-metastatic protein B (GPNMB). This review focusses on the emerging potential of GPNMB as a biomarker of stressed macrophages in LSDs as well as in acquired pathologies accompanied by an excessive lysosomal substrate load in macrophages.
Collapse
Affiliation(s)
| | - Paulo Gaspar
- Leiden Institute of Chemistry, Leiden University, 2333 CC Leiden, The Netherlands.
| | - Rolf Boot
- Leiden Institute of Chemistry, Leiden University, 2333 CC Leiden, The Netherlands.
| | - Johannes M F G Aerts
- Leiden Institute of Chemistry, Leiden University, 2333 CC Leiden, The Netherlands.
| | - Marco van Eijk
- Leiden Institute of Chemistry, Leiden University, 2333 CC Leiden, The Netherlands.
| |
Collapse
|
82
|
Ding HR, Wang JL, Ren HZ, Shi XL. Lipometabolism and Glycometabolism in Liver Diseases. BIOMED RESEARCH INTERNATIONAL 2018; 2018:1287127. [PMID: 31205932 PMCID: PMC6530156 DOI: 10.1155/2018/1287127] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 10/10/2018] [Accepted: 12/03/2018] [Indexed: 12/12/2022]
Abstract
The liver is the main metabolic organ in the body especially in lipometabolism and glycometabolism. Carbohydrates and fats disorders can result in insulin resistance in the liver. Metabolic imbalance can even lead to life-threatening conditions. Therefore, it is essential to maintain the normal metabolic function of the liver. When the liver is in a pathological state, liver metabolism homeostasis is damaged, and metabolic disorders will further aggravate liver disease. Consequently, it is essential to determine the relationship between liver diseases and metabolic disorders. Here we review a lot of evidence that liver diseases are closely related to lipometabolism and glycometabolism. Although the disorder of the liver metabolism is caused by different liver diseases, the break of metabolic balance is determined by changes in the state of the liver. We discuss the relationship between liver disease and metabolic changes, outline the process of how metabolic changes are regulated by liver diseases, and describe the role which metabolic changes play in the process and prognosis of liver disease.
Collapse
Affiliation(s)
- Hao-ran Ding
- Department of Hepatobiliary Surgery, Nanjing Drum Tower Hospital Clinical College of Nanjing Medical University, Nanjing, China
- Department of Hepatobiliary Surgery, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, China
| | - Jing-lin Wang
- Department of Hepatobiliary Surgery, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, China
| | - Hao-zhen Ren
- Department of Hepatobiliary Surgery, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, China
| | - Xiao-lei Shi
- Department of Hepatobiliary Surgery, Nanjing Drum Tower Hospital Clinical College of Nanjing Medical University, Nanjing, China
- Department of Hepatobiliary Surgery, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, China
| |
Collapse
|
83
|
Pericleous M, Kelly C, Ala A, De Lusignan S. The role of the chronic care model in promoting the management of the patient with rare liver disease. Expert Rev Gastroenterol Hepatol 2018; 12:829-841. [PMID: 29976101 DOI: 10.1080/17474124.2018.1497483] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/28/2022]
Abstract
INTRODUCTION The chronic care model (CCM) provides a holistic approach for managing chronic illnesses. Patients with rare liver diseases (RLD) have complex needs, impaired quality of life and often life-threatening complications. Most RLD meet the criteria for a long-term chronic condition and should be viewed through the prism of CCM. We aimed to ascertain whether the CCM has been considered for the frequently-encountered RLD. METHODS MEDLINE®/PubMed®/Cochrane/EMBASE were searched to identify publications relating to the use of the CCM for the management of six RLD. We identified 33 articles eligible for inclusion. RESULTS Six, eleven, one, thirteen, two and zero studies, discussed individual components of the CCM for autoimmune hepatitis (AIH), primary biliary cholangitis (PBC), primary sclerosing cirrhosis (PSC), Wilsons disease (WD), alpha-1 antitrypsin deficiency (A1AD) and lysosomal acid lipase deficiency (LALd) respectively. We have not identified studies using the full CCM for any of the aforementioned RLD. DISCUSSION Unlike in common chronic conditions e.g. diabetes, there has been limited consideration of the use of CCM (or its components) for the management of RLD. This may reflect a reluctance of the clinical community to view these diseases as chronic or lack of healthcare policy investment in rare diseases in general.
Collapse
Affiliation(s)
- Marinos Pericleous
- a Department of Gastroenterology and Hepatology , Royal Surrey County Hospital NHS Foundation Trust , Guildford , UK.,b Department of Clinical and experimental medicine , University of Surrey , Guildford , UK
| | - Claire Kelly
- a Department of Gastroenterology and Hepatology , Royal Surrey County Hospital NHS Foundation Trust , Guildford , UK.,b Department of Clinical and experimental medicine , University of Surrey , Guildford , UK
| | - Aftab Ala
- a Department of Gastroenterology and Hepatology , Royal Surrey County Hospital NHS Foundation Trust , Guildford , UK.,b Department of Clinical and experimental medicine , University of Surrey , Guildford , UK
| | - Simon De Lusignan
- b Department of Clinical and experimental medicine , University of Surrey , Guildford , UK
| |
Collapse
|
84
|
Access to care in rare liver diseases: New challenges and new opportunities. J Hepatol 2018; 68:577-585. [PMID: 29113911 DOI: 10.1016/j.jhep.2017.11.004] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/29/2017] [Revised: 10/12/2017] [Accepted: 11/01/2017] [Indexed: 12/13/2022]
Abstract
Patients with rare diseases are often disadvantaged, particularly those with rare liver diseases. Reasons for disadvantage include delayed or overlooked diagnosis, lack of local expertise and high-quality care, poor scientific understanding of the disease process and limited therapeutic options. In adult liver disease this can be compounded by prejudices towards patients with liver disease in general, because of a perception (incorrect for all rare liver diseases) that liver disease is lifestyle related and thus "self-inflicted". In paediatric rare liver diseases, such as biliary atresia, optimising outcomes requires a particularly timely diagnosis. Irrespective of patient age, the scientific and medical community must rise to the challenge of advancing our understanding of rare liver disease, searching for more effective and specific therapies, and providing the infrastructure to provide the best care for all patients, infants, children, young and older adults. The European Reference Network for Rare Liver Diseases is an important step in this direction.
Collapse
|
85
|
Abstract
Lysosomal acid lipase (LAL) deficiency is an autosomal recessive lysosomal storage disorder caused by mutations in the LIPA gene that leads to premature organ damage and mortality. We present retrospective data from medical records of 5 Brazilian patients, showing the broad clinical spectrum of the disease.
Collapse
|