1
|
Vejux A, Ghzaiel I, Mackrill JJ, Dias IHK, Rezig L, Ksila M, Zarrouk A, Nury T, Brahmi F, El Midaoui A, Meziane S, Atanasov AG, Hammami S, Latruffe N, Jouanny P, Lizard G. Oxysterols, age-related-diseases and nutritherapy: Focus on 7-ketocholesterol and 7β-hydroxycholesterol. Prostaglandins Other Lipid Mediat 2025; 178:106993. [PMID: 40216356 DOI: 10.1016/j.prostaglandins.2025.106993] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2024] [Revised: 03/31/2025] [Accepted: 04/08/2025] [Indexed: 04/25/2025]
Abstract
Age-related diseases are often associated with a disruption of RedOx balance that can lead to lipid peroxidation with the formation of oxysterols, especially those oxidized on carbon-7: 7-ketocholesterol (also known as 7-oxo-cholesterol) and 7β-hydroxycholesterol. Like cholesterol, these oxysterols have 27 carbons, they are composed of a sterane nucleus and have a hydroxyl function in position 3. The oxysterols 7-ketocholesterol and 7β-hydroxycholesterol are mainly formed by cholesterol autoxidation and are biomarkers of oxidative stress. These two oxysterols are frequently found at increased levels in the biological fluids (plasma, cerebrospinal fluid), tissues and/or organs (arterial wall, retina, brain) of patients with age-related diseases, especially cardiovascular diseases, neurodegenerative diseases (mainly Alzheimer's disease), ocular diseases (cataract, age-related macular degeneration), and sarcopenia. Depending on the cell type considered, 7-ketocholesterol and 7β-hydroxycholesterol induce either caspase- dependent or -independent types of cell death associated with mitochondrial and peroxisomal dysfunctions, autophagy and oxidative stress. The caspase dependent type of cell death associated with oxidative stress and autophagy is defined as oxiapoptophagy. These two oxysterols are also inducers of inflammation. These biological features associated with the toxicity of 7-ketocholesterol, and 7β-hydroxycholesterol are often observed in patients with age-related diseases, suggesting an involvement of these oxysterols in the pathophysiology of these disorders. The cytotoxic effects of 7-ketocholesterol and 7β-hydroxycholesterol are counteracted on different cell models by representative nutrients of the Mediterranean diet: ω3 and ω9 fatty acids, polyphenols, and tocopherols. There are also evidences, mainly in cardiovascular diseases, of the benefits of α-tocopherol and phenolic compounds. These in vitro and in vivo observations on 7-ketocholesterol and 7β-hydroxycholesterol, which are frequently increased in age-related diseases, reinforce the interest of nutritherapeutic treatments to prevent and/or cure age-related diseases currently without effective therapies.
Collapse
Affiliation(s)
- Anne Vejux
- Centre des Sciences du Goût et de l'Alimentation, CNRS, INRAE, Institut Agro Dijon, Université de Bourgogne Europe, 21000 Dijon, France; Team 'Biochemistry of the Peroxisome, Inflammation and Lipid Metabolism' EA7270/Inserm, Université de Bourgogne Europe, 21000 Dijon, France.
| | - Imen Ghzaiel
- Team 'Biochemistry of the Peroxisome, Inflammation and Lipid Metabolism' EA7270/Inserm, Université de Bourgogne Europe, 21000 Dijon, France; Lab-NAFS 'Nutrition-Functional Food & Vascular Health', Faculty of Medicine, University of Monastir, LR12ES05, Monastir 5000, Tunisia
| | - John J Mackrill
- Department of Physiology, University College Cork, Western Gateway Building, Western Road, Cork T12 XF62, Ireland
| | - Irundika H K Dias
- Aston Medical School, College of Health and Life Sciences, Aston University, Birmingham, UK
| | - Leila Rezig
- University of Carthage, National Institute of Applied Sciences and Technology, LR11ES26, LIP-MB 'Laboratory of Protein Engineering and Bioactive Molecules', Tunis 1080, Tunisia; University of Carthage, High Institute of Food Industries, 58 Alain Savary Street, El Khadra City, Tunis 1003, Tunisia
| | - Mohamed Ksila
- Team 'Biochemistry of the Peroxisome, Inflammation and Lipid Metabolism' EA7270/Inserm, Université de Bourgogne Europe, 21000 Dijon, France
| | - Amira Zarrouk
- Lab-NAFS 'Nutrition-Functional Food & Vascular Health', Faculty of Medicine, University of Monastir, LR12ES05, Monastir 5000, Tunisia; Faculty of Medicine of Sousse, avenue Mohamed Karaoui, 4002 Sousse, Tunisia
| | - Thomas Nury
- Team 'Biochemistry of the Peroxisome, Inflammation and Lipid Metabolism' EA7270/Inserm, Université de Bourgogne Europe, 21000 Dijon, France
| | - Fatiha Brahmi
- Laboratory of Biomathematics, Biophysics, Biochemistry, and Scientometrics (L3BS), Faculty of Nature and Life Sciences, University of Bejaia, 06000 Bejaia, Algeria
| | - Adil El Midaoui
- Department of Biology, Faculty of Sciences and Techniques of Errachidia, Moulay Ismail University of Meknes, Meknes 50050, Morocco; Department of Pharmacology and Physiology, Faculty of Medicine, University of Montreal, Montreal, QC H3C 3J7, Canada
| | - Smail Meziane
- Institut Européen des Antioxydants, 1B Rue Victor de Lespinats, 54230 Neuves-Maisons, France
| | - Atanas G Atanasov
- Institute of Genetics and Animal Biotechnology of the Polish Academy of Sciences, Magdalenka, Jastrzebiec, Poland
| | - Sonia Hammami
- Lab-NAFS 'Nutrition-Functional Food & Vascular Health', Faculty of Medicine, University of Monastir, LR12ES05, Monastir 5000, Tunisia
| | - Norbert Latruffe
- Centre des Sciences du Goût et de l'Alimentation, CNRS, INRAE, Institut Agro Dijon, Université de Bourgogne Europe, 21000 Dijon, France; Team 'Biochemistry of the Peroxisome, Inflammation and Lipid Metabolism' EA7270/Inserm, Université de Bourgogne Europe, 21000 Dijon, France
| | - Pierre Jouanny
- Geriatric Internal Medicine Department (Champmaillot), University Hospital Center, Université de Bourgogne Europe, 21000 Dijon, France
| | - Gérard Lizard
- Team 'Biochemistry of the Peroxisome, Inflammation and Lipid Metabolism' EA7270/Inserm, Université de Bourgogne Europe, 21000 Dijon, France.
| |
Collapse
|
2
|
de Castro Lopez MJ, White FJ, Holmes V, Roberts J, Wu THY, Cooper JA, Church HJ, Petts G, Wynn RF, Jones SA, Ghosh A. Does Early Diagnosis and Treatment Alter the Clinical Course of Wolman Disease? Divergent Trajectories in Two Siblings and a Consideration for Newborn Screening. Int J Neonatal Screen 2025; 11:17. [PMID: 40136632 PMCID: PMC11943304 DOI: 10.3390/ijns11010017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/03/2024] [Revised: 01/03/2025] [Accepted: 01/15/2025] [Indexed: 03/27/2025] Open
Abstract
Wolman disease (WD) is a lethal disorder defined by the deficiency of the lysosomal acid lipase enzyme. Patients present with intestinal failure, malnutrition, and hepatosplenomegaly. Enzyme replacement therapy (ERT) with dietary substrate reduction (DSR) significantly improves survival. We sought to determine the outcomes of two siblings with WD treated after the onset of symptoms (sibling 1) and presymptomatic (sibling 2). A chart review was conducted on two siblings with WD treated with ERT and DSR at 4 months of age (sibling 1) and immediately after birth (sibling 2) to determine clinical outcomes based on survival, laboratory results, growth, dietary records, and gut biopsies. Sibling 1 presented with hepatosplenomegaly and liver dysfunction and developed hemophagocytic lymphohistiocytosis despite treatment. She received a bone marrow transplant at 8 months of age but died at 13 months. Sibling 2 is alive at 16 months of age with height, weight, and MUAC above the 95th centile, fully orally fed, with no gastrointestinal symptoms, normal liver function, and normal oxysterols. Sibling 2 duodenal biopsies show normal villus architecture with no foamy macrophage infiltration. Initiation of treatment prior to the onset of symptoms can prevent clinical manifestations and increase survival. The divergent trajectory in these siblings raises the question of WD's candidacy for newborn screening.
Collapse
Affiliation(s)
- Maria Jose de Castro Lopez
- Manchester Centre for Genomic Medicine, Manchester University NHS Foundation Trust, Oxford Road, Manchester M13 9WL, UK; (M.J.d.C.L.); (F.J.W.); (V.H.); (J.R.); (T.H.Y.W.); (H.J.C.); (A.G.)
| | - Fiona J. White
- Manchester Centre for Genomic Medicine, Manchester University NHS Foundation Trust, Oxford Road, Manchester M13 9WL, UK; (M.J.d.C.L.); (F.J.W.); (V.H.); (J.R.); (T.H.Y.W.); (H.J.C.); (A.G.)
- Department of Therapy and Dietetics, Manchester University NHS Foundation Trust, Oxford Road, Manchester M13 9WL, UK
| | - Victoria Holmes
- Manchester Centre for Genomic Medicine, Manchester University NHS Foundation Trust, Oxford Road, Manchester M13 9WL, UK; (M.J.d.C.L.); (F.J.W.); (V.H.); (J.R.); (T.H.Y.W.); (H.J.C.); (A.G.)
- Department of Therapy and Dietetics, Manchester University NHS Foundation Trust, Oxford Road, Manchester M13 9WL, UK
| | - Jane Roberts
- Manchester Centre for Genomic Medicine, Manchester University NHS Foundation Trust, Oxford Road, Manchester M13 9WL, UK; (M.J.d.C.L.); (F.J.W.); (V.H.); (J.R.); (T.H.Y.W.); (H.J.C.); (A.G.)
| | - Teresa H. Y. Wu
- Manchester Centre for Genomic Medicine, Manchester University NHS Foundation Trust, Oxford Road, Manchester M13 9WL, UK; (M.J.d.C.L.); (F.J.W.); (V.H.); (J.R.); (T.H.Y.W.); (H.J.C.); (A.G.)
| | - James A. Cooper
- Manchester Centre for Genomic Medicine, Manchester University NHS Foundation Trust, Oxford Road, Manchester M13 9WL, UK; (M.J.d.C.L.); (F.J.W.); (V.H.); (J.R.); (T.H.Y.W.); (H.J.C.); (A.G.)
| | - Heather J. Church
- Manchester Centre for Genomic Medicine, Manchester University NHS Foundation Trust, Oxford Road, Manchester M13 9WL, UK; (M.J.d.C.L.); (F.J.W.); (V.H.); (J.R.); (T.H.Y.W.); (H.J.C.); (A.G.)
| | - Gemma Petts
- Department of Paediatric Histopathology, Royal Manchester Children’s Hospital, Oxford Road, Manchester M13 9WL, UK;
| | - Robert F. Wynn
- Department of Blood and Marrow Transplantation, Royal Manchester Children’s Hospital, Oxford Road, Manchester M13 9WL, UK;
| | - Simon A. Jones
- Manchester Centre for Genomic Medicine, Manchester University NHS Foundation Trust, Oxford Road, Manchester M13 9WL, UK; (M.J.d.C.L.); (F.J.W.); (V.H.); (J.R.); (T.H.Y.W.); (H.J.C.); (A.G.)
| | - Arunabha Ghosh
- Manchester Centre for Genomic Medicine, Manchester University NHS Foundation Trust, Oxford Road, Manchester M13 9WL, UK; (M.J.d.C.L.); (F.J.W.); (V.H.); (J.R.); (T.H.Y.W.); (H.J.C.); (A.G.)
| |
Collapse
|
3
|
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
|
4
|
Canbay E, Canda E, Yazıcı H, Kasıkcı GK, Durmaz B, Copur O, Tahhan B, Düzgün D, Koru ZE, Sezer E, Aydın D, Levent RE, Ucar SK, Coker M, Sozmen EY. Determination of selected oxysterol levels, oxidative stress, and macrophage activation indicators in children and adolescents with familial hypercholesterolemia. Lipids Health Dis 2024; 23:374. [PMID: 39538217 PMCID: PMC11562613 DOI: 10.1186/s12944-024-02371-y] [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: 07/03/2024] [Accepted: 11/10/2024] [Indexed: 11/16/2024] Open
Abstract
AIM Elevated levels of cholesterol in the bloodstream, also referred to as hypercholesterolemia, pose a significant risk for the onset of cardiovascular and cerebrovascular diseases. Oxysterols, cholesterol-derived oxidized compounds that form enzymatically or non-enzymatically, contribute to the development of atherosclerosis and coronary artery disease. This study aimed to examine the critical oxysterol levels in children and adolescents with hypercholesterolemia and explore the correlation between these levels, oxidative stress, and atherosclerosis progression. MATERIALS AND METHODS The study included 20 patients with familial hypercholesterolemia (FH) and 20 healthy individuals aged between 6 and 18 years. Participants were categorized into children (6-9 years) and adolescents (10-18 years). Pediatric and adolescent patients were selected from among subjects with LDL-C ≥ 130 mg/dL and diagnosed with heterozygous familial hypercholesterolemia (HeFH) based on the presence of mutations in the LDL receptor (LDL-R) gene. Patients with HeFH who were receiving regular atorvastatin therapy were included in the study. RESULTS There were no notable differences in catalase and paraoxonase (PON1) activities among the groups. However, the patient group displayed substantially higher levels of malondialdehyde (MDA) (P = 0.0108) and superoxide dismutase (SOD) activity (P = 0.0103). Compared to the healthy control group, serum chitotriosidase (CHITO) activity (P = 0.037) and chitinase 3-like protein 1 (YKL-40) levels (P = 0.0027) were significantly elevated in the patient group. Furthermore, the carotid intima-media thickness (CIMT) measurements of the patient group were significantly greater than those of the healthy group (**P < 0.0001****). The patient group exhibited significantly elevated levels of 5,6-α-epoxycholesterol, Cholestane-3β,5α,6β-triol (C-triol), and 7-ketocholesterol (7-KC), whereas 27-hydroxycholesterol (27-OHC) was significantly more abundant in the healthy group. On the other hand, while 27-OHC/Total cholesterol (Total-C) levels were significantly higher in healthy individuals, the C-Triol/Total-C ratio was significantly higher in patients. No significant differences were found between the groups in terms of 7-KC/Total-C and 5,6-α-epoxycholesterol/Total-C levels. CONCLUSION This study highlights the key roles of oxysterols, oxidative stress, and macrophage activation in the development of atherosclerosis in pediatric and adolescent patients with FH. Elevated C-Triol levels in FH patients, alongside increased CIMT, point to early vascular changes despite atorvastatin therapy. In contrast, higher 27-OHC levels in healthy controls suggest differential oxysterol regulation due to cholesterol-lowering treatments in FH patients. C-Triol and 27-OHC/Total-C ratios showed potential as biomarkers to distinguish patients with FH. These findings emphasize the need for therapies targeting oxidative stress and macrophage activation in addition to cholesterol-lowering interventions.
Collapse
Affiliation(s)
- Erhan Canbay
- Department of Medical Biochemistry, Faculty of Medicine, Ege University, Bornova, Izmir, 35100, Turkey.
| | - Ebru Canda
- Department of Pediatric Metabolic Disease, Faculty of Medicine, Ege University, Bornova, Izmir, Turkey
| | - Havva Yazıcı
- Department of Pediatric Metabolic Disease, Faculty of Medicine, Ege University, Bornova, Izmir, Turkey
| | - Gulcin Kayan Kasıkcı
- Department of Pediatric Cardiology, Faculty of Medicine, Ege University Izmir, Bornova, Izmir, Turkey
| | - Burak Durmaz
- Department of Medical Biochemistry, Faculty of Medicine, Ege University, Bornova, Izmir, 35100, Turkey
- Department of Medical Biochemistry, Faculty of Medicine, Near East University, Nicosia, Cyprus
| | - Oznur Copur
- Department of Medical Biochemistry, Faculty of Medicine, Ege University, Bornova, Izmir, 35100, Turkey
- Department of Medical Pharmacology, Faculty of Medicine, Lokman Hekim University, Ankara, Türkiye
| | - Begüm Tahhan
- Department of Medical Biochemistry, Faculty of Medicine, Ege University, Bornova, Izmir, 35100, Turkey
| | - Dilek Düzgün
- Department of Biochemistry, Faculty of Science, Ege University, Bornova, Izmir, Turkey
| | - Zeynep Elçim Koru
- Department of Biochemistry, Faculty of Science, Ege University, Bornova, Izmir, Turkey
| | - Ebru Sezer
- Department of Medical Biochemistry, Faculty of Medicine, Ege University, Bornova, Izmir, 35100, Turkey
| | - Derya Aydın
- Department of Pediatric Cardiology, Faculty of Medicine, Ege University Izmir, Bornova, Izmir, Turkey
| | - Resit Erturk Levent
- Department of Pediatric Cardiology, Faculty of Medicine, Ege University Izmir, Bornova, Izmir, Turkey
| | - Sema Kalkan Ucar
- Department of Pediatric Metabolic Disease, Faculty of Medicine, Ege University, Bornova, Izmir, Turkey
| | - Mahmut Coker
- Department of Pediatric Metabolic Disease, Faculty of Medicine, Ege University, Bornova, Izmir, Turkey
| | - Eser Yıldırım Sozmen
- Department of Medical Biochemistry, Faculty of Medicine, Tınaztepe University, Buca, Izmir, Turkey
| |
Collapse
|
5
|
Motamed-Gorji N, Khalil Y, Gonzalez-Robles C, Khan S, Mills P, Garcia-Moreno H, Ging H, Tariq A, Clayton PT, Giunti P. Elevated Bile Acid 3β,5α,6β-Trihydroxycholanoyl Glycine in a Subset of Adult Ataxias Including Niemann-Pick Type C. Antioxidants (Basel) 2024; 13:561. [PMID: 38790666 PMCID: PMC11117656 DOI: 10.3390/antiox13050561] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2024] [Revised: 04/18/2024] [Accepted: 04/22/2024] [Indexed: 05/26/2024] Open
Abstract
Ataxia is a common neurological feature of Niemann-Pick disease type C (NPC). In this disease, unesterified cholesterol accumulates in lysosomes of the central nervous system and hepatic cells. Oxidation by reactive oxygen species produces oxysterols that can be metabolised to specific bile acids. These bile acids have been suggested as useful biomarkers to detect NPC. Concentrations of 3β,5α,6β-trihydroxycholanyl glycine (3β,5α,6β-triOH-Gly) and 3β,7β-dihydroxy-5-cholenyl glycine (3β,7β-diOH-Δ5-Gly) were measured in plasma of 184 adults with idiopathic ataxia. All patients were tested with whole genome sequencing containing hereditary ataxia panels, which include NPC1 and NPC2 mutations and other genetic causes of ataxia. Plasma 3β,5α,6β-triOH-Gly above normal (>90 nM) was found in 8 out of 184 patients. One patient was homozygous for the p.(Val1165Met) mutation in the NPC1 gene. The remaining seven included one patient with Friedreich's ataxia and three patients with autoimmune diseases. Oxidative stress is known to be increased in Friedreich's ataxia and in autoimmune diseases. Therefore, this subset of patients possibly shares a common mechanism that determines the increase of this bile acid. In a large cohort of adults with ataxia, plasma 3β,5α,6β-triOH-Gly was able to detect the one patient in the cohort with NPC1 disease, but also detected oxidation of cholesterol by ROS in other disorders. Plasma 3β,7β-diOH-Δ5-Gly is not a potential biomarker for NPC1.
Collapse
Affiliation(s)
- Nazgol Motamed-Gorji
- Ataxia Centre, Department of Clinical and Movement Neurosciences, UCL Queen Square Institute of Neurology, Queen Square, London WC1N 3BG, UK (H.G.-M.)
| | - Youssef Khalil
- Inborn Errors of Metabolism, Genetics and Genomic Medicine, UCL Great Ormond Street Institute of Child Health, 30 Guilford Street, London WC1N 1EH, UK (P.M.)
| | - Cristina Gonzalez-Robles
- Ataxia Centre, Department of Clinical and Movement Neurosciences, UCL Queen Square Institute of Neurology, Queen Square, London WC1N 3BG, UK (H.G.-M.)
| | - Shamsher Khan
- Ataxia Centre, Department of Clinical and Movement Neurosciences, UCL Queen Square Institute of Neurology, Queen Square, London WC1N 3BG, UK (H.G.-M.)
| | - Philippa Mills
- Inborn Errors of Metabolism, Genetics and Genomic Medicine, UCL Great Ormond Street Institute of Child Health, 30 Guilford Street, London WC1N 1EH, UK (P.M.)
| | - Hector Garcia-Moreno
- Ataxia Centre, Department of Clinical and Movement Neurosciences, UCL Queen Square Institute of Neurology, Queen Square, London WC1N 3BG, UK (H.G.-M.)
| | - Heather Ging
- Ataxia Centre, Department of Clinical and Movement Neurosciences, UCL Queen Square Institute of Neurology, Queen Square, London WC1N 3BG, UK (H.G.-M.)
| | - Ambreen Tariq
- Ataxia Centre, Department of Clinical and Movement Neurosciences, UCL Queen Square Institute of Neurology, Queen Square, London WC1N 3BG, UK (H.G.-M.)
| | - Peter T. Clayton
- Inborn Errors of Metabolism, Genetics and Genomic Medicine, UCL Great Ormond Street Institute of Child Health, 30 Guilford Street, London WC1N 1EH, UK (P.M.)
| | - Paola Giunti
- Ataxia Centre, Department of Clinical and Movement Neurosciences, UCL Queen Square Institute of Neurology, Queen Square, London WC1N 3BG, UK (H.G.-M.)
| |
Collapse
|
6
|
van der Ham M, Gerrits J, Prinsen B, van Hasselt P, Fuchs S, Jans J, Willems A, de Sain-van der Velden M. UPLC-Orbitrap-HRMS application for analysis of plasma sterols. Anal Chim Acta 2024; 1296:342347. [PMID: 38401937 DOI: 10.1016/j.aca.2024.342347] [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: 09/20/2023] [Revised: 01/11/2024] [Accepted: 02/04/2024] [Indexed: 02/26/2024]
Abstract
Correct identification and quantification of different sterol biomarkers can be used as a first-line diagnostic approach for inherited metabolic disorders (IMD). The main drawbacks of current methodologies are related to lack of selectivity and sensitivity for some of these compounds. To address this, we developed and validated two sensitive and selective assays for quantification of six cholesterol biosynthesis pathway intermediates (total amount (free and esterified form) of 7-dehydrocholesterol (7-DHC), 8-dehydrocholesterol (8-DHC), desmosterol, lathosterol, lanosterol and cholestanol), two phytosterols (total amount (free and esterified form) of campesterol and sitosterol) and free form of two oxysterols (7-ketocholesterol (7-KC) and 3β,5α,6β-cholestane-triol (C-triol). For quantification of four cholesterol intermediates we based our analytical approach on sterol derivatization with 4-phenyl-1,2,4-triazoline-3,5-dione (PTAD). Quantification of all analytes is performed using UPLC coupled to an Orbitrap high resolution mass spectrometry (HRMS) system, with detection of target ions through full scan acquisition using positive atmospheric pressure chemical ionization (APCI) mode. UPLC and MS parameters were optimized to achieve high sensitivity and selectivity. Analog stable isotope labeled for each compound was used for proper quantification and correction for recovery, matrix effects and process efficiency. Precision (2.4%-12.3% inter-assay variation), lower limit of quantification (0.027 nM-50.5 nM) and linearity (5.5 μM (R2 0.999) - 72.3 μM (R2 0.997)) for phyto- and oxysterols were determined. The diagnostic potential of these two assays in a cohort of patients (n = 31, 50 samples) diagnosed with IMD affecting cholesterol and lysosomal/peroxisomal homeostasis is demonstrated.
Collapse
Affiliation(s)
- Maria van der Ham
- Section Metabolic Diagnostics, Department of Genetics, University Medical Centre Utrecht, the Netherlands
| | - Johan Gerrits
- Section Metabolic Diagnostics, Department of Genetics, University Medical Centre Utrecht, the Netherlands
| | - Berthil Prinsen
- Section Metabolic Diagnostics, Department of Genetics, University Medical Centre Utrecht, the Netherlands
| | - Peter van Hasselt
- Section of Metabolic Diseases, Wilhelmina Children's Hospital, University Medical Center Utrecht, the Netherlands
| | - Sabine Fuchs
- Section of Metabolic Diseases, Wilhelmina Children's Hospital, University Medical Center Utrecht, the Netherlands
| | - Judith Jans
- Section Metabolic Diagnostics, Department of Genetics, University Medical Centre Utrecht, the Netherlands
| | - Anke Willems
- Section Metabolic Diagnostics, Department of Genetics, University Medical Centre Utrecht, the Netherlands
| | | |
Collapse
|
7
|
Lopergolo D, Bianchi S, Gallus GN, Locci S, Pucci B, Leoni V, Gasparini D, Tardelli E, Chincarini A, Sestini S, Santorelli FM, Zetterberg H, De Stefano N, Mignarri A. Familial Alzheimer's disease associated with heterozygous NPC1 mutation. J Med Genet 2024; 61:332-339. [PMID: 37989569 DOI: 10.1136/jmg-2023-109219] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Accepted: 10/14/2023] [Indexed: 11/23/2023]
Abstract
INTRODUCTION NPC1 mutations are responsible for Niemann-Pick disease type C (NPC), a rare autosomal recessive neurodegenerative disease. Patients harbouring heterozygous NPC1 mutations may rarely show parkinsonism or dementia. Here, we describe for the first time a large family with an apparently autosomal dominant late-onset Alzheimer's disease (AD) harbouring a novel heterozygous NPC1 mutation. METHODS All the five living siblings belonging to the family were evaluated. We performed clinical evaluation, neuropsychological tests, assessment of cerebrospinal fluid markers of amyloid deposition, tau pathology and neurodegeneration (ATN), structural neuroimaging and brain amyloid-positron emission tomography. Oxysterol serum levels were also tested. A wide next-generation sequencing panel of genes associated with neurodegenerative diseases and a whole exome sequencing analysis were performed. RESULTS We detected the novel heterozygous c.3034G>T (p.Gly1012Cys) mutation in NPC1, shared by all the siblings. No other point mutations or deletions in NPC1 or NPC2 were found. In four siblings, a diagnosis of late-onset AD was defined according to clinical characterisation and ATN biomarkers (A+, T+, N+) and serum oxysterol analysis showed increased 7-ketocholesterol and cholestane-3β,5α,6β-triol. DISCUSSION We describe a novel NPC1 heterozygous mutation harboured by different members of a family with autosomal dominant late-onset amnesic AD without NPC-associated features. A missense mutation in homozygous state in the same aminoacidic position has been previously reported in a patient with NPC with severe phenotype. The alteration of serum oxysterols in our family corroborates the pathogenic role of our NPC1 mutation. Our work, illustrating clinical and biochemical disease hallmarks associated with NPC1 heterozygosity in patients affected by AD, provides relevant insights into the pathogenetic mechanisms underlying this possible novel association.
Collapse
Affiliation(s)
- Diego Lopergolo
- Department of Medicine, Surgery and Neurosciences, University of Siena, Siena, Italy
- UOC Neurologia e Malattie Neurometaboliche, Azienda Ospedaliero-Universitaria Senese, Siena, Italy
| | - Silvia Bianchi
- Department of Medicine, Surgery and Neurosciences, University of Siena, Siena, Italy
- UOC Neurologia e Malattie Neurometaboliche, Azienda Ospedaliero-Universitaria Senese, Siena, Italy
| | - Gian Nicola Gallus
- Department of Medicine, Surgery and Neurosciences, University of Siena, Siena, Italy
- UOC Neurologia e Malattie Neurometaboliche, Azienda Ospedaliero-Universitaria Senese, Siena, Italy
| | - Sara Locci
- Department of Medicine, Surgery and Neurosciences, University of Siena, Siena, Italy
- UOC Neurologia e Malattie Neurometaboliche, Azienda Ospedaliero-Universitaria Senese, Siena, Italy
| | - Barbara Pucci
- Department of Medicine, Surgery and Neurosciences, University of Siena, Siena, Italy
- UOC Neurologia e Neurofisiologia Clinica, Azienda Ospedaliero-Universitaria Senese, Siena, Italy
| | - Valerio Leoni
- Laboratory of Clinical Chemistry, Hospital of Desio, ASST Brianza, School of Medicine and Surgery, University of Milano Bicocca, Milan, Italy
| | - Daniele Gasparini
- Department of Medicine, Surgery and Neurosciences, University of Siena, Siena, Italy
- UOC Neurologia e Malattie Neurometaboliche, Azienda Ospedaliero-Universitaria Senese, Siena, Italy
| | - Elisa Tardelli
- Unit of Nuclear Medicine, Department of Diagnostic Imaging, PO - S. Stefano, Azienda U.S.L. Toscana Centro, Prato, italy
| | | | - Stelvio Sestini
- Unit of Nuclear Medicine, Department of Diagnostic Imaging, PO - S. Stefano, Azienda U.S.L. Toscana Centro, Prato, italy
| | - Filippo Maria Santorelli
- Molecular Medicine for Neurodegenerative and Neuromuscular Diseases Unit, IRCCS Stella Maris Foundation, Calambrone, Italy
| | - Henrik Zetterberg
- Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
- UK Dementia Research Institute at UCL, London, UK
- Department of Neurodegenerative Disease, UCL Institute of Neurology, London, UK
- Hong Kong Center for Neurodegenerative Diseases, Hong Kong Special Administrative Region, People's Republic of China
| | - Nicola De Stefano
- Department of Medicine, Surgery and Neurosciences, University of Siena, Siena, Italy
- UOC Neurologia e Malattie Neurometaboliche, Azienda Ospedaliero-Universitaria Senese, Siena, Italy
| | - Andrea Mignarri
- Department of Medicine, Surgery and Neurosciences, University of Siena, Siena, Italy
- UOC Neurologia e Malattie Neurometaboliche, Azienda Ospedaliero-Universitaria Senese, Siena, Italy
| |
Collapse
|
8
|
Jerves Serrano T, Gold J, Cooper JA, Church HJ, Tylee KL, Wu HY, Kim SY, Stepien KM. Hepatomegaly and Splenomegaly: An Approach to the Diagnosis of Lysosomal Storage Diseases. J Clin Med 2024; 13:1465. [PMID: 38592278 PMCID: PMC10932313 DOI: 10.3390/jcm13051465] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2024] [Revised: 02/28/2024] [Accepted: 02/29/2024] [Indexed: 04/10/2024] Open
Abstract
Clinical findings of hepatomegaly and splenomegaly, the abnormal enlargement of the liver and spleen, respectively, should prompt a broad differential diagnosis that includes metabolic, congestive, neoplastic, infectious, toxic, and inflammatory conditions. Among the metabolic diseases, lysosomal storage diseases (LSDs) are a group of rare and ultrarare conditions with a collective incidence of 1 in 5000 live births. LSDs are caused by genetic variants affecting the lysosomal enzymes, transporters, or integral membrane proteins. As a result, abnormal metabolites accumulate in the organelle, leading to dysfunction. Therapeutic advances, including early diagnosis and disease-targeted management, have improved the life expectancy and quality of life of people affected by certain LSDs. To access these new interventions, LSDs must be considered in patients presenting with hepatomegaly and splenomegaly throughout the lifespan. This review article navigates the diagnostic approach for individuals with hepatosplenomegaly particularly focusing on LSDs. We provide hints in the history, physical exam, laboratories, and imaging that may identify LSDs. Additionally, we discuss molecular testing, arguably the preferred confirmatory test (over biopsy), accompanied by enzymatic testing when feasible.
Collapse
Affiliation(s)
| | - Jessica Gold
- Division of Genetics, Department of Pediatrics, Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA;
| | - James A. Cooper
- Willink Biochemical Genetics Laboratory, St Mary’s Hospital, Manchester University NHS Foundation Trust, Manchester M13 9WL, UK; (J.A.C.); (H.J.C.); (K.L.T.); (H.Y.W.)
| | - Heather J. Church
- Willink Biochemical Genetics Laboratory, St Mary’s Hospital, Manchester University NHS Foundation Trust, Manchester M13 9WL, UK; (J.A.C.); (H.J.C.); (K.L.T.); (H.Y.W.)
| | - Karen L. Tylee
- Willink Biochemical Genetics Laboratory, St Mary’s Hospital, Manchester University NHS Foundation Trust, Manchester M13 9WL, UK; (J.A.C.); (H.J.C.); (K.L.T.); (H.Y.W.)
| | - Hoi Yee Wu
- Willink Biochemical Genetics Laboratory, St Mary’s Hospital, Manchester University NHS Foundation Trust, Manchester M13 9WL, UK; (J.A.C.); (H.J.C.); (K.L.T.); (H.Y.W.)
| | - Sun Young Kim
- Division of Human Genetics, Department of Pediatrics, Cincinnati Children’s Hospital Medical Center, University of Cincinnati, Cincinnati, OH 45219, USA;
| | - Karolina M. Stepien
- Salford Royal Organization, Northern Care Alliance NHS Foundation Trust, Adult Inherited Metabolic Diseases Department, Salford M6 8HD, UK
- Division of Cardiovascular Sciences, University of Manchester, Manchester M13 9PL, UK
| |
Collapse
|
9
|
de Médina P, Ayadi S, Soulès R, Payre B, Rup-Jacques S, Silvente-Poirot S, Samadi M, Poirot M. Chemical synthesis and biochemical properties of cholestane-5α,6β-diol-3-sulfonate: A non-hydrolysable analogue of cholestane-5α,6β-diol-3β-sulfate. J Steroid Biochem Mol Biol 2023; 234:106396. [PMID: 37683773 DOI: 10.1016/j.jsbmb.2023.106396] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Revised: 08/22/2023] [Accepted: 09/04/2023] [Indexed: 09/10/2023]
Abstract
Cholestane-3β,5α,6β-triol (CT) is a primary metabolite of 5,6-epoxycholesterols (5,6-EC) that is catalyzed by the cholesterol-5,6-epoxide hydrolase (ChEH). CT is a well-known biomarker for Niemann-Pick disease type C (NP-C), a progressive inherited neurodegenerative disease. On the other hand, CT is known to be metabolized by the 11β-hydroxysteroid-dehydrogenase of type 2 (11β-HSD2) into a tumor promoter named oncosterone that stimulates the growth of breast cancer tumors. Sulfation is a major metabolic transformation leading to the production of sulfated oxysterols. The production of cholestane-5α,6β-diol-3β-O-sulfate (CDS) has been reported in breast cancer cells. However, no data related to CDS biological properties have been reported so far. These studies have been hampered because sulfate esters of sterols and steroids are rapidly hydrolyzed by steroid sulfatase to give free steroids and sterols. In order to get insight into the biological properties of CDS, we report herein the synthesis and the characterization of cholestane-5α,6β-diol-3β-sulfonate (CDSN), a non-hydrolysable analogue of CDS. We show that CDSN is a potent inhibitor of 11β-HSD2 that blocks oncosterone production on cell lysate. The inhibition of oncosterone biosynthesis of a whole cell assay was observed but results from the blockage by CDSN of the uptake of CT in MCF-7 cells. While CDSN inhibits MCF-7 cell proliferation, we found that it potentiates the cytotoxic activity of post-lanosterol cholesterol biosynthesis inhibitors such as tamoxifen and PBPE. This effect was associated with an increase of free sterols accumulation and the appearance of giant multilamellar bodies, a structural feature reminiscent of Type C Niemann-Pick disease cells and consistent with a possible inhibition by CDSN of NPC1. Altogether, our data showed that CDSN is biologically active and that it is a valuable tool to study the biological properties of CDS and more specifically its impact on immunity and viral infection.
Collapse
Affiliation(s)
- Philippe de Médina
- Cancer Research Center of Toulouse (CRCT), Inserm, CNRS, University of Toulouse, Team INOV: Cholesterol Metabolism and Therapeutic Innovations, Toulouse, France; Equipe labellisée par la Ligue Nationale contre le Cancer, France; French network for Nutrition physical Acitivity And Cancer Research (NACRe network), France.
| | - Silia Ayadi
- Cancer Research Center of Toulouse (CRCT), Inserm, CNRS, University of Toulouse, Team INOV: Cholesterol Metabolism and Therapeutic Innovations, Toulouse, France; Equipe labellisée par la Ligue Nationale contre le Cancer, France
| | - Régis Soulès
- Cancer Research Center of Toulouse (CRCT), Inserm, CNRS, University of Toulouse, Team INOV: Cholesterol Metabolism and Therapeutic Innovations, Toulouse, France; Equipe labellisée par la Ligue Nationale contre le Cancer, France; French network for Nutrition physical Acitivity And Cancer Research (NACRe network), France
| | - Bruno Payre
- Centre de Microscopie Electronique Appliquée à la Biologie, Faculté de Médecine Rangueil, Toulouse, France
| | - Sandrine Rup-Jacques
- Laboratory of Chemistry and Physics Multi-Scale Approach to Complex Environments, Department of Chemistry, University Lorraine, 57070 Metz, France
| | - Sandrine Silvente-Poirot
- Cancer Research Center of Toulouse (CRCT), Inserm, CNRS, University of Toulouse, Team INOV: Cholesterol Metabolism and Therapeutic Innovations, Toulouse, France; Equipe labellisée par la Ligue Nationale contre le Cancer, France; French network for Nutrition physical Acitivity And Cancer Research (NACRe network), France.
| | - Mohammad Samadi
- Laboratory of Chemistry and Physics Multi-Scale Approach to Complex Environments, Department of Chemistry, University Lorraine, 57070 Metz, France.
| | - Marc Poirot
- Cancer Research Center of Toulouse (CRCT), Inserm, CNRS, University of Toulouse, Team INOV: Cholesterol Metabolism and Therapeutic Innovations, Toulouse, France; Equipe labellisée par la Ligue Nationale contre le Cancer, France; French network for Nutrition physical Acitivity And Cancer Research (NACRe network), France.
| |
Collapse
|
10
|
Arslan N, Coker M, Gokcay GF, Kiykim E, Onenli Mungan HN, Ezgu F. Expert opinion on patient journey, diagnosis and clinical monitoring in acid sphingomyelinase deficiency in Turkey: a pediatric metabolic disease specialist's perspective. Front Pediatr 2023; 11:1113422. [PMID: 37435168 PMCID: PMC10330960 DOI: 10.3389/fped.2023.1113422] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Accepted: 06/06/2023] [Indexed: 07/13/2023] Open
Abstract
This review by a panel of pediatric metabolic disease specialists aimed to provide a practical and implementable guidance document to assist clinicians in best clinical practice in terms of recognition, diagnosis and management of patients with acid sphingomyelinase deficiency (ASMD). The participating experts consider the clinical suspicion of ASMD by the physician to be of utmost importance in the prevention of diagnostic delay and strongly suggest the use of a diagnostic algorithm including/starting with dried blood spots assay in the timely diagnosis of ASMD in patients presenting with hepatosplenomegaly and a need for increased awareness among physicians in this regard to consider ASMD in the differential diagnosis. In anticipation of the introduction of enzyme replacement therapy, raising awareness of the disease among physicians to prevent diagnostic delay and further investigation addressing natural history of ASMD across the disease spectrum, potential presenting characteristics with a high index of suspicion, as well as biomarkers and genotype-phenotype correlations suggestive of poor prognosis seem important in terms of implementation of best practice patterns.
Collapse
Affiliation(s)
- Nur Arslan
- Division of Pediatric Metabolism, Department of Pediatrics, Dokuz Eylul University Faculty of Medicine, Izmir, Türkiye
| | - Mahmut Coker
- Division of Pediatric Metabolism, Department of Pediatrics, Ege University Faculty of Medicine, Izmir, Türkiye
| | - Gulden Fatma Gokcay
- Division of Pediatric Metabolism, Department of Pediatrics, Istanbul University Istanbul Faculty of Medicine, Istanbul, Türkiye
| | - Ertugrul Kiykim
- Division of Pediatric Metabolism, Department of Pediatrics, Istanbul University Cerrahpasa Faculty of Medicine, Istanbul, Türkiye
| | | | - Fatih Ezgu
- Division of Pediatric Metabolism and Pediatric Genetics, Department of Pediatrics, Gazi University Faculty of Medicine, Ankara, Türkiye
| |
Collapse
|
11
|
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
|
12
|
A Novel Variant in the LIPA Gene Associated with Distinct Phenotype. Balkan J Med Genet 2022; 25:93-100. [PMID: 36880034 PMCID: PMC9985358 DOI: 10.2478/bjmg-2022-0010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/06/2023] Open
Abstract
Deficiency of lysosomal acid lipase (LAL-D) is caused by biallelic pathogenic variants in the LIPA gene. Spectrum of LAL-D ranges from early onset of hepatosplenomegaly and psychomotor regression (Wolman disease) to a more chronic course (cholesteryl ester storage disease - CESD). The diagnosis is based on lipid and biomarker profiles, specific liver histopathology, enzyme deficiency, and identification of causative genetic variants. Biomarker findings are a useful for diagnostics of LAL-D, including high plasma concentration of chitotriosidase as well as elevated oxysterols. Current treatment options include enzyme replacement therapy (sebelipase-alpha), statins, liver transplantation, and stem cell transplantation. We present two pairs of siblings from Serbia with a distinctive phenotype resembling LAL-D with a novel variant of unknown significance (VUS) detected in the LIPA gene and residual LAL activity. All patients presented with hepatosplenomegaly at early childhood. In siblings from family 1, compound heterozygosity for a pathogenic c.419G>A (p.Trp140Ter) variant and a novel VUS c.851C>T (p.Ser284Phe) was detected. Patients from family 2 were homozygous for c.851C>T VUS and both have typical histopathologic findings for LAL-D in the liver. Enzyme activity of LAL was tested in three patients and reported as sufficient, and therefore enzyme replacement therapy could not be approved. When confronted with a challenge of diagnosing an inherited metabolic disorder, several aspects are taken into consideration: clinical manifestations, specific biomarkers, enzyme assay results, and molecular genetic findings. This report brings cases to light which have a considerable discrepancy between those aspects, namely the preserved LAL enzyme activity in presence of clinical manifestations and rare variants in the LIPA gene.
Collapse
|
13
|
Leal AF, Nieto WG, Candelo E, Pachajoa H, Alméciga-Díaz CJ. Hematological Findings in Lysosomal Storage Disorders: A Perspective from the Medical Laboratory. EJIFCC 2022; 33:28-42. [PMID: 35645695 PMCID: PMC9092717] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Abstract
Lysosomal storage disorders (LSDs) are a group of rare and genetic diseases produced by mutations in genes coding for proteins involved in lysosome functioning. Protein defect leads to the lysosomal accumulation of undegraded macromolecules including glycoproteins, glycosaminoglycans, lipids, and glycogen. Depending on the stored substrate, several pathogenic cascades may be activated leading to multisystemic and progressive disorders affecting the brain, eye, ear, lungs, heart, liver, spleen, kidney, skin, or bone. In addition, for some of these disorders, hematological findings have been also reported. In this paper, we review the major hematological alterations in LSDs based on 56 case reports published between 2010 and 2020. Hematological alterations were reported in sphingolipidosis, mucopolysaccharidoses, mucolipidoses, neuronal ceroid lipofuscinosis, glycogenosis, glycoproteinosis, cystinosis, and cholesteryl ester storage disease. They were reported alterations in red cell linage and leukocytes, such as anemia and morphology changes in eosinophils, neutrophils, monocytes, and lymphocytes. In addition, changes in platelet counts (thrombocytopenia) and leukocyte abnormalities on non-peripheral blood samples were also reported for some LSDs. Although in most of the cases these hematological alterations are not pathognomonic of a specific disease or group of LSDs, since they can be easily identified in general clinical laboratories, their identification may contribute to the diagnosis of these disorders. In this sense, we hope that this review contributes to the awareness of the importance of hematological alterations in the diagnosis of LSDs.
Collapse
Affiliation(s)
- Andrés Felipe Leal
- Institute for the Study of Inborn Errors of Metabolism, Faculty of Science, Pontificia Universidad Javeriana, Bogotá D.C., Colombia,Co-corresponding authors: Andrés Felipe Leal, B.Sc., M.Sc. Carlos J. Alméciga-Díaz, BPharm, Ph.D. Institute for the Study of Inborn Errors of Metabolism, Faculty of Science Pontificia Universidad Javeriana, Cra. 7 No. 43-82 Building 54, Room 305A Bogotá D.C., 110231 Colombia Tel: +57-1 3208320 Ext 4140 Fax: +57-1 3208320 Ext 4099 E-mail: E-mail:
| | - Wendy G. Nieto
- Translational Biomedical Research Group, Centro de Investigaciones, Fundación Cardiovascular de Colombia, Santander, Colombia
| | - Estephania Candelo
- Department of Medical Basic Sciences, Faculty of Health Sciences, Universidad Icesi, Cali, Colombia,Centro de Investigaciones Clínicas, Fundación Valle del Lili, Cali, Colombia,Congenital Abnormalities and Rare Disease Centre (CIACER), Cali, Colombia
| | - Harry Pachajoa
- Department of Medical Basic Sciences, Faculty of Health Sciences, Universidad Icesi, Cali, Colombia, Department of Medical Genetics, Fundación Valle del Lili, Cali, Colombia,Congenital Abnormalities and Rare Disease Centre (CIACER), Cali, Colombia
| | - Carlos Javier Alméciga-Díaz
- Institute for the Study of Inborn Errors of Metabolism, Faculty of Science, Pontificia Universidad Javeriana, Bogotá D.C., Colombia
| |
Collapse
|
14
|
Risso D, Leoni V, Canzoneri F, Arveda M, Zivoli R, Peraino A, Poli G, Menta R. Presence of cholesterol oxides in milk chocolates and their correlation with milk powder freshness. PLoS One 2022; 17:e0264288. [PMID: 35312699 PMCID: PMC8936476 DOI: 10.1371/journal.pone.0264288] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2021] [Accepted: 02/04/2022] [Indexed: 12/18/2022] Open
Abstract
Cholesterol oxidation products (COPs) of non-enzymatic origin are mainly found in meat, fish, eggs and milk, mostly originating from the type of feeding, processing and storage. To verify the significance of COPs as biomarkers of cholesterol autoxidation and milk freshness, we quantified them in chocolates containing whole milk powders (WMPs) of increasing shelf-lives (i.e. 20, 120, and 180 days). Non-enzymatic total COPs (both free and esterified) ranged from 256.57 ± 11.97 to 445.82 ± 11.88 ng/g, increasing proportionally to the shelf-life of the WMPs, thus reflecting the ingredients’ freshness. Based on the expected theoretical COPs, the effect of processing was quantitatively less significant in the generation of oxysterols (41–44%) than the contribution of the autoxidation of the WMPs over time (56–59%), pointing to the shelf-life as the primary determinant of COPs. Lastly, we quantified COPs of major commercial milk chocolates on the Italian market, which followed a similar distribution (from 240.79 ± 11.74 to 475.12 ± 12.58 ng/g). Although further replications of this work are needed, this study reports preliminary results and a practical example of a first application of non-enzymatic COPs as markers to further quantify and characterize the nutritional quality and freshness, not only of ingredients but also of composite products.
Collapse
Affiliation(s)
- Davide Risso
- Soremartec Italia Srl, Ferrero Group, Alba, Italy
| | - Valerio Leoni
- Laboratory of Clinical Chemistry, Hospital of Desio and Monza, ASST-Monza, School of Medicine and Surgery, University of Milano Bicocca, Milan, Italy
| | | | | | | | | | - Giuseppe Poli
- Department of Clinical and Biological Sciences, University of Torino, San Luigi Hospital, Turin, Italy
| | | |
Collapse
|
15
|
Diagnosis, treatment, and follow-up of a case of Wolman disease with hemophagocytic lymphohistiocytosis. Mol Genet Metab Rep 2022; 30:100833. [PMID: 35242567 PMCID: PMC8856920 DOI: 10.1016/j.ymgmr.2021.100833] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2021] [Revised: 12/06/2021] [Accepted: 12/07/2021] [Indexed: 12/14/2022] Open
Abstract
Wolman Disease (WD) is a severe multi-system metabolic disease due to lysosomal acid lipase (LAL) deficiency. We report on a WD infant who developed an unusual hemophagocytic lymphohistiocytosis (HLH) phenotype related to WD treated with sebelipase alfa. A male baby came to our attention at six months of life for respiratory insufficiency and sepsis, abdominal distension, severe hepatosplenomegaly, diarrhea, and severe growth retardation. HLH was diagnosed and treated with intravenous immunoglobulin, steroids, cyclosporine, broad-spectrum antimicrobial therapy, and finally with the anti-IL-6 drug tocilizumab. WD was suspected for the presence of adrenal calcifications and it was confirmed by LAL enzyme activity and by molecular analysis of LIPA. Plasma oxysterols cholestan-3β,5α,6β-triol (C-triol), and 7-ketocholesterol (7-KC) were markedly increased. Sebelipase alfa was started with progressive amelioration of biochemical and clinical features. The child died from sepsis, 2 months after sebelipase discontinuation requested by parents. Our case shows the importance of an early diagnosis of WD and confirms the difficulty to reach a diagnosis in the HLH phenotype. Sebelipase alpha is an effective treatment for LAL deficiency, also in children affected by WD. Further data are necessary to confirm the utility of measuring plasma c-triol as a biochemical marker of the disease.
Collapse
|
16
|
Jiang X, Ory DS. Advancing Diagnosis and Treatment of Niemann-Pick C disease through Biomarker Discovery. EXPLORATION OF NEUROPROTECTIVE THERAPY 2021; 1:146-158. [PMID: 35356760 PMCID: PMC8963791 DOI: 10.37349/ent.2021.00012] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/25/2021] [Accepted: 11/05/2021] [Indexed: 05/30/2023]
Abstract
Niemann-Pick C is a rare neurodegenerative, lysosomal storage disease caused by accumulation of unesterified cholesterol. Diagnosis of the disease is often delayed due to its rarity, the heterogeneous presentation and the early non-specific symptoms. The discovery of disease-specific biomarkers - cholestane-3β,5α,6β-triol (C-triol), trihydroxycholanic acid glycinate (TCG) and N-palmitoyl-O-phosphocholineserine (PPCS, initially referred to as lysoSM-509) - has led to development of non-invasive, blood-based diagnostics. Dissemination of these rapid, sensitive, and specific clinical assays has accelerated diagnosis. Moreover, the superior receiver operating characteristic of the TCG bile acid biomarker and its detection in dried blood spots has also facilitated development of a newborn screen for NPC, which is currently being piloted in New York state. The C-triol, TCG and PPCS biomarkers have also proven useful for monitoring treatment response in peripheral tissues, but are uninformative with respect to treatment efficacy in the central nervous system (CNS). A major gap for the field is the lack of a validated, non-invasive biomarker to monitor the course of disease and CNS response to therapy.
Collapse
Affiliation(s)
- Xuntian Jiang
- Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110
| | | |
Collapse
|
17
|
Rojas MM, Villalpando DM, Alexander-Aguilera A, Ferrer M, García HS. Effect of CLA supplementation on factors related to vascular dysfunction in arteries of orchidectomized rats. Prostaglandins Other Lipid Mediat 2021; 157:106586. [PMID: 34438054 DOI: 10.1016/j.prostaglandins.2021.106586] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2021] [Revised: 08/17/2021] [Accepted: 08/19/2021] [Indexed: 01/22/2023]
Abstract
The vascular endothelium is a monolayer of flat epithelial cells located between the circulating blood and the underlying connective tissue. It conveys key functions that when impaired, lead to endothelial dysfunction. This condition is responsible for the pathogenesis of vascular diseases. The cardioprotective effect of sex hormones is widely known; hence, a murine orchidectomized model has been employed to study the effects caused by their deficiency. In the search for approaches to maintain vascular health, the effect of dietary fatty acids as CLA on cardiovascular diseases has been studied. Some proven beneficial properties of CLA are antioxidant, antiatherogenic and anti-inflammatory. Our objective was to evaluate the effect of a diet supplemented with 1.8 % (w/w) of CLA, administered during eight weeks, on the amount of cholesterol oxidation products (COPs) produced by orchidectomy and on factors related to vascular dysfunction in the aorta and the mesenteric arteries. The diet with CLA prevented the increase in prostanoids formation and maintained the normal physiological conditions of NO and antioxidant activity. In addition, it prevented the increase in cholesterol and COPs at the vascular wall. CLA-supplemented diet prevented the orchidectomy-induced alterations on prostanoids, NO and COPs and also improved the antioxidant activity. These findings could contribute to understand the mechanisms of actions of CLA involved in the prevention of cardiovascular diseases.
Collapse
Affiliation(s)
- Mibsam M Rojas
- Food Research and Development Unit, National Technology of Mexico/Technological Institute of Veracruz, Ver., Mexico
| | - Diva M Villalpando
- Physiology Department, Faculty of Medicine, Autonomous University of Madrid, Madrid, Spain
| | | | - Mercedes Ferrer
- Physiology Department, Faculty of Medicine, Autonomous University of Madrid, Madrid, Spain.
| | - Hugo S García
- Food Research and Development Unit, National Technology of Mexico/Technological Institute of Veracruz, Ver., Mexico.
| |
Collapse
|
18
|
Guidara W, Messedi M, Maalej M, Naifar M, Khrouf W, Grayaa S, Maalej M, Bonnefont-Rousselot D, Lamari F, Ayadi F. Plasma oxysterols: Altered level of plasma 24-hydroxycholesterol in patients with bipolar disorder. J Steroid Biochem Mol Biol 2021; 211:105902. [PMID: 33901658 DOI: 10.1016/j.jsbmb.2021.105902] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/02/2020] [Revised: 04/09/2021] [Accepted: 04/20/2021] [Indexed: 12/13/2022]
Abstract
Cholesterol and its oxygenated metabolites, including oxysterols, are intensively investigated as potential players in the pathophysiology of brain disorders. Altered oxysterol levels have been described in patients with numerous neuropsychiatric disorders. Recent studies have shown that Bipolar disorder (BD) is associated with the disruption of cholesterol metabolism. The present study was aimed at investigating the profile of oxysterols in plasma, their ratio to total cholesterol and their association with clinical parameters in patients with BD. Thirty three men diagnosed with BD and forty healthy controls matched for age and sex were included in the study. Oxysterol levels were measured by isotope-dilution ultra-performance liquid chromatography-tandem mass spectrometry. Significantly higher levels were observed for cholestane-3β,5α,6β-triol, 27-hydroxycholesterol (27-OHC) and Cholestanol in patients with BD. The concentration of 24-hydroxycholesterol (24-OHC) was significantly lower in patients compared to controls. 24-OHC was also negatively correlated to MAS subscale score (r =-0.343; p = 0.049). In patients, 24-OHC was inversely correlated with age (r = -0.240; p = 0.045). Multivariate analysis found that BD acute decompensation was independently related to the rise in plasma 24-OHC (p = 0.002; OR = 0.966, 95 % CI [0.945 - 0.987]). However, the 24-OHC assay relevance as a biomarker of this disease deserves further investigation in other studies.
Collapse
Affiliation(s)
- Wassim Guidara
- Laboratory of Research "Molecular Basis of Human Diseases", LR19ES13, Faculty of Medicine, University of Sfax, Sfax, Tunisia.
| | - Meriam Messedi
- Laboratory of Research "Molecular Basis of Human Diseases", LR19ES13, Faculty of Medicine, University of Sfax, Sfax, Tunisia
| | - Manel Maalej
- Psychiatry C-department, University of Sfax & Hédi Chaker Hospital, Sfax, Tunisia
| | - Manel Naifar
- Laboratory of Research "Molecular Basis of Human Diseases", LR19ES13, Faculty of Medicine, University of Sfax, Sfax, Tunisia; Laboratory of Biochemistry, University of Sfax & Habib Bourguiba Hospital, Sfax, Tunisia
| | - Walid Khrouf
- AP-HP, Sorbonne University, La Pitié-Salpêtrière University Hospital, Department of Metabolic Biochemistry, Paris, France
| | - Sahar Grayaa
- Laboratory of Research "Molecular Basis of Human Diseases", LR19ES13, Faculty of Medicine, University of Sfax, Sfax, Tunisia
| | - Mohamed Maalej
- Psychiatry C-department, University of Sfax & Hédi Chaker Hospital, Sfax, Tunisia
| | - Dominique Bonnefont-Rousselot
- AP-HP, Sorbonne University, La Pitié-Salpêtrière University Hospital, Department of Metabolic Biochemistry, Paris, France; UTCBS, U1267 Inserm, UMR 8258 CNRS, Université de Paris, Paris, France
| | - Foudil Lamari
- AP-HP, Sorbonne University, La Pitié-Salpêtrière University Hospital, Department of Metabolic Biochemistry, Paris, France
| | - Fatma Ayadi
- Laboratory of Research "Molecular Basis of Human Diseases", LR19ES13, Faculty of Medicine, University of Sfax, Sfax, Tunisia; Laboratory of Biochemistry, University of Sfax & Habib Bourguiba Hospital, Sfax, Tunisia
| |
Collapse
|
19
|
Reynolds M, Linneman LA, Luna S, Warner BB, Turmelle YP, Kulkarni SS, Jiang X, Khanna G, Shinawi M, Porter FD, Ory DS, Cole FS, Dickson PI. A phase 1/2 open label nonrandomized clinical trial of intravenous 2-hydroxypropyl-β-cyclodextrin for acute liver disease in infants with Niemann-Pick C1. Mol Genet Metab Rep 2021; 28:100772. [PMID: 34113546 PMCID: PMC8170172 DOI: 10.1016/j.ymgmr.2021.100772] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2021] [Accepted: 05/14/2021] [Indexed: 11/27/2022] Open
Abstract
Introduction Niemann-Pick C (NPC) is an autosomal recessive disease due to defective NPC1 or NPC2 proteins resulting in endo-lysosomal storage of unesterified cholesterol in the central nervous system and liver. Acute liver disease in the newborn period may be self-limited or fatal. 2-hydroxypropyl-β-cyclodextrin (2HPBCD) is a cholesterol-binding agent that reduces lysosomal cholesterol storage. We have enrolled 3 infants 0–6 months old with direct hyperbilirubinemia due to NPC1 or NPC2 liver disease in a Phase I/II open label clinical trial of intravenous 2HPBCD. Methods Infants received intravenous 2HPBCD twice a week for 6 weeks, followed by monthly infusion for 6-months. Primary outcome measure was reduction of plasma (3β,5α,6β-trihydroxy-cholan-24-oyl) glycine (TCG), a bile acid generated from cholesterol sequestered in lysosome. Results Three participants completed this protocol. A fourth patient received intravenous 2HPBCD under an emergency investigational new drug study but later expired from her underlying condition. The three protocol patients are living and have improved liver enzymes and TCG. No patient has experienced a drug-related adverse event. Conclusion Intravenous 2HPBCD was tolerated in three infants with liver disease due to NPC.
Collapse
Affiliation(s)
- Margaret Reynolds
- Washington University in St. Louis School of Medicine, St. Louis, MO, United States of America
| | - Laura A Linneman
- Washington University in St. Louis School of Medicine, St. Louis, MO, United States of America
| | - Sofia Luna
- Washington University in St. Louis School of Medicine, St. Louis, MO, United States of America
| | - Barbara B Warner
- Washington University in St. Louis School of Medicine, St. Louis, MO, United States of America
| | - Yumirle P Turmelle
- Washington University in St. Louis School of Medicine, St. Louis, MO, United States of America
| | - Sakil S Kulkarni
- Washington University in St. Louis School of Medicine, St. Louis, MO, United States of America
| | - Xuntian Jiang
- Washington University in St. Louis School of Medicine, St. Louis, MO, United States of America
| | - Geetika Khanna
- Washington University in St. Louis School of Medicine, St. Louis, MO, United States of America
| | - Marwan Shinawi
- Washington University in St. Louis School of Medicine, St. Louis, MO, United States of America
| | - Forbes D Porter
- Eunice Kennedy Shriver National Institute of Child Health and Human Development, NIH, DHHS, Bethesda, MD, United States of America
| | - Daniel S Ory
- Washington University in St. Louis School of Medicine, St. Louis, MO, United States of America
| | - F Sessions Cole
- Washington University in St. Louis School of Medicine, St. Louis, MO, United States of America
| | - Patricia I Dickson
- Washington University in St. Louis School of Medicine, St. Louis, MO, United States of America
| |
Collapse
|
20
|
Potter JE, Petts G, Ghosh A, White FJ, Kinsella JL, Hughes S, Roberts J, Hodgkinson A, Brammeier K, Church H, Merrigan C, Hughes J, Evans P, Campbell H, Bonney D, Newman WG, Bigger BW, Broomfield A, Jones SA, Wynn RF. Enzyme replacement therapy and hematopoietic stem cell transplant: a new paradigm of treatment in Wolman disease. Orphanet J Rare Dis 2021; 16:235. [PMID: 34020687 PMCID: PMC8139039 DOI: 10.1186/s13023-021-01849-7] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2021] [Accepted: 05/04/2021] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Wolman disease is a rare, lysosomal storage disorder in which biallelic variants in the LIPA gene result in reduced or complete lack of lysosomal acid lipase. The accumulation of the substrates; cholesterol esters and triglycerides, significantly impacts cellular function. Untreated patients die within the first 12 months of life. Clinically, patients present severely malnourished, with diarrhoea and hepatosplenomegaly, many have an inflammatory phenotype, including with hemophagocytic lymphohistiocytosis (HLH). Hematopoietic stem cell transplant (HCT) had been historically the only treatment available but has a high procedure-related mortality because of disease progression and disease-associated morbidities. More recently, enzyme replacement therapy (ERT) with dietary substrate reduction (DSR) has significantly improved patient survival. However, ERT is life long, expensive and its utility is limited by anti-drug antibodies (ADA) and the need for central venous access. RESULTS We describe five Wolman disease patients diagnosed in infancy that were treated at Royal Manchester Children's Hospital receiving ERT with DSR then HCT-multimodal therapy. In 3/5 an initial response to ERT was attenuated by ADA with associated clinical and laboratory features of deterioration. 1/5 developed anaphylaxis to ERT and the other patient died post HCT with ongoing HLH. All patients received allogeneic HCT. 4/5 patients are alive, and both disease phenotype and laboratory parameters are improved compared to when they were on ERT alone. The gastrointestinal symptoms are particularly improved after HCT, with reduced diarrhoea and vomiting. This allows gradual structured normalisation of diet with improved tolerance of dietary fat. Histologically there are reduced cholesterol clefts, fewer foamy macrophages and an improved villous structure. Disease biomarkers also show improvement with ERT, immunotherapy and HCT. Three patients have mixed chimerism after HCT, indicating a likely engraftment-defect in this condition. CONCLUSION We describe combined ERT, DSR and HCT, multimodal treatment for Wolman disease. ERT and DSR stabilises the sick infant and reduces the formerly described prohibitively high, transplant-associated mortality in this condition. HCT abrogates the problems of ERT, namely attenuating ADA, the need for continuing venous access, and continuing high cost drug treatment. HCT also brings improved efficacy, particularly evident in improved gastrointestinal function and histology. Multimodal therapy should be considered a new paradigm of treatment for Wolman disease patients where there is an attenuated response to ERT, and for all patients where there is a well-matched transplant donor, in order to improve long term gut function, tolerance of a normal diet and quality of life.
Collapse
Affiliation(s)
- Jane E Potter
- Department of Blood and Marrow Transplantation, Royal Manchester Children's Hospital, Oxford Road, Manchester, UK.
| | - Gemma Petts
- Department of Paediatric Histopathology, Royal Manchester Children's Hospital, Oxford Road, Manchester, UK
| | - Arunabha Ghosh
- Manchester Centre for Genomic Medicine, Manchester University NHS Foundation Trust, Oxford Road, Manchester, UK
| | - Fiona J White
- Manchester Centre for Genomic Medicine, Manchester University NHS Foundation Trust, Oxford Road, Manchester, UK
- Department of Therapy and Dietetics, Manchester University NHS Foundation Trust, Oxford Road, Manchester, UK
| | - Jane L Kinsella
- Department of Blood and Marrow Transplantation, Royal Manchester Children's Hospital, Oxford Road, Manchester, UK
| | - Stephen Hughes
- Paediatric Allergy and Immunology Department, Royal Manchester Children's Hospital, Oxford Road, Manchester, UK
| | - Jane Roberts
- Manