1
|
Wang L, Yu H, Wang D, Yin G, Chen S, Zhang X, Yu W, Meng D, Liu H, Jiang W, Zhang F. Diosgenin alleviates lipid accumulation in NAFLD through the pathways of ferroptosis defensive and executive system. J Nutr Biochem 2025; 140:109886. [PMID: 40023201 DOI: 10.1016/j.jnutbio.2025.109886] [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: 08/26/2024] [Revised: 02/17/2025] [Accepted: 02/21/2025] [Indexed: 03/04/2025]
Abstract
The most prevalent liver condition globally is non-alcoholic fatty liver disease (NAFLD), for which no approved therapies currently exist. Diosgenin, an important component in plants from the Leguminosae, Dioscoreaceae, and Solanaceae families, has demonstrated considerable anti-inflammatory and antioxidant effects. Nonetheless, the specific mechanism by which it may act in managing NAFLD remains unclear. Our research aims to explore the effects and molecular mechanisms of DG on NAFLD by utilizing both in vivo and in vitro experimental approaches. To investigate the effect of DG on hepatic steatosis, we used Sprague-Dawley rats induced by a high-fat diet (HFD) and HepG2 cells exposed to free fatty acids. Oil red O staining and hematoxylin-eosin (H&E) staining were used to explore lipid accumulation and hepatic degeneration. ROS staining, SOD, MDA, and Fe2+kits were used to detect the indexes related to oxidative stress in ferroptosis in hepatic tissues and cells. IFSP1 and pcDNA3.1-ACSL4 plasmid were used to knock down Ferroptosis suppressor protein1 (FSP1) and promote the expression of acyl-CoA synthetase long-chain family member 4 (ACSL4) in HepG2 cells. DG improved lipid metabolism disorders and liver damage induced by a high-fat diet in rats with NAFLD. Furthermore, the administration of DG notably decreased oxidative stress levels and liver Fe2+ concentrations in rats. Additionally, in vitro experiments demonstrated that DG treatment markedly attenuated ferroptosis and ROS accumulation in HepG2 cells induced by FFAs. Moreover, overexpression of hepatic ACSL4 expression by pcDNA3.1-ACSL4 plasmid promoted the regulatory effects of DG on LPCAT3 and ALOX15. Our research shows that DG can alleviate NAFLD by regulating the FSP1/COQ10 pathway of the ferroptosis defense system and the ACSL4/LPCAT3/ALOX15 pathway of the ferroptosis execution system. Therefore, DG may serve as a novel inhibitor of ferroptosis for the treatment of NAFLD.
Collapse
Affiliation(s)
- Linya Wang
- Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Hongzhuan Yu
- Weifang Traditional Chinese Medicine Hospital, Shandong, China
| | - Dongxian Wang
- Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Guoliang Yin
- Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Suwen Chen
- Department of Cardiology, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Xin Zhang
- Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Wenfei Yu
- Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Decheng Meng
- Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Hongshuai Liu
- Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Wenying Jiang
- Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Fengxia Zhang
- Department of Neurology, the first Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Shandong, China.
| |
Collapse
|
2
|
de las Heras J, Almohalla C, Blasco-Alonso J, Bourbon M, Couce ML, de Castro López MJ, García Jiménez MC, Gil Ortega D, González-Diéguez L, Meavilla S, Moreno-Álvarez A, Pastor-Rosado J, Sánchez-Pintos P, Serrano-Gonzalo I, López E, Valdivielso P, Yahyaoui R, Quintero J. Practical Recommendations for the Diagnosis and Management of Lysosomal Acid Lipase Deficiency with a Focus on Wolman Disease. Nutrients 2024; 16:4309. [PMID: 39770929 PMCID: PMC11678757 DOI: 10.3390/nu16244309] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2024] [Revised: 12/03/2024] [Accepted: 12/07/2024] [Indexed: 01/11/2025] Open
Abstract
Lysosomal acid lipase deficiency (LAL-D) is an ultra-rare lysosomal storage disease with two distinct phenotypes, an infantile-onset form (formerly Wolman disease) and a later-onset form (formerly cholesteryl ester storage disease). The objective of this narrative review is to examine the most important aspects of the diagnosis and treatment of LAL-D and to provide practical expert recommendations. The infantile-onset form occurs in the first weeks of life and is characterized by malnourishment and failure to thrive due to gastrointestinal impairment (vomiting, diarrhea, malabsorption), as well as systemic inflammation, hepatosplenomegaly, and adrenal calcifications. Mortality is close to 100% before one year of life in the absence of specific treatment. The later-onset form can be diagnosed in childhood or adulthood and is characterized by chronic liver injury and/or lipid profile alterations. When LAL-D is suspected, enzyme activity should be determined to confirm the diagnosis, with analysis from a dried blood spot sample being the quickest and most reliable method. In infantile-onset LAL-D, the initiation of enzyme replacement therapy (sebelipase α) and careful nutritional management with a low-lipid diet is very urgent, as prognosis is directly linked to the early initiation of specific treatment. In recent years, our knowledge of the management of LAL-D has increased considerably, with improvements regarding the initial enzyme replacement therapy dose and careful nutritional treatment with a low-lipid diet to decrease lipid deposition and systemic inflammation, leading to better outcomes. In this narrative review we offer a quick guide for the initial management of infantile-onset LAL-D.
Collapse
Affiliation(s)
- Javier de las Heras
- Division of Pediatric Metabolism, Cruces University Hospital, CIBER-ER, Metab-ERN, University of the Basque Country (UPV/EHU), Biobizkaia Health Research Institute, 48903 Bilbao, Spain
| | - Carolina Almohalla
- Unidad de Hepatología, Hospital Universitario Río Hortega, 47012 Valladolid, Spain
| | - Javier Blasco-Alonso
- Unidad de Diagnóstico y Tratamiento de Enfermedades Metabólicas Hereditarias, UGC Pediatría, Hospital Regional Universitario de Málaga, 29010 Málaga, Spain
| | - Mafalda Bourbon
- Grupo de Investigação Cardiovascular, Departamento de Promoção da Saúde e Prevenção de Doenças não Transmissíveis, Instituto Nacional de Saúde Doutor Ricardo Jorge, 1649-016 Lisboa, Portugal
- BioISI, Biosystems & Integrative Sciences Institute, Faculdade de Ciências, Universidade de Lisboa, 1649-004 Lisboa, Portugal
| | - Maria-Luz Couce
- Metabolic Unit, Department of Forensic Sciences, Pathology, Gynecology and Obstetrics, Pediatrics, Health Research Institute of Santiago de Compostela (IDIS), Hospital Clínico Universitario de Santiago de Compostela, CIBERER, MetabERN, 15706 Santiago de Compostela, Spain
| | - María José de Castro López
- Willink Biochemical Genetics Unit, St Mary’s Hospital, Manchester University Foundation Trust, University of Manchester, Manchester M13 9WL, UK
| | - Mª Concepción García Jiménez
- NeuroMetabolic Unit, Pediatría, Hospital Universitario Miguel Servet, Aragon Health Research Institute (IIS Aragón), 50009 Zaragoza, Spain
| | - David Gil Ortega
- Unidad de Gastroenterología, Hepatología y Nutrición Pediátrica, Hospital Universitario Virgen de la Arrixaca, 30120 Murcia, Spain
| | - Luisa González-Diéguez
- Liver Unit, Division of Gastroenterology and Hepatology, Hospital Universitario Central de Asturias, 33011 Oviedo, Spain
| | - Silvia Meavilla
- Metabolic Unit, Gastroenterology, Hepatology and Nutrition Department, Sant Joan de Déu Hospital, 08950 Barcelona, Spain
| | - Ana Moreno-Álvarez
- Pediatric Gastroenterology, Hepatology and Nutrition Unit, Department of Pediatrics, A Coruña University Hospital, 15006 A Coruña, Spain
| | - José Pastor-Rosado
- Lipid Unit, Department of Pediatrics, Hospital General Universitario de Elche, Universidad Miguel Hernandez de Elche, 03202 Elche, Spain
| | - Paula Sánchez-Pintos
- Metabolic Unit, Department of Forensic Sciences, Pathology, Gynecology and Obstetrics, Pediatrics, Health Research Institute of Santiago de Compostela (IDIS), Hospital Clínico Universitario de Santiago de Compostela, CIBERER, MetabERN, 15706 Santiago de Compostela, Spain
| | - Irene Serrano-Gonzalo
- Fundación Española Para el Estudio y Terapéutica de la Enfermedad de Gaucher y Otras Lisosomales (FEETEG), 50009 Zaragoza, Spain
- GIIS-012 Group, Aragon Health Research Institute (IIS Aragón), 50009 Zaragoza, Spain
| | - Eduardo López
- Spanish LAL-D Patient Organization, 08918 Badalona, Spain
| | - Pedro Valdivielso
- Unidad de Lípidos, Hospital Universitario Virgen de la Victoria, Instituto de Investigación Biomédica de Málaga (IBIMA), Universidad de Málaga, 29010 Málaga, Spain
| | - Raquel Yahyaoui
- Clinical Laboratory, Laboratory of Inherited Metabolic Disorders, Hospital Regional Universitario de Málaga, Instituto de Investigación Biomédica de Málaga IBIMA-Plataforma BIONAND, 29590 Málaga, Spain
| | - Jesús Quintero
- Pediatric Hepatology and Liver Transplant Unit, Department of Pediatrics, ERN Rare Liver ERN TransplantChild, Vall d’Hebron Barcelona Hospital Campus, Universitat Autònoma de Barcelona, 08193 Barcelona, Spain
| |
Collapse
|
3
|
Chen S, Sun S, Feng Y, Li X, Yin G, Liang P, Yu W, Meng D, Zhang X, Liu H, Zhang F. Diosgenin attenuates nonalcoholic hepatic steatosis through the hepatic FXR-SHP-SREBP1C/PPARα/CD36 pathway. Eur J Pharmacol 2023; 952:175808. [PMID: 37263401 DOI: 10.1016/j.ejphar.2023.175808] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2023] [Revised: 05/16/2023] [Accepted: 05/24/2023] [Indexed: 06/03/2023]
Abstract
Nonalcoholic fatty liver disease (NAFLD) is the most common liver disease worldwide and has no approved treatment. The hepatic farnesoid X receptor (FXR) is one of the most promising therapeutic targets for NAFLD. Diosgenin (DG), a natural compound extracted from Chinese herbal medicine, is very effective in preventing metabolic diseases. Our research aims to determine the effects and molecular mechanisms of DG on NAFLD in vivo and in vitro. The effect of DG on hepatic steatosis was evaluated in Sprague‒Dawley (SD) rats induced by a high-fat diet (HFD) and in HepG2 cells exposed to free fatty acids (FFAs, sodium oleate:sodium palmitate = 2:1). DG treatment efficiently managed hepatic lipid deposition in vivo and in vitro. Mechanistically, DG upregulated the expression of FXR and small heterodimer partner (SHP) and downregulated the expression of genes involved in hepatic de novo lipogenesis (DNL), including sterol regulatory element-binding protein 1C (SREBP1C), acetyl-CoA carboxylase 1 (ACC1), and fatty acid synthase (FASN). Moreover, DG promoted the expression of peroxisome proliferator-activated receptor alpha (PPARα), which is related to fatty acid oxidation. In addition, DG inhibited the expression of the CD36 molecule (CD36) related to fatty acid uptake. However, hepatic FXR silencing weakened the regulatory effects of DG on these genes. Collectively, our data show that DG has a good effect on alleviating nonalcoholic hepatic steatosis via the hepatic FXR-SHP-SREBP1C/PPARα/CD36 pathway. DG promises to be a novel candidate FXR activator that can be utilized to treat NAFLD.
Collapse
Affiliation(s)
- Suwen Chen
- Shandong University of Traditional Chinese Medicine, Jinan, 250013, China
| | - Shangwen Sun
- Department of Cardiology, Qilu Hospital of Shandong University, Jinan, 250012, Shandong, China
| | - Yanan Feng
- Shandong University of Traditional Chinese Medicine, Jinan, 250013, China
| | - Xiu Li
- Department of Endocrinology, Affiliated Linshu County People's Hospital, Linyi, 276799, China
| | - Guoliang Yin
- Shandong University of Traditional Chinese Medicine, Jinan, 250013, China
| | - Pengpeng Liang
- Shandong University of Traditional Chinese Medicine, Jinan, 250013, China
| | - Wenfei Yu
- Shandong University of Traditional Chinese Medicine, Jinan, 250013, China
| | - Decheng Meng
- Shandong University of Traditional Chinese Medicine, Jinan, 250013, China
| | - Xin Zhang
- Shandong University of Traditional Chinese Medicine, Jinan, 250013, China
| | - Hongshuai Liu
- Shandong University of Traditional Chinese Medicine, Jinan, 250013, China
| | - Fengxia Zhang
- Department of Neurology, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, 250013, China.
| |
Collapse
|
4
|
Musso G, Saba F, Cassader M, Gambino R. Lipidomics in pathogenesis, progression and treatment of nonalcoholic steatohepatitis (NASH): Recent advances. Prog Lipid Res 2023; 91:101238. [PMID: 37244504 DOI: 10.1016/j.plipres.2023.101238] [Citation(s) in RCA: 28] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2023] [Revised: 05/20/2023] [Accepted: 05/21/2023] [Indexed: 05/29/2023]
Abstract
Nonalcoholic fatty liver disease (NAFLD) is a chronic liver disease affecting up to 30% of the general adult population. NAFLD encompasses a histological spectrum ranging from pure steatosis to non-alcoholic steatohepatitis (NASH). NASH can progress to cirrhosis and is becoming the most common indication for liver transplantation, as a result of increasing disease prevalence and of the absence of approved treatments. Lipidomic readouts of liver blood and urine samples from experimental models and from NASH patients disclosed an abnormal lipid composition and metabolism. Collectively, these changes impair organelle function and promote cell damage, necro-inflammation and fibrosis, a condition termed lipotoxicity. We will discuss the lipid species and metabolic pathways leading to NASH development and progression to cirrhosis, as well as and those species that can contribute to inflammation resolution and fibrosis regression. We will also focus on emerging lipid-based therapeutic opportunities, including specialized proresolving lipid molecules and macrovesicles contributing to cell-to-cell communication and NASH pathophysiology.
Collapse
Affiliation(s)
- Giovanni Musso
- Dept of Emergency Medicine, San Luigi Gonzaga University Hospital, Orbassano, Turin, Italy.
| | - Francesca Saba
- Dept. of Medical Sciences, San Giovanni Battista Hospital, University of Turin, Turin, Italy
| | - Maurizio Cassader
- Dept. of Medical Sciences, San Giovanni Battista Hospital, University of Turin, Turin, Italy
| | - Roberto Gambino
- Dept. of Medical Sciences, San Giovanni Battista Hospital, University of Turin, Turin, Italy
| |
Collapse
|
5
|
Giraldo P, López de Frutos L, Cebolla JJ. Recommendations for overcoming challenges in the diagnosis of lysosomal acid lipase deficiency. Expert Opin Orphan Drugs 2022. [DOI: 10.1080/21678707.2022.2131393] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Affiliation(s)
- Pilar Giraldo
- Hematology. Hospital Quironsalud. Zaragoza. SPAIN
- Fundación Española para el Estudio y Terapéutica de la Enfermedad de Gaucher y otras lisosomales (FEETEG). Zaragoza. SPAIN
- Grupo de Investigación en Enfermedades Metabólicas y Hematológicas Raras (GIIS-012). Instituto de Investigación Sanitaria Aragón (ISS Aragón). SPAIN
| | - Laura López de Frutos
- Fundación Española para el Estudio y Terapéutica de la Enfermedad de Gaucher y otras lisosomales (FEETEG). Zaragoza. SPAIN
- Grupo de Investigación en Enfermedades Metabólicas y Hematológicas Raras (GIIS-012). Instituto de Investigación Sanitaria Aragón (ISS Aragón). SPAIN
| | - Jorge J Cebolla
- Grupo de Investigación en Enfermedades Metabólicas y Hematológicas Raras (GIIS-012). Instituto de Investigación Sanitaria Aragón (ISS Aragón). SPAIN
- Departamento de Bioquímica, Biología Molecular y Celular. Universidad de Zaragoza. SPAIN
| |
Collapse
|
6
|
Park EJ, Yoon C, Han JS, Lee GH, Kim DW, Park EJ, Lim HJ, Kang MS, Han HY, Seol HJ, Kim KP. Effect of PM10 on pulmonary immune response and fetus development. Toxicol Lett 2020; 339:1-11. [PMID: 33301788 DOI: 10.1016/j.toxlet.2020.11.024] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2020] [Revised: 11/19/2020] [Accepted: 11/29/2020] [Indexed: 01/12/2023]
Abstract
Despite numerous reports that ambient particulate matter is a key determinant for human health, toxicity data produced based on physicochemical properties of particulate matters is very lack, suggesting lack of scientific evidence for regulation. In this study, we sampled inhalable particulate matters (PM10) in northern Seoul, Korea. PM10 showed atypical- and fiber-type particles with the average size and the surface charge of 1,598.1 ± 128.7 nm and -27.5 ± 2.8, respectively, and various toxic elements were detected in the water extract. On day 90 after the first pulmonary exposure, total cell number dose-dependently increased in the lungs of both sexes of mice. PM10 induced Th1-dominant immune response with pathological changes in both sexes of mice. Meanwhile, composition of total cells and expression of proteins which functions in cell-to-cell communication showed different trends between sexes. Following, male and female mice were mated to identify effects of PM10 to the next generation. PM10 remained in the lung of dams until day 21 after birth, and the levels of IgA and IgE increased in the blood of dams exposed to the maximum dose compared to control. In addition, the interval between births of fetuses, the number of offspring, the neonatal survival rate (day 4 after birth) and the sex ratio seemed to be affected at the maximum dose, and particularly, all offspring from one dam were stillborn. In addition, expression of HIF-1α protein increased in the lung tissue of dams exposed to PM10, and level of hypoxia-related proteins was notably enhanced in PM10-exposed bronchial epithelial cells compared to control. Taken together, we suggest that inhaled PM10 may induce Th1-shifting immune response in the lung, and that it may affect reproduction (fetus development) by causing lung hypoxia. Additionally, we propose that further study is needed to identify particle-size-dependent effects on development of the next generation.
Collapse
Affiliation(s)
- Eun-Jung Park
- East-West Medical Science Research Institute, Kyung Hee Medical Science Research Institute, Kyung Hee University, Seoul, Republic of Korea.
| | - Cheolho Yoon
- Seoul Center, Korea Basic Science Institute, Seoul, Republic of Korea
| | - Ji-Seok Han
- Department of Advanced Toxicology Research, Korea Institute of Toxicology, Daejeon, Republic of Korea
| | - Gwang-Hee Lee
- School of Civil, Environmental, and Architectural Engineering, Korea University, Seoul, Republic of Korea
| | - Dong-Wan Kim
- School of Civil, Environmental, and Architectural Engineering, Korea University, Seoul, Republic of Korea
| | - Eun-Jun Park
- East-West Medical Science Research Institute, Kyung Hee Medical Science Research Institute, Kyung Hee University, Seoul, Republic of Korea
| | - Hyun-Ji Lim
- East-West Medical Science Research Institute, Kyung Hee Medical Science Research Institute, Kyung Hee University, Seoul, Republic of Korea
| | - Min-Sung Kang
- General Toxicology & Research Group, Jeonbuk Branch Institute, Korea Institute of Toxicology, Jeollabuk-do, Republic of Korea
| | - Hyoung-Yun Han
- Department of Predictive Toxicology, Korea Institute of Toxicology, Daejeon, Republic of Korea
| | - Hyun-Joo Seol
- Department of Obstetrics & Gynecology, Kyung Hee University Hospital at Gangdong, Kyung Hee University, Seoul, Republic of Korea
| | - Kwang Pyo Kim
- Department of Applied Chemistry, Institute of Natural Science, Global Center for Pharmaceutical Ingredient Materials, Kyung Hee University, Yongin, Republic of Korea; Department of Biomedical Science and technology, Kyung Hee Medical Science Research Institute, Kyung Hee University, Seoul, Republic of Korea
| |
Collapse
|
7
|
Du J, Ji Y, Qiao L, Liu Y, Lin J. Cellular endo-lysosomal dysfunction in the pathogenesis of non-alcoholic fatty liver disease. Liver Int 2020; 40:271-280. [PMID: 31765080 DOI: 10.1111/liv.14311] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/19/2019] [Revised: 11/09/2019] [Accepted: 11/13/2019] [Indexed: 12/13/2022]
Abstract
Non-alcoholic fatty liver disease (NAFLD), an increasingly devastating human disorder, is characterized by intrahepatic fat accumulation. Although important progress has been made in understanding NAFLD, the fundamental mechanisms involved in the pathogenesis of NAFLD have not been fully explained. The endo-lysosomal trafficking network is central to lipid metabolism, protein degradation and signal transduction, which are involved in a variety of diseases. In recent years, many genes and pathways in the endo-lysosomal trafficking network and involved in lysosomal biogenesis have been associated with the development and progression of NAFLD. Mutations of these genes and impaired signalling lead to dysfunction in multiple steps of the endo-lysosomal network (endocytic trafficking, membrane fusion and lysosomal degradation), resulting in the accumulation of pathogenic proteins. In this review, we will focus on how alterations in these genes and pathways affect endo-lysosomal trafficking as well as the pathophysiology of NAFLD.
Collapse
Affiliation(s)
- Jiang Du
- College of Biomedical Engineering, Xinxiang Medical University, Xinxiang, China.,Stem Cell and Biotherapy Engineering Research Center of Henan, Xinxiang Medical University, Xinxiang, China
| | - Yu Ji
- College of Biomedical Engineering, Xinxiang Medical University, Xinxiang, China
| | - Liang Qiao
- Stem Cell and Biotherapy Engineering Research Center of Henan, Xinxiang Medical University, Xinxiang, China
| | - Yanli Liu
- Stem Cell and Biotherapy Engineering Research Center of Henan, Xinxiang Medical University, Xinxiang, China
| | - Juntang Lin
- Stem Cell and Biotherapy Engineering Research Center of Henan, Xinxiang Medical University, Xinxiang, China
| |
Collapse
|
8
|
Zharkova M, Nekrasova T, Ivashkin V, Maevskaya M, Strokova T. Fatty Liver and Systemic Atherosclerosis in a Young, Lean Patient: Rule Out Lysosomal Acid Lipase Deficiency. Case Rep Gastroenterol 2019; 13:498-507. [PMID: 31911762 DOI: 10.1159/000504646] [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] [Received: 09/05/2019] [Accepted: 11/06/2019] [Indexed: 11/19/2022] Open
Abstract
Lysosomal acid lipase deficiency (LALD) is a rare genetic disease characterized by the accumulation of cholesteryl esters and triglycerides in many organs, including the liver, spleen, lymph nodes, bone marrow, and vascular endothelium. Patients with LALD can appear asymptomatic until liver failure or premature sudden death from coronary artery disease, stroke, and aneurysm, which lead to the diagnosis. Herein, we present a diagnostic workup in a young 17-year-old female patient who manifested hepatosplenomegaly, elevated liver enzymes, severe dyslipidemia, and systemic atherosclerosis. Liver biopsy demonstrated over 90% diffuse microvesicular steatosis, lipid accumulation in Kupffer cells, and birefringent cholesteryl ester crystals. The diagnosis of LALD was proven by the decrease of lysosomal acid lipase activity in dried blood spots and by the detection of two compound heterozygous mutations in the LIPA gene: nonsense mutation G796T (Gly266Term) and splicing site mutation G894A (E8SJM). The patient started enzyme replacement therapy with sebelipase alfa. Following the 1-year treatment, the patient remained asymptomatic, her serum aminotransferase levels were normal, liver density increased due to lipid resorption, and plaque-associated stenosis of carotid artery regressed. Moreover, liver biopsy showed a decrease of cholesteryl ester crystals in Kupffer cells.
Collapse
Affiliation(s)
- Maria Zharkova
- Vasilenko Clinic of Internal Diseases Propedeutics, Gastroenterology and Hepatology, University Clinical Hospital № 2, I.M. Sechenov First Moscow State Medical University, Moscow, Russian Federation
| | - Tatiana Nekrasova
- Department of Pathology, I.M. Sechenov First Moscow State Medical University, Moscow, Russian Federation
| | - Vladimir Ivashkin
- Vasilenko Clinic of Internal Diseases Propedeutics, Gastroenterology and Hepatology, University Clinical Hospital № 2, I.M. Sechenov First Moscow State Medical University, Moscow, Russian Federation
| | - Marina Maevskaya
- Vasilenko Clinic of Internal Diseases Propedeutics, Gastroenterology and Hepatology, University Clinical Hospital № 2, I.M. Sechenov First Moscow State Medical University, Moscow, Russian Federation
| | - Tatyana Strokova
- Federal Research Center for Nutrition and Biotechnology, Moscow, Russian Federation
| |
Collapse
|
9
|
Lopez AM, Chuang JC, Turley SD. Impact of loss of SOAT2 function on disease progression in the lysosomal acid lipase-deficient mouse. Steroids 2018; 130:7-14. [PMID: 29246491 PMCID: PMC5760480 DOI: 10.1016/j.steroids.2017.11.015] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/08/2017] [Revised: 11/28/2017] [Accepted: 11/30/2017] [Indexed: 01/27/2023]
Abstract
Although only a small proportion of cholesterol in the body is esterified, in several diseases marked expansion of the esterified cholesterol (EC) pool occurs. These include Wolman disease (WD) and Cholesteryl Ester Storage Disease (CESD) which both result from mutations in LIPA, the gene that encodes lysosomal acid lipase (LAL). The respective contributions that our three cholesterol esterifying enzymes make to EC production, especially in disorders like CESD, are not well defined. The current studies represent a detailed exploration of our earlier findings in young male LAL-deficient mice also missing sterol O-acyltransferase 2 (SOAT2, also called ACAT2). Here we show that, even as they aged, male and female Lal-/-: Soat2- /- mice, compared to Lal-/-: Soat2+/+ littermates, had appreciably less hepatomegaly as well as a marked reduction in the level of sequestration of EC, in liver transaminase activities, and in hepatic mRNA expression levels for markers of inflammation. Loss of SOAT2 function also dramatically curtailed EC entrapment in the small intestine of the LAL-deficient mice. Together, these data imply that SOAT2 inhibition, if applied concurrently with enzyme replacement therapy for LAL deficiency, may blunt the re-esterification of newly released unesterified cholesterol thereby improving clinical outcomes.
Collapse
Affiliation(s)
- Adam M Lopez
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390, United States.
| | - Jen-Chieh Chuang
- Children's Research Institute, University of Texas Southwestern Medical Center, Dallas, TX 75390, United States.
| | - Stephen D Turley
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390, United States.
| |
Collapse
|
10
|
Vázquez-Frias R, García-Ortiz J, Valencia-Mayoral P, Castro-Narro G, Medina-Bravo P, Santillán-Hernández Y, Flores-Calderón J, Mehta R, Arellano-Valdés C, Carbajal-Rodríguez L, Navarrete-Martínez J, Urbán-Reyes M, Valadez-Reyes M, Zárate-Mondragón F, Consuelo-Sánchez A. Mexican consensus on lysosomal acid lipase deficiency diagnosis. REVISTA DE GASTROENTEROLOGÍA DE MÉXICO (ENGLISH EDITION) 2018. [DOI: 10.1016/j.rgmxen.2018.01.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/18/2022] Open
|
11
|
Vázquez-Frias R, García-Ortiz JE, Valencia-Mayoral PF, Castro-Narro GE, Medina-Bravo PG, Santillán-Hernández Y, Flores-Calderón J, Mehta R, Arellano-Valdés CA, Carbajal-Rodríguez L, Navarrete-Martínez JI, Urbán-Reyes ML, Valadez-Reyes MT, Zárate-Mondragón F, Consuelo-Sánchez A. Mexican consensus on lysosomal acid lipase deficiency diagnosis. REVISTA DE GASTROENTEROLOGIA DE MEXICO (ENGLISH) 2018; 83:51-61. [PMID: 29287906 DOI: 10.1016/j.rgmx.2017.08.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/20/2017] [Revised: 07/09/2017] [Accepted: 08/16/2017] [Indexed: 02/07/2023]
Abstract
INTRODUCTION Lysosomal acid lipase deficiency (LAL-D) causes progressive cholesteryl ester and triglyceride accumulation in the lysosomes of hepatocytes and monocyte-macrophage system cells, resulting in a systemic disease with various manifestations that may go unnoticed. It is indispensable to recognize the deficiency, which can present in patients at any age, so that specific treatment can be given. The aim of the present review was to offer a guide for physicians in understanding the fundamental diagnostic aspects of LAL-D, to successfully aid in its identification. METHODS The review was designed by a group of Mexican experts and is presented as an orienting algorithm for the pediatrician, internist, gastroenterologist, endocrinologist, geneticist, pathologist, radiologist, and other specialists that could come across this disease in their patients. An up-to-date review of the literature in relation to the clinical manifestations of LAL-D and its diagnosis was performed. The statements were formulated based on said review and were then voted upon. The structured quantitative method employed for reaching consensus was the nominal group technique. RESULTS A practical algorithm of the diagnostic process in LAL-D patients was proposed, based on clinical and laboratory data indicative of the disease and in accordance with the consensus established for each recommendation. CONCLUSION The algorithm provides a sequence of clinical actions from different studies for optimizing the diagnostic process of patients suspected of having LAL-D.
Collapse
Affiliation(s)
- R Vázquez-Frias
- Departamento de Gastroenterología y Nutrición, Hospital Infantil de México Federico Gómez, Secretaría de Salud, Ciudad de México, México
| | - J E García-Ortiz
- Centro de Investigación Biomédica de Occidente, CMNO-IMSS, Guadalajara, Jalisco, México
| | - P F Valencia-Mayoral
- Dirección de Planeación, Hospital Infantil de México Federico Gómez, Secretaría de Salud, Ciudad de México, México
| | - G E Castro-Narro
- Departamento de Gastroenterología, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Ciudad de México, México
| | - P G Medina-Bravo
- Departamento de Endocrinología, Hospital Infantil de México Federico Gómez, Secretaría de Salud, Ciudad de México, México
| | - Y Santillán-Hernández
- Departamento de Genética, Centro Médico Nacional 20 de Noviembre, ISSSTE, Ciudad de México, México
| | - J Flores-Calderón
- Servicio de Gastroenterología, Hospital de Pediatría «Dr. Silvestre Frenk Freund», Centro Médico Nacional Siglo XXI, IMSS, Ciudad de México, México
| | - R Mehta
- Departamento de Endocrinología, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Ciudad de México, México
| | | | - L Carbajal-Rodríguez
- Departamento de Medicina Interna, Instituto Nacional de Pediatría, Ciudad de México, México
| | - J I Navarrete-Martínez
- Departamento de Genética, Hospital Central Sur de Alta Especialidad de Petróleos Mexicanos, Ciudad de México, México
| | - M L Urbán-Reyes
- Departamento de Gastroenterología y Nutrición, Hospital Infantil de México Federico Gómez, Secretaría de Salud, Ciudad de México, México
| | - M T Valadez-Reyes
- Departamento de Imagenología, Hospital Infantil de México Federico Gómez, Secretaría de Salud, Ciudad de México, México
| | - F Zárate-Mondragón
- Departamento de Gastroenterología y Nutrición, Instituto Nacional de Pediatría, Ciudad de México, México
| | - A Consuelo-Sánchez
- Departamento de Gastroenterología y Nutrición, Hospital Infantil de México Federico Gómez, Secretaría de Salud, Ciudad de México, México.
| |
Collapse
|
12
|
Abstract
INTRODUCTION With the growing obesity epidemic, nonalcoholic fatty liver disease (NAFLD) is rapidly becoming one of the leading causes of liver disease worldwide. Although obesity is a main risk factor for the development of NAFLD, it can also develop in lean subjects and can be encountered in different clinical setting and in association with an array of genetic, metabolic, nutritional, infectious and drug-induced disorders. Areas covered: This article discusses causes of fatty liver in non-obese subjects focusing on Lysosomal acid lipase deficiency (LAL-D), a commonly overlooked disorder reviewing its prevalence, genetics, pathogenesis, clinical features, diagnosis and treatment. It will also review other causes of non-alcoholic fatty liver disease, which can be encountered in the absence of obesity and metabolic syndrome. Expert commentary: Although the prevalence of LAL-D has been estimated in the range of 1 in 40,000 and 1 in 300,000, this estimate is much more than the identified cases reported in the literature, which suggests that that the disease may be considerably under-diagnosed. There is a pressing need to educate clinicians about the disease, especially with the development of new promising therapeutic modalities.
Collapse
Affiliation(s)
- Hassan H A-Kader
- a Division of Gastroenterology, Hepatology and Nutrition, Department of Pediatrics , The University of Arizona , Tucson , AZ , USA
| |
Collapse
|
13
|
Pericleous M, Kelly C, Wang T, Livingstone C, Ala A. Wolman's disease and cholesteryl ester storage disorder: the phenotypic spectrum of lysosomal acid lipase deficiency. Lancet Gastroenterol Hepatol 2017; 2:670-679. [PMID: 28786388 DOI: 10.1016/s2468-1253(17)30052-3] [Citation(s) in RCA: 94] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/03/2016] [Revised: 02/02/2017] [Accepted: 02/07/2017] [Indexed: 02/07/2023]
Abstract
Lysosomal acid lipase deficiency is a rare, autosomal recessive condition caused by mutations in the gene encoding lysosomal acid lipase (LIPA) that result in reduced or absent activity of this essential enzyme. The severity of the resulting disease depends on the nature of the underlying mutation and magnitude of its effect on enzymatic function. Wolman's disease is a severe disorder that presents during infancy, resulting in failure to thrive, hepatomegaly, and hepatic failure, and an average life expectancy of less than 4 months. Cholesteryl ester storage disorder arises later in life and is less severe, although the two diseases share many common features, including dyslipidaemia and transaminitis. The prevalence of these diseases has been estimated at one in 40 000 to 300 000, but many cases are undiagnosed and unreported, and awareness among clinicians is low. Lysosomal acid lipase deficiency-which can be diagnosed using dry blood spot testing-is often misdiagnosed as non-alcoholic fatty liver disease (NAFLD), non-alcoholic steatohepatitis (NASH), hereditary dyslipidaemia, or cryptogenic cirrhosis. There are no formal guidelines for treatment of these patients, and treatment options are limited. In this Review we appraise the existing literature on Wolman's disease and cholesteryl ester storage disease, and discuss available treatments, including enzyme replacement therapy, oral lipid-lowering therapy, stem-cell transplantation, and liver transplantation.
Collapse
Affiliation(s)
- Marinos Pericleous
- Department of Gastroenterology and Hepatology, Royal Surrey County Hospital NHS Foundation Trust, Guildford, Surrey, UK; Department of Clinical and Experimental Medicine, University of Surrey, Guildford, Surrey, UK
| | - Claire Kelly
- Department of Gastroenterology and Hepatology, Royal Surrey County Hospital NHS Foundation Trust, Guildford, Surrey, UK; Department of Clinical and Experimental Medicine, University of Surrey, Guildford, Surrey, UK
| | - Tim Wang
- Department of Clinical Biochemistry, Royal Surrey County Hospital NHS Foundation Trust, Guildford, Surrey, UK; School of Biosciences and Medicine, University of Surrey, Guildford, Surrey, UK
| | - Callum Livingstone
- Department of Clinical Biochemistry, Royal Surrey County Hospital NHS Foundation Trust, Guildford, Surrey, UK; School of Biosciences and Medicine, University of Surrey, Guildford, Surrey, UK
| | - Aftab Ala
- Department of Gastroenterology and Hepatology, Royal Surrey County Hospital NHS Foundation Trust, Guildford, Surrey, UK; Department of Clinical and Experimental Medicine, University of Surrey, Guildford, Surrey, UK.
| |
Collapse
|
14
|
Angelico F, Corradini SG, Pastori D, Fargion S, Fracanzani AL, Angelico M, Bolondi L, Tozzi G, Pujatti PL, Labbadia G, Corazza GR, Averna M, Perticone F, Croce G, Persico M, Bucci T, Baratta F, Polimeni L, Del Ben M, Violi F. Severe reduction of blood lysosomal acid lipase activity in cryptogenic cirrhosis: A nationwide multicentre cohort study. Atherosclerosis 2017; 262:179-184. [PMID: 28396038 DOI: 10.1016/j.atherosclerosis.2017.03.038] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/30/2017] [Revised: 03/22/2017] [Accepted: 03/29/2017] [Indexed: 02/07/2023]
Abstract
BACKGROUND AND AIMS Blood lysosomal acid lipase (LAL) is reduced in non-alcoholic steatohepatitis, which is the major cause of cryptogenic cirrhosis (CC); few data on LAL activity in CC do exist. We investigated LAL activity in a cohort of patients with liver cirrhosis. METHODS This is a multicentre cohort study including 274 patients with liver cirrhosis of different aetiology from 19 centres of Internal Medicine, Gastroenterology and Hepatology distributed throughout Italy. Blood LAL activity (nmol/spot/h) was measured with dried blood spot extracts using Lalistat 2. RESULTS Overall, 133 patients had CC, and 141 patients had cirrhosis by other causes (61 viral, 53 alcoholic, 20 alcoholic + viral, 7 autoimmune). Mean age was 64.2 ± 13.4 years, and 28.5% were women. Patients with CC were older compared to other aetiology-cirrhosis, with a lower Child-Turcotte-Pugh (CTP, p=0.003) and MELD (p=0.009) score, and a higher prevalence of cardio-metabolic risk factors and previous ischemic events. In the whole cohort, median LAL activity value was 0.58 nmol/spot/h, 0.49 and 0.65 in the groups of CC and known-aetiology cirrhosis, respectively (p=0.002). The difference remained significant after adjustment for white blood cells count (p=0.001). Multivariable linear regression analysis showed that CC (vs. known aetiology, Beta = -0.144, p=0.018), platelet count (Beta = 0.398, p < 0.001) and CTP score (Beta = -0.133, p=0.022) were associated with log-LAL activity. Similar results were found using MELD as covariate. CONCLUSIONS We found a marked reduction of LAL activity in patients with cryptogenic cirrhosis compared to the other known aetiologies. A prospective study will clarify the role of LAL in chronic liver diseases.
Collapse
Affiliation(s)
- Francesco Angelico
- I Clinica Medica, Atherothrombosis Center, Department of Internal Medicine and Medical Specialties, Sapienza University of Rome, Italy; Department of Public Health and Infectious Diseases, Sapienza University of Rome, Italy.
| | | | - Daniele Pastori
- I Clinica Medica, Atherothrombosis Center, Department of Internal Medicine and Medical Specialties, Sapienza University of Rome, Italy; Department of Anatomical, Histological, Forensic Medicine and Orthopedics Sciences - Sapienza University of Rome, Italy
| | - Silvia Fargion
- Department of Pathophysiology and Transplantation, Ca' Granda Foundation IRCCS Maggiore Policlinico Hospital, University of Milan, Milan, Italy
| | - Anna Ludovica Fracanzani
- Department of Pathophysiology and Transplantation, Ca' Granda Foundation IRCCS Maggiore Policlinico Hospital, University of Milan, Milan, Italy
| | | | - Luigi Bolondi
- Department of Medical and Surgical Sciences, University of Bologna, Italy
| | - Giulia Tozzi
- Unit for Neuromuscular and Neurodegenerative Diseases, Children's Hospital and Research Institute "Bambino Gesù", Rome, Italy
| | - Pietro Luigi Pujatti
- Department of Internal Medicine, Ospedale di Arzignano, ULSS n.5 "Ovest Vicentino", Italy
| | - Giancarlo Labbadia
- I Clinica Medica, Atherothrombosis Center, Department of Internal Medicine and Medical Specialties, Sapienza University of Rome, Italy
| | - Gino Roberto Corazza
- First Department of Internal Medicine, Fondazione IRCCS Policlinico San Matteo, University of Pavia, Italy
| | - Maurizio Averna
- Department of Internal Medicine and Medical Specialties - DIBIMIS, School of Medicine, University of Palermo, Palermo, Italy
| | - Francesco Perticone
- Department of Medical and Surgical Sciences, University Magna Græcia of Catanzaro, Italy
| | - Giuseppe Croce
- Internal Medicine Unit, Giuseppe Mazzini Hospital, Teramo, Italy
| | - Marcello Persico
- Internal Medicine and Hepatology Unit, Salerno University of Medicine, Salerno, Italy
| | - Tommaso Bucci
- Internal Medicine and Hepatology Unit, Salerno University of Medicine, Salerno, Italy
| | - Francesco Baratta
- I Clinica Medica, Atherothrombosis Center, Department of Internal Medicine and Medical Specialties, Sapienza University of Rome, Italy; Department of Anatomical, Histological, Forensic Medicine and Orthopedics Sciences - Sapienza University of Rome, Italy
| | - Licia Polimeni
- I Clinica Medica, Atherothrombosis Center, Department of Internal Medicine and Medical Specialties, Sapienza University of Rome, Italy
| | - Maria Del Ben
- I Clinica Medica, Atherothrombosis Center, Department of Internal Medicine and Medical Specialties, Sapienza University of Rome, Italy
| | - Francesco Violi
- I Clinica Medica, Atherothrombosis Center, Department of Internal Medicine and Medical Specialties, Sapienza University of Rome, Italy
| |
Collapse
|
15
|
Valayannopoulos V, Mengel E, Brassier A, Grabowski G. Lysosomal acid lipase deficiency: Expanding differential diagnosis. Mol Genet Metab 2017; 120:62-66. [PMID: 27876313 DOI: 10.1016/j.ymgme.2016.11.002] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/09/2016] [Revised: 11/08/2016] [Accepted: 11/08/2016] [Indexed: 10/20/2022]
Abstract
The differential diagnoses for metabolic liver diseases may be challenging in clinical settings, which represents a critical issue for disorders such as lysosomal acid lipase deficiency (LAL-D). LAL-D is caused by deficient activity of the LAL enzyme, resulting in the accumulation of cholesteryl esters and triglycerides throughout the body, predominately in the liver, spleen, gastrointestinal tract, and blood vessel walls. LAL-D is a progressive, multi-organ disease with early mortality and significant morbidity characterized by a combination of hepatic dysfunction and dyslipidemia. Evidence suggests LAL-D may be substantially underdiagnosed or misdiagnosed, which is critical given that disease progression can be unpredictable, with liver failure and/or accelerated atherosclerosis potentially contributing to early mortality. However, given the development of a simple diagnostic test and recently approved treatment, LAL-D should be incorporated into the differential diagnosis in relevant clinical settings. LAL-D can be diagnosed using an LAL enzyme-based biochemical test, thereby allowing for active monitoring of patients to detect potential disease complications and consider treatment options including diet, lipid-lowering medication, and treatment with sebelipase alfa, a recombinant enzyme replacement therapy shown to provide clinical benefit and improve disease-relevant markers in clinical trials. To illustrate the complexity of diagnosing LAL-D, this manuscript will describe the path to diagnosing LAL-D in a series of patient cases in which LAL-D was diagnosed as well as in patients where other diseases, such as Gaucher disease and Niemann-Pick disease, were initially suspected.
Collapse
Affiliation(s)
- Vassili Valayannopoulos
- Hôpital Necker, Enfants Malades, Paris, France; Sanofi Genzyme Corporation, Cambridge, MA, USA.
| | - Eugen Mengel
- Villa Metabolica, Center for Pediatric and Adolescent Medicine, University Medical Center of the Johannes Gutenberg-University, Mainz, Germany
| | - Anais Brassier
- Hôpital Necker, Enfants Malades, Paris, France; Sanofi Genzyme Corporation, Cambridge, MA, USA
| | | |
Collapse
|
16
|
Abstract
Sebelipase alfa (Kanuma®, Kanuma™), the first commercially available recombinant human lysosomal acid lipase (LAL), is approved in various countries worldwide, including those of the EU, the USA and Japan, as a long-term enzyme replacement therapy for patients diagnosed with LAL deficiency (LAL-D), an ultra-rare, autosomal recessive, progressive metabolic liver disease. In an ongoing study in nine infants presenting with early-onset LAL-D (Wolman disease), open-label treatment with sebelipase alfa significantly improved 1-year survival compared with historical controls. A substantial mortality benefit was maintained at 2 years of age, as was a reduction in disease-related activity. In an ongoing study of 66 children and adults with late-onset LAL-D (cholesteryl ester storage disease), 20 weeks' double-blind treatment with sebelipase alfa significantly reduced multiple disease-related hepatic and lipid abnormalities compared with placebo. Sustained improvements in markers of liver damage and dyslipidaemia were seen after 76 weeks' open-label treatment in an extension of this trial and, similarly, after 2 years' open-label treatment in an extension of another study in nine adults with late-onset LAL-D. Sebelipase alfa therapy has thus far been generally well tolerated, with signs and symptoms consistent with anaphylaxis being the most serious adverse reactions experienced by patients receiving the drug in clinical trials. Due to the rarity of the disease, these studies have enrolled a limited number of patients. Nonetheless, the available data indicate that sebelipase alfa is an effective disease-specific therapy for individuals with LAL-D who have historically been managed using supportive therapies (e.g. cholesterol reduction, hematopoietic stem cell transplantation, and liver transplantation).
Collapse
Affiliation(s)
- James E Frampton
- Springer, Private Bag 65901, Mairangi Bay, 0754, Auckland, New Zealand.
| |
Collapse
|
17
|
Burton BK, Deegan PB, Enns GM, Guardamagna O, Horslen S, Hovingh GK, Lobritto SJ, Malinova V, McLin VA, Raiman J, Di Rocco M, Santra S, Sharma R, Sykut-Cegielska J, Whitley CB, Eckert S, Valayannopoulos V, Quinn AG. Clinical Features of Lysosomal Acid Lipase Deficiency. J Pediatr Gastroenterol Nutr 2015; 61:619-25. [PMID: 26252914 PMCID: PMC4645959 DOI: 10.1097/mpg.0000000000000935] [Citation(s) in RCA: 79] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/19/2015] [Accepted: 07/31/2015] [Indexed: 12/13/2022]
Abstract
OBJECTIVE The aim of this study was to characterize key clinical manifestations of lysosomal acid lipase deficiency (LAL D) in children and adults. METHODS Investigators reviewed medical records of LAL D patients ages ≥5 years, extracted historical data, and obtained prospective laboratory and imaging data on living patients to develop a longitudinal dataset. RESULTS A total of 49 patients were enrolled; 48 had confirmed LAL D. Mean age at first disease-related abnormality was 9.0 years (range 0-42); mean age at diagnosis was 15.2 years (range 1-46). Twenty-nine (60%) were male patients, and 27 (56%) were <20 years of age at the time of consent/assent. Serum transaminases were elevated in most patients with 458 of 499 (92%) of alanine aminotransferase values and 265 of 448 (59%) of aspartate aminotransferase values above the upper limit of normal. Most patients had elevated low-density lipoprotein (64% patients) and total cholesterol (63%) at baseline despite most being on lipid-lowering therapies, and 44% had high-density lipoprotein levels below the lower limit of normal. More than half of the patients with liver biopsies (n = 31, mean age 13 years) had documented evidence of steatosis (87%) and/or fibrosis (52%). Imaging assessments revealed that the median liver volume was ∼1.15 multiples of normal (MN) and median spleen volume was ∼2.2 MN. Six (13%) patients had undergone a liver transplant (ages 9-43.5 years). CONCLUSION This study provides the largest longitudinal case review of patients with LAL D and confirms that LAL D is predominantly a pediatric disease causing early and progressive hepatic dysfunction associated with dyslipidemia that often leads to liver failure and transplantation.
Collapse
Affiliation(s)
- Barbara K. Burton
- Division of Genetics, Birth Defects and Metabolism, Ann & Robert H. Lurie Children's Hospital of Chicago, Chicago, IL
| | - Patrick B. Deegan
- Department of Medicine, Addenbrooke's Hospital NHS Trust, Cambridge, UK
| | - Gregory M. Enns
- Medical Genetics Division, Stanford University, Stanford, CA
| | | | | | - Gerard K. Hovingh
- Department of Vascular Medicine-Internal Medicine, Academic Medical Center, Amsterdam, The Netherlands
| | - Steve J. Lobritto
- New York-Presbyterian/Columbia University Medical Center, New York, NY
| | - Vera Malinova
- Department of Pediatrics, First Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Valerie A. McLin
- Departement de l’Enfant et de l’Adolescent, Hopitaux Universitaires de Geneve, Geneva, Switzerland
| | - Julian Raiman
- Division of Clinical and Metabolic Genetics, Hospital for Sick Children, Toronto, Ontario, Canada
| | - Maja Di Rocco
- Department of Pediatrics, Unit of Rare Diseases, Gaslini Institute Genoa, Genova, Italy
| | - Saikat Santra
- Department of Inherited Metabolic Disorders, Birmingham Children's Hospital, Birmingham, UK
| | - Reena Sharma
- Department of Adult Inherited Metabolic Diseases, Salford Royal NHS Foundation, Salford, UK
| | | | | | | | | | | |
Collapse
|
18
|
Lonardo A, Bellentani S, Argo CK, Ballestri S, Byrne CD, Caldwell SH, Cortez-Pinto H, Grieco A, Machado MV, Miele L, Targher G. Epidemiological modifiers of non-alcoholic fatty liver disease: Focus on high-risk groups. Dig Liver Dis 2015; 47:997-1006. [PMID: 26454786 DOI: 10.1016/j.dld.2015.08.004] [Citation(s) in RCA: 345] [Impact Index Per Article: 34.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/15/2015] [Revised: 07/27/2015] [Accepted: 08/06/2015] [Indexed: 02/07/2023]
Abstract
An improved understanding of non-alcoholic fatty liver disease epidemiology would lead to identification of individuals at high risk of developing chronic liver disease and extra-hepatic complications, thus contributing to more effective case finding of non-alcoholic fatty liver disease among selected groups. We aimed to illustrate the epidemiology of non-alcoholic fatty liver disease in high-risk groups, which were identified based on existing literature. To this end, PubMed was searched to retrieve original articles published until May 2015 using relevant and pertinent keywords "nonalcoholic fatty liver disease" and "diabetes", "obesity", "hyperlipidaemia", "familial heterozygous hypobetalipoproteinaemia", "hypertension", "metabolic syndrome", "ethnicity", "family history" or "genetic polymorphisms". We found that age, sex and ethnicity are major physiological modifiers of the risk of non-alcoholic fatty liver disease, along with belonging to "non-alcoholic fatty liver disease families" and carrying risk alleles for selected genetic polymorphisms. Metabolic syndrome, diabetes, obesity, mixed hyperlipidaemia and hypocholesterolaemia due to familial hypobetalipoproteinaemia are the major metabolic modifiers of non-alcoholic fatty liver disease risk. Compared with these metabolic conditions, however, arterial hypertension appears to carry a relatively more modest risk of non-alcoholic fatty liver disease. A better understanding of the epidemiology of non-alcoholic fatty liver disease may result in a more liberal policy of case finding among high-risk groups.
Collapse
Affiliation(s)
- Amedeo Lonardo
- Internal Medicine and Outpatient Liver Clinic, NOCSAE Baggiovara, Azienda USL di Modena, Modena, Italy.
| | - Stefano Bellentani
- Internal Medicine and Outpatient Liver Clinic, NOCSAE Baggiovara, Azienda USL di Modena, Modena, Italy; Department of Gastroenterology and Endoscopy, NOCSE Baggiovara, Azienda USL di Modena Modena, Italy
| | | | - Stefano Ballestri
- Internal Medicine Pavullo Hospital, Azienda USL di Modena, Modena, Italy
| | - Christopher D Byrne
- Nutrition and Metabolism, University of Southampton, Southampton National Institute for Health Research Biomedical Research Centre, Southampton, UK
| | | | - Helena Cortez-Pinto
- Department of Gastroenterology, University Hospital of Santa Maria, Faculty of Medicine, Lisbon, Portugal
| | - Antonio Grieco
- Institute of Internal Medicine, Catholic University of Rome, Rome, Italy
| | - Mariana V Machado
- Department of Gastroenterology, University Hospital of Santa Maria, Faculty of Medicine, Lisbon, Portugal
| | - Luca Miele
- Institute of Internal Medicine, Catholic University of Rome, Rome, Italy
| | - Giovanni Targher
- Section of Endocrinology, Diabetes and Metabolism, Department of Medicine, University of Verona and Azienda Ospedaliera Universitaria Integrata di Verona, Verona, Italy
| |
Collapse
|
19
|
Baratta F, Pastori D, Polimeni L, Tozzi G, Violi F, Angelico F, Del Ben M. Does Lysosomial Acid Lipase Reduction Play a Role in Adult Non-Alcoholic Fatty Liver Disease? Int J Mol Sci 2015; 16:28014-28021. [PMID: 26602919 PMCID: PMC4691032 DOI: 10.3390/ijms161226085] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2015] [Revised: 11/06/2015] [Accepted: 11/17/2015] [Indexed: 12/15/2022] Open
Abstract
Lysosomal Acid Lipase (LAL) is a key enzyme involved in lipid metabolism, responsible for hydrolysing the cholesteryl esters and triglycerides. Wolman Disease represents the early onset phenotype of LAL deficiency rapidly leading to death. Cholesterol Ester Storage Disease is a late onset phenotype that occurs with fatty liver, elevated aminotransferase levels, hepatomegaly and dyslipidaemia, the latter characterized by elevated LDL-C and low HDL-C. The natural history and the clinical manifestations of the LAL deficiency in adults are not well defined, and the diagnosis is often incidental. LAL deficiency has been suggested as an under-recognized cause of dyslipidaemia and fatty liver. Therefore, LAL activity may be reduced also in non-obese patients presenting non-alcoholic fatty liver disease (NAFLD), unexplained persistently elevated liver transaminases or with elevation in LDL cholesterol. In these patients, it could be indicated to test LAL activity. So far, very few studies have been performed to assess LAL activity in representative samples of normal subjects or patients with NAFLD. Moreover, no large study has been carried out in adult subjects with NAFLD or cryptogenic cirrhosis.
Collapse
Affiliation(s)
- Francesco Baratta
- Department of Internal Medicine and Medical Specialities and Department of Anatomical, Histological, Forensic Medicine and Orthopedics Sciences-Sapienza University, Rome 00185, Italy.
| | - Daniele Pastori
- Department of Internal Medicine and Medical Specialities and Department of Anatomical, Histological, Forensic Medicine and Orthopedics Sciences-Sapienza University, Rome 00185, Italy.
| | - Licia Polimeni
- Department of Internal Medicine and Medical Specialities and Department of Anatomical, Histological, Forensic Medicine and Orthopedics Sciences-Sapienza University, Rome 00185, Italy.
| | - Giulia Tozzi
- Unit for Neuromuscular and Neurodegenerative Diseases, Children's Hospital and Research Institute "Bambino Gesù", Rome 00165, Italy.
| | - Francesco Violi
- Department of Internal Medicine and Medical Specialities, Sapienza University, Rome 00185, Italy.
| | - Francesco Angelico
- Department of Public Health and Infectious Diseases, Sapienza University, Policlinico Umberto I, I Clinica Medica, Viale del Policlinico 155, Rome 00161, Italy.
| | - Maria Del Ben
- Department of Internal Medicine and Medical Specialities, Sapienza University, Rome 00185, Italy.
| |
Collapse
|
20
|
Abstract
Sebelipase alfa (Kanuma™) is a recombinant human lysosomal acid lipase (LAL) developed by Synageva BioPharma Corp. (now Alexion Pharmaceuticals, Inc.) for long-term enzyme replacement therapy in patients with LAL deficiency. The agent, administered by intravenous infusion once weekly or once every other week, acts to replace the deficient enzyme activity in patients with LAL deficiency, reducing lysosomal lipid accumulation, and thereby improving disease-related abnormalities such as dyslipidaemia and liver abnormalities. Sebelipase alfa received its first global approval, in the EU, in August 2015 for long-term enzyme replacement therapy in patients of all ages with LAL deficiency. Regulatory submissions have also been filed in the USA, Mexico and Japan for use in this indication. This article summarizes the milestones in the development of sebelipase alfa leading to this first approval for the treatment of LAL deficiency.
Collapse
Affiliation(s)
- Matt Shirley
- Springer, Private Bag 65901, Mairangi Bay, 0754, Auckland, New Zealand.
| |
Collapse
|
21
|
Burton BK, Balwani M, Feillet F, Barić I, Burrow TA, Camarena Grande C, Coker M, Consuelo-Sánchez A, Deegan P, Di Rocco M, Enns GM, Erbe R, Ezgu F, Ficicioglu C, Furuya KN, Kane J, Laukaitis C, Mengel E, Neilan EG, Nightingale S, Peters H, Scarpa M, Schwab KO, Smolka V, Valayannopoulos V, Wood M, Goodman Z, Yang Y, Eckert S, Rojas-Caro S, Quinn AG. A Phase 3 Trial of Sebelipase Alfa in Lysosomal Acid Lipase Deficiency. N Engl J Med 2015; 373:1010-20. [PMID: 26352813 DOI: 10.1056/nejmoa1501365] [Citation(s) in RCA: 168] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
BACKGROUND Lysosomal acid lipase is an essential lipid-metabolizing enzyme that breaks down endocytosed lipid particles and regulates lipid metabolism. We conducted a phase 3 trial of enzyme-replacement therapy in children and adults with lysosomal acid lipase deficiency, an underappreciated cause of cirrhosis and severe dyslipidemia. METHODS In this multicenter, randomized, double-blind, placebo-controlled study involving 66 patients, we evaluated the safety and effectiveness of enzyme-replacement therapy with sebelipase alfa (administered intravenously at a dose of 1 mg per kilogram of body weight every other week); the placebo-controlled phase of the study was 20 weeks long and was followed by open-label treatment for all patients. The primary end point was normalization of the alanine aminotransferase level. Secondary end points included additional disease-related efficacy assessments, safety, and side-effect profile. RESULTS Substantial disease burden at baseline included a very high level of low-density lipoprotein cholesterol (≥190 mg per deciliter) in 38 of 66 patients (58%) and cirrhosis in 10 of 32 patients (31%) who underwent biopsy. A total of 65 of the 66 patients who underwent randomization completed the double-blind portion of the trial and continued with open-label treatment. At 20 weeks, the alanine aminotransferase level was normal in 11 of 36 patients (31%) in the sebelipase alfa group and in 2 of 30 (7%) in the placebo group (P=0.03), with mean changes from baseline of -58 U per liter versus -7 U per liter (P<0.001). With respect to prespecified key secondary efficacy end points, we observed improvements in lipid levels and reduction in hepatic fat content (P<0.001 for all comparisons, except P=0.04 for triglycerides). The number of patients with adverse events was similar in the two groups; most events were mild and were considered by the investigator to be unrelated to treatment. CONCLUSIONS Sebelipase alfa therapy resulted in a reduction in multiple disease-related hepatic and lipid abnormalities in children and adults with lysosomal acid lipase deficiency. (Funded by Synageva BioPharma and others; ARISE ClinicalTrials.gov number, NCT01757184.).
Collapse
Affiliation(s)
- Barbara K Burton
- From the Northwestern University Feinberg School of Medicine and the Ann and Robert H. Lurie Children's Hospital, Chicago (B.K.B.); Icahn School of Medicine, Mount Sinai, New York (M.B.), and Women and Children's Hospital of Buffalo, Buffalo (R.E.) - both in New York; Centre Hospitalier Universitaire Brabois-Hôpital d'Enfants, Vandoeuvre-lès-Nancy (F.F.), and University Hospital Necker-Enfants Malades and Imagine Institute, Paris (V.V.) - both in France; University Hospital Center Zagreb and University of Zagreb, School of Medicine, Zagreb, Croatia (I.B.); Cincinnati Children's Hospital Medical Center, Cincinnati (T.A.B.); Hospital Universitario La Paz, Madrid (C.C.G.); Ege University Medical Faculty, Izmir (M.C.), and Gazi University Medical Faculty, Ankara (F.E.) - both in Turkey; Hospital Infantil de México Federico Gómez, Mexico City (A.C.-S.); Cambridge University Hospitals, Cambridge, United Kingdom (P.D.); Unit of Rare Diseases, Department of Pediatrics, Gaslini Institute, Genoa (M.D.R.), and University of Padua, Padua (M.S.) - both in Italy; Stanford University, Palo Alto (G.M.E.), and University of California, San Francisco, San Francisco ( J.K.) - both in California; Children's Hospital of Philadelphia, Philadelphia (C.F.); Alfred I. duPont Hospital for Children, Wilmington, DE (K.N.F.); University of Arizona Cancer Center, Tucson (C.L.); Villa Metabolica, Center of Pediatric and Adolescent Medicine, University of Mainz, Mainz (E.M.), and University Hospital Freiburg, Freiburg (K.O.S.) - both in Germany; Boston Children's Hospital, Boston (E.G.N.), and Synageva BioPharma, Lexington (Y.Y., S.E., S.R.-C., A.G.Q.) - both in Massachusetts; John Hunter Children's Hospital, and Discipline of Paediatrics and Child Health, University of Newcastle, Newcastle, NSW (S.N.), Royal Children's Hospital, Parkville, VIC (H.P.), and Royal Brisbane and Women's Hospital, Brisbane, QLD (M.W.) - all in Australia; Faculty Hospital, Palacky University, Olomouc, Czech Republic
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
22
|
Baratta F, Pastori D, Del Ben M, Polimeni L, Labbadia G, Di Santo S, Piemonte F, Tozzi G, Violi F, Angelico F. Reduced Lysosomal Acid Lipase Activity in Adult Patients With Non-alcoholic Fatty Liver Disease. EBioMedicine 2015; 2:750-754. [PMID: 26288848 PMCID: PMC4534687 DOI: 10.1016/j.ebiom.2015.05.018] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2015] [Revised: 05/18/2015] [Accepted: 05/19/2015] [Indexed: 12/15/2022] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) is characterized by intra-hepatic fat accumulation and mechanisms involved in its pathogenesis are not fully explained. Lysosomal Acid Lipase (LAL) is a key enzyme in lipid metabolism. We investigated its activity in patients with fatty liver. LAL activity (nmol/spot/h) was measured in 100 adult healthy subjects (HS) and in 240 NAFLD patients. A sub-analysis on 35 patients with biopsy-proven non-alcoholic steatohepatitis (NASH) was performed. Median LAL activity was 1.15 (0.95-1.72) in HS. It was significantly reduced in NAFLD [0.78 (0.61-1.01), p < 0.001 vs. HS]. A further reduction was observed in the subgroup of NASH [0.67 (0.51-0.77), p < 0.001 vs. HS]. Patients with LAL activity below median had higher values of serum total cholesterol (p < 0.05) and LDL-c (p < 0.05), and increased serum liver enzymes (ALT, p < 0.001; AST, p < 0.01; GGT, p < 0.01). At multivariable logistic regression analysis, factors associated with LAL activity below median were ALT (OR: 1.018, 95% CI 1.004-1.032, p = 0.011) and metabolic syndrome (OR: 2.551, 95% CI 1.241-5.245, p = 0.011), whilst statin use predicted a better LAL function (OR: 0.464, 95% CI 0.248-0.866, p = 0.016). Our findings suggest a strong association between impaired LAL activity and NAFLD. A better knowledge of the role of LAL may provide new insights in NAFLD pathogenesis.
Collapse
Affiliation(s)
- Francesco Baratta
- Department of Internal Medicine and Medical Specialties, Sapienza University of Rome, Italy
- Department of Anatomical, Histological, Forensic Medicine and Orthopedics Sciences, Sapienza University of Rome, Italy
| | - Daniele Pastori
- Department of Internal Medicine and Medical Specialties, Sapienza University of Rome, Italy
- Department of Anatomical, Histological, Forensic Medicine and Orthopedics Sciences, Sapienza University of Rome, Italy
| | - Maria Del Ben
- Department of Internal Medicine and Medical Specialties, Sapienza University of Rome, Italy
| | - Licia Polimeni
- Department of Internal Medicine and Medical Specialties, Sapienza University of Rome, Italy
- Department of Anatomical, Histological, Forensic Medicine and Orthopedics Sciences, Sapienza University of Rome, Italy
| | - Giancarlo Labbadia
- Department of Internal Medicine and Medical Specialties, Sapienza University of Rome, Italy
| | - Serena Di Santo
- Department of Public Health and Infectious Diseases, Sapienza University of Rome, Italy
| | - Fiorella Piemonte
- Unit for Neuromuscular and Neurodegenerative Diseases, Children's Hospital and Research Institute “Bambino Gesù”, Rome, Italy
| | - Giulia Tozzi
- Unit for Neuromuscular and Neurodegenerative Diseases, Children's Hospital and Research Institute “Bambino Gesù”, Rome, Italy
| | - Francesco Violi
- Department of Internal Medicine and Medical Specialties, Sapienza University of Rome, Italy
| | - Francesco Angelico
- Department of Public Health and Infectious Diseases, Sapienza University of Rome, Italy
| |
Collapse
|
23
|
Santillán-Hernández Y, Almanza-Miranda E, Xin WW, Goss K, Vera-Loaiza A, Mora MTGDL, Piña-Aguilar RE. Novel LIPA mutations in Mexican siblings with lysosomal acid lipase deficiency. World J Gastroenterol 2015; 21:1001-8. [PMID: 25624737 PMCID: PMC4299316 DOI: 10.3748/wjg.v21.i3.1001] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/18/2014] [Revised: 08/12/2014] [Accepted: 09/29/2014] [Indexed: 02/06/2023] Open
Abstract
Lysosomal acid lipase (LAL) deficiency is an under-recognized lysosomal disease caused by deficient enzymatic activity of LAL. In this report we describe two affected female Mexican siblings with early hepatic complications. At two months of age, the first sibling presented with alternating episodes of diarrhea and constipation, and later with hepatomegaly, elevated transaminases, high levels of total and low-density lipoprotein cholesterol, and low levels of high-density lipoprotein. Portal hypertension and grade 2 esophageal varices were detected at four years of age. The second sibling presented with hepatomegaly, elevated transaminases and mildly elevated low-density lipoprotein and low high-density lipoprotein at six months of age. LAL activity was deficient in both patients. Sequencing of LIPA revealed two previously unreported heterozygous mutations in exon 4: c.253C>A and c.294C>G. These cases highlight the clinical continuum between the so-called Wolman disease and cholesteryl ester storage disease, and underscore that LAL deficiency represents a single disease with a degree of clinical heterogeneity.
Collapse
|
24
|
Lopez AM, Posey KS, Turley SD. Deletion of sterol O-acyltransferase 2 (SOAT2) function in mice deficient in lysosomal acid lipase (LAL) dramatically reduces esterified cholesterol sequestration in the small intestine and liver. Biochem Biophys Res Commun 2014; 454:162-6. [PMID: 25450374 PMCID: PMC4312202 DOI: 10.1016/j.bbrc.2014.10.063] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2014] [Accepted: 10/13/2014] [Indexed: 11/21/2022]
Abstract
Sterol O-acyltransferase 2 (SOAT2), also known as ACAT2, is the major cholesterol esterifying enzyme in the liver and small intestine (SI). Esterified cholesterol (EC) carried in certain classes of plasma lipoproteins is hydrolyzed by lysosomal acid lipase (LAL) when they are cleared from the circulation. Loss-of-function mutations in LIPA, the gene that encodes LAL, result in Wolman disease (WD) or cholesteryl ester storage disease (CESD). Hepatomegaly and a massive increase in tissue EC levels are hallmark features of both disorders. While these conditions can be corrected with enzyme replacement therapy, the question arose as to what effect the loss of SOAT2 function might have on tissue EC sequestration in LAL-deficient mice. When weaned at 21 days, Lal(-)(/)(-):Soat2(+)(/)(+) mice had a whole liver cholesterol content (mg/organ) of 24.7 mg vs 1.9mg in Lal(+/+):Soat2(+/+) littermates, with almost all the excess sterol being esterified. Over the next 31 days, liver cholesterol content in the Lal(-)(/)(-):Soat2(+)(/)(+) mice increased to 145 ± 2 mg but to only 29 ± 2 mg in their Lal(-)(/)(-):Soat2(-)(/)(-) littermates. The level of EC accumulation in the SI of the Lal(-)(/)(-):Soat2(-)(/)(-) mice was also much less than in their Lal(-)(/)(-):Soat2(+)(/)(+) littermates. In addition, there was a >70% reduction in plasma transaminase activities in the Lal(-)(/)(-):Soat2(-)(/)(-) mice. These studies illustrate how the severity of disease in a mouse model for CESD can be substantially ameliorated by elimination of SOAT2 function.
Collapse
Affiliation(s)
- Adam M Lopez
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390-9151, United States.
| | - Kenneth S Posey
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390-9151, United States.
| | - Stephen D Turley
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390-9151, United States.
| |
Collapse
|
25
|
Sebelipase alfa over 52 weeks reduces serum transaminases, liver volume and improves serum lipids in patients with lysosomal acid lipase deficiency. J Hepatol 2014; 61:1135-42. [PMID: 24993530 PMCID: PMC4203712 DOI: 10.1016/j.jhep.2014.06.022] [Citation(s) in RCA: 72] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/10/2014] [Revised: 05/21/2014] [Accepted: 06/23/2014] [Indexed: 02/06/2023]
Abstract
BACKGROUND & AIMS Lysosomal acid lipase deficiency is an autosomal recessive enzyme deficiency resulting in lysosomal accumulation of cholesteryl esters and triglycerides. LAL-CL04, an ongoing extension study, investigates the long-term effects of sebelipase alfa, a recombinant human lysosomal acid lipase. METHODS Sebelipase alfa (1mg/kg or 3mg/kg) was infused every-other-week to eligible subjects. Safety and tolerability assessments, including liver function, lipid profiles and liver volume assessment, were carried out at regular intervals. RESULTS 216 infusions were administered to eight adult subjects through week 52 during LAL-CL04. At week 52, mean alanine aminotransferase and aspartate aminotransferase levels were normal with mean change from baseline of -58% and -40%. Mean changes for low-density lipoprotein, total cholesterol, triglyceride and high-density lipoprotein were -60%, -39%, -36%, and +29%, respectively. Mean liver volume by magnetic resonance imaging and hepatic proton density fat fraction decreased (12% and 55%, respectively). Adverse events were mainly mild and unrelated to sebelipase alfa. Infusion-related reactions were uncommon: three events of moderate severity were reported in two subjects; one patient's event was suggestive of a hypersensitivity-like reaction, but additional testing did not confirm this, and the subject has successfully re-started sebelipase alfa. Of samples tested to date, no anti-drug antibodies have been detected. CONCLUSIONS Long-term dosing with sebelipase alfa in lysosomal acid lipase-deficient patients is well tolerated and produces sustained reductions in transaminases, improvements in serum lipid profile and reduction in the hepatic fat fraction. A randomized, placebo-controlled phase 3 trial in children and adults is underway (ARISE: NCT01757184).
Collapse
|
26
|
Sun Y, Xu YH, Du H, Quinn B, Liou B, Stanton L, Inskeep V, Ran H, Jakubowitz P, Grilliot N, Grabowski GA. Reversal of advanced disease in lysosomal acid lipase deficient mice: a model for lysosomal acid lipase deficiency disease. Mol Genet Metab 2014; 112:229-41. [PMID: 24837159 DOI: 10.1016/j.ymgme.2014.04.006] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/20/2014] [Revised: 04/23/2014] [Accepted: 04/23/2014] [Indexed: 01/08/2023]
Abstract
Lysosomal acid lipase (LAL) is an essential enzyme that hydrolyzes triglycerides (TG) and cholesteryl esters (CE) in lysosomes. Mutations of the LIPA gene lead to Wolman disease (WD) and cholesterol ester storage disease (CESD). The disease hallmarks include hepatosplenomegaly and extensive storage of CE and/or TG. The effects of intravenous investigational enzyme therapy (ET) on survival and efficacy were evaluated in Lipa knock out, lal-/- mice with advanced disease using recombinant human LAL (rhLAL). Comparative ET was conducted with lower doses (weekly, 0.8 and 3.2mg/kg) beginning at 16 weeks (study 1), and with higher dose (10mg/kg) in early (8-weeks), middle (16-weeks) and late (24-weeks) disease stages (study 2). In study 1, rhLAL extended the life span of lal-/- mice in a dose dependent manner by 52 (0.8 mg/kg) or 94 (3.2mg/kg) days. This was accompanied by partial correction of cholesterol and TG levels in spleen and liver. In study 2, the high dose resulted in a significant improvement in organ size (liver, spleen and small intestine) and tissue histology as well as significant decreases in cholesterol and TG in all three groups. In the treated livers and spleens the cholesterol and TG levels were reduced to below treatment initiation levels indicating a reversal of disease manifestations, even in advanced disease. ET diminished liver fibrosis and macrophage proliferation. These results show that LAL deficiency can be improved biochemically and histopathologically by various dosages of ET, even in advanced disease.
Collapse
Affiliation(s)
- Ying Sun
- The Division of Human Genetics, Cincinnati Children's Hospital Research Foundation, USA; The Department of Pediatrics, University of Cincinnati College of Medicine Cincinnati, OH 45229, USA.
| | - You-Hai Xu
- The Division of Human Genetics, Cincinnati Children's Hospital Research Foundation, USA; The Department of Pediatrics, University of Cincinnati College of Medicine Cincinnati, OH 45229, USA
| | - Hong Du
- The Department of Pathology and Laboratory Medicine, Indiana University School of Medicine, Indianapolis, IN 46202-5120, USA
| | - Brian Quinn
- The Division of Human Genetics, Cincinnati Children's Hospital Research Foundation, USA
| | - Benjamin Liou
- The Division of Human Genetics, Cincinnati Children's Hospital Research Foundation, USA
| | - Lori Stanton
- The Division of Human Genetics, Cincinnati Children's Hospital Research Foundation, USA
| | - Venette Inskeep
- The Division of Human Genetics, Cincinnati Children's Hospital Research Foundation, USA
| | - Huimin Ran
- The Division of Human Genetics, Cincinnati Children's Hospital Research Foundation, USA
| | - Phillip Jakubowitz
- The Division of Human Genetics, Cincinnati Children's Hospital Research Foundation, USA; University of Cincinnati, OH 45221, USA
| | - Nicholas Grilliot
- The Division of Human Genetics, Cincinnati Children's Hospital Research Foundation, USA; University of Cincinnati, OH 45221, USA
| | - Gregory A Grabowski
- The Division of Human Genetics, Cincinnati Children's Hospital Research Foundation, USA; The Department of Pediatrics, University of Cincinnati College of Medicine Cincinnati, OH 45229, USA; Synageva BioPharma Corp., Lexington, MA 02421, USA
| |
Collapse
|
27
|
Reiner Ž, Guardamagna O, Nair D, Soran H, Hovingh K, Bertolini S, Jones S, Ćorić M, Calandra S, Hamilton J, Eagleton T, Ros E. Lysosomal acid lipase deficiency--an under-recognized cause of dyslipidaemia and liver dysfunction. Atherosclerosis 2014; 235:21-30. [PMID: 24792990 DOI: 10.1016/j.atherosclerosis.2014.04.003] [Citation(s) in RCA: 200] [Impact Index Per Article: 18.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/20/2013] [Revised: 04/04/2014] [Accepted: 04/05/2014] [Indexed: 12/15/2022]
Abstract
Lysosomal acid lipase deficiency (LAL-D) is a rare autosomal recessive lysosomal storage disease caused by deleterious mutations in the LIPA gene. The age at onset and rate of progression vary greatly and this may relate to the nature of the underlying mutations. Patients presenting in infancy have the most rapidly progressive disease, developing signs and symptoms in the first weeks of life and rarely surviving beyond 6 months of age. Children and adults typically present with some combination of dyslipidaemia, hepatomegaly, elevated transaminases, and microvesicular hepatosteatosis on biopsy. Liver damage with progression to fibrosis, cirrhosis and liver failure occurs in a large proportion of patients. Elevated low-density lipoprotein cholesterol levels and decreased high-density lipoprotein cholesterol levels are common features, and cardiovascular disease may manifest as early as childhood. Given that these clinical manifestations are shared with other cardiovascular, liver and metabolic diseases, it is not surprising that LAL-D is under-recognized in clinical practice. This article provides practical guidance to lipidologists, endocrinologists, cardiologists and hepatologists on how to recognize individuals with this life-limiting disease. A diagnostic algorithm is proposed with a view to achieving definitive diagnosis using a recently developed blood test for lysosomal acid lipase. Finally, current management options are reviewed in light of the ongoing development of enzyme replacement therapy with sebelipase alfa (Synageva BioPharma Corp., Lexington, MA, USA), a recombinant human lysosomal acid lipase enzyme.
Collapse
Affiliation(s)
- Željko Reiner
- University Hospital Center, School of Medicine, University of Zagreb, Kispaticeva 12, 10000 Zagreb, Croatia.
| | - Ornella Guardamagna
- Department of Public and Health Sciences, Medical School, University of Turin, Piazza Polonia 94, I-10126 Turin, Italy
| | - Devaki Nair
- Department of Clinical Biochemistry, Royal Free Hospital NHS Foundation Trust, Pond Street, London NW3 2QG, UK
| | - Handrean Soran
- Cardiovascular Trials Unit, Central Manchester University Hospitals NHS Foundation Trust, Oxford Road, Manchester M13 9WL, UK
| | - Kees Hovingh
- Department of Vascular Medicine, Academic Medical Center, Meibergdreef 9, Amsterdam 1105 AZ, Netherlands
| | - Stefano Bertolini
- Department of Internal Medicine, University of Genoa, Viale Benedetto XV n. 6, 16132 Genoa, Italy
| | - Simon Jones
- Willink Biochemical Genetics Unit, Manchester Centre for Genomic Medicine, Central Manchester University Hospitals NHS Foundation Trust, St. Mary's Hospital, Oxford Road, Manchester M13 9WL, UK
| | - Marijana Ćorić
- University Hospital Center, School of Medicine, University of Zagreb, Kispaticeva 12, 10000 Zagreb, Croatia
| | - Sebastiano Calandra
- Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, Via Campi 287, I-41125 Modena, Italy
| | - John Hamilton
- Biochemistry Department, Yorkhill Hospital, Glasgow, G3 8SJ, UK
| | - Terence Eagleton
- Synageva BioPharma Corp., 33 Hayden Ave., Lexington, MA 02421, USA
| | - Emilio Ros
- Lipid Clinic, Endocrinology & Nutrition Service, Institut d'Investigations Biomèdiques August Pi Sunyer, Hospital Clínic, C. Villarroel, 170, 08036 Barcelona, Spain; CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), Instituto de Salud Carloss III (ISCIII), Spain.
| |
Collapse
|
28
|
Stitziel NO, Fouchier SW, Sjouke B, Peloso GM, Moscoso AM, Auer PL, Goel A, Gigante B, Barnes TA, Melander O, Orho-Melander M, Duga S, Sivapalaratnam S, Nikpay M, Martinelli N, Girelli D, Jackson RD, Kooperberg C, Lange LA, Ardissino D, McPherson R, Farrall M, Watkins H, Reilly MP, Rader DJ, de Faire U, Schunkert H, Erdmann J, Samani NJ, Charnas L, National Heart, Lung and Blood Institute GO Exome Sequencing Project, Altshuler D, Gabriel S, Kastelein JJ, Defesche JC, Nederveen AJ, Kathiresan S, Hovingh GK. Exome sequencing and directed clinical phenotyping diagnose cholesterol ester storage disease presenting as autosomal recessive hypercholesterolemia. Arterioscler Thromb Vasc Biol 2013; 33:2909-14. [PMID: 24072694 PMCID: PMC4002172 DOI: 10.1161/atvbaha.113.302426] [Citation(s) in RCA: 75] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2013] [Accepted: 09/10/2013] [Indexed: 01/12/2023]
Abstract
OBJECTIVE Autosomal recessive hypercholesterolemia is a rare inherited disorder, characterized by extremely high total and low-density lipoprotein cholesterol levels, that has been previously linked to mutations in LDLRAP1. We identified a family with autosomal recessive hypercholesterolemia not explained by mutations in LDLRAP1 or other genes known to cause monogenic hypercholesterolemia. The aim of this study was to identify the molecular pathogenesis of autosomal recessive hypercholesterolemia in this family. APPROACH AND RESULTS We used exome sequencing to assess all protein-coding regions of the genome in 3 family members and identified a homozygous exon 8 splice junction mutation (c.894G>A, also known as E8SJM) in LIPA that segregated with the diagnosis of hypercholesterolemia. Because homozygosity for mutations in LIPA is known to cause cholesterol ester storage disease, we performed directed follow-up phenotyping by noninvasively measuring hepatic cholesterol content. We observed abnormal hepatic accumulation of cholesterol in the homozygote individuals, supporting the diagnosis of cholesterol ester storage disease. Given previous suggestions of cardiovascular disease risk in heterozygous LIPA mutation carriers, we genotyped E8SJM in >27 000 individuals and found no association with plasma lipid levels or risk of myocardial infarction, confirming a true recessive mode of inheritance. CONCLUSIONS By integrating observations from Mendelian and population genetics along with directed clinical phenotyping, we diagnosed clinically unapparent cholesterol ester storage disease in the affected individuals from this kindred and addressed an outstanding question about risk of cardiovascular disease in LIPA E8SJM heterozygous carriers.
Collapse
Affiliation(s)
- Nathan O. Stitziel
- Cardiovascular Division, Department of Medicine, Washington University School of Medicine, Saint Louis MO 63110, USA
- Division of Statistical Genomics, Washington University School of Medicine, Saint Louis MO 63110, USA
| | - Sigrid W. Fouchier
- Department of Vascular Medicine, Academic Medical Center, Amsterdam, 1105 AZ, The Netherlands
- Department of Experimental Vascular Medicine, Academic Medical Center, Amsterdam, 1105 AZ, The Netherlands
| | - Barbara Sjouke
- Department of Vascular Medicine, Academic Medical Center, Amsterdam, 1105 AZ, The Netherlands
- Department of Experimental Vascular Medicine, Academic Medical Center, Amsterdam, 1105 AZ, The Netherlands
| | - Gina M. Peloso
- Center for Human Genetic Research, Boston MA, 02114, USA
- Program in Medical and Population Genetics, Broad Institute, Cambridge MA 02142, USA
| | - Alessa M. Moscoso
- Center for Human Genetic Research, Boston MA, 02114, USA
- Program in Medical and Population Genetics, Broad Institute, Cambridge MA 02142, USA
| | - Paul L. Auer
- Fred Hutchinson Cancer Research Center, Seattle WA 98109, USA
- School of Public Health, University of Wisconsin-Milwaukee, Milwaukee, WI, 53201
| | - Anuj Goel
- Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, UK
- Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford, UK
| | - Bruna Gigante
- Division of Cardiovascular Epidemiology, Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Timothy A. Barnes
- Department of Cardiovascular Sciences, University of Leicester and Leicester NIHR Biomedical Research Unit in Cardiovascular Disease, Glenfield Hospital, Leicester, LE3 9QP, UK
| | - Olle Melander
- Department of Clinical Sciences, Hypertension and Cardiovascular Diseases, Skania University Hospital, Lund University, Malmö, Sweden
| | - Marju Orho-Melander
- Diabetes and Cardiovascular Disease Genetic Epidemiology, Skania University Hospital, Lund University, Malmö, Sweden
| | - Stefano Duga
- Dipartimento di Biotecnologie Mediche e Medicina Traslazionale, Università degli Studi di Milano
| | - Suthesh Sivapalaratnam
- Department of Vascular Medicine, Academic Medical Center, Amsterdam, 1105 AZ, The Netherlands
- Department of Experimental Vascular Medicine, Academic Medical Center, Amsterdam, 1105 AZ, The Netherlands
| | - Majid Nikpay
- Atherogenomics Laboratory, University of Ottawa Heart Institute, Ottawa, ON, Canada
| | | | | | - Rebecca D. Jackson
- Division of Endocrinology, Diabetes and Metabolism, Ohio State University, Columbus, OH 43210, USA
| | | | - Leslie A. Lange
- Departments of Epidemiology, Genetics and Biostatistics, Bioinformatics and Computational Biology, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Diego Ardissino
- Division of Cardiology, Azienda Ospedaliero-Universitaria di Parma, Parma, Italy
| | - Ruth McPherson
- The John & Jennifer Ruddy Canadian Cardiovascular Genetics Centre, University of Ottawa Heart Institute, Ottawa, ON, Canada
| | - Martin Farrall
- Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, UK
- Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford, UK
| | - Hugh Watkins
- Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, UK
- Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford, UK
| | - Muredach P. Reilly
- The Institute for Translational Medicine and Therapeutics and The Cardiovascular Institute, Perleman School of Medicine at the University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Daniel J. Rader
- The Institute for Translational Medicine and Therapeutics and The Cardiovascular Institute, Perleman School of Medicine at the University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Ulf de Faire
- Division of Cardiovascular Epidemiology, Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Heribert Schunkert
- Deutsches Herzzentrum München, München, Germany
- Technische Universität München, München, Germany
- DZHK (German Research Centre for Cardiovascular Research), partner site München, München, Germany
| | - Jeanette Erdmann
- Institut für Integrative und Experimentelle Genomik, Universität zu Lübeck, Lübeck, Germany
- DZHK (German Research Centre for Cardiovascular Research), partner site Hamburg/Kiel/Lübeck, Lübeck, Germany
| | - Nilesh J. Samani
- Department of Cardiovascular Sciences, University of Leicester and Leicester NIHR Biomedical Research Unit in Cardiovascular Disease, Glenfield Hospital, Leicester, LE3 9QP, UK
| | | | | | - David Altshuler
- Center for Human Genetic Research, Boston MA, 02114, USA
- Program in Medical and Population Genetics, Broad Institute, Cambridge MA 02142, USA
- Department of Molecular Biology, Massachusetts General Hospital, Boston, MA, 02114, USA
- Department of Genetics, Harvard Medical School, Boston, MA, 02115, USA
| | - Stacey Gabriel
- Program in Medical and Population Genetics, Broad Institute, Cambridge MA 02142, USA
| | - John J.P. Kastelein
- Department of Vascular Medicine, Academic Medical Center, Amsterdam, 1105 AZ, The Netherlands
- Department of Experimental Vascular Medicine, Academic Medical Center, Amsterdam, 1105 AZ, The Netherlands
| | - Joep C. Defesche
- Department of Vascular Medicine, Academic Medical Center, Amsterdam, 1105 AZ, The Netherlands
- Department of Experimental Vascular Medicine, Academic Medical Center, Amsterdam, 1105 AZ, The Netherlands
| | - Aart J. Nederveen
- Department of Vascular Medicine, Academic Medical Center, Amsterdam, 1105 AZ, The Netherlands
- Department of Experimental Vascular Medicine, Academic Medical Center, Amsterdam, 1105 AZ, The Netherlands
| | - Sekar Kathiresan
- Center for Human Genetic Research, Boston MA, 02114, USA
- Program in Medical and Population Genetics, Broad Institute, Cambridge MA 02142, USA
- Cardiovascular Research Center, Massachusetts General Hospital, Boston, MA, 02114, USA
- Department of Medicine, Harvard Medical School, Boston, MA, 02115, USA
| | - G. Kees Hovingh
- Department of Vascular Medicine, Academic Medical Center, Amsterdam, 1105 AZ, The Netherlands
- Department of Experimental Vascular Medicine, Academic Medical Center, Amsterdam, 1105 AZ, The Netherlands
| |
Collapse
|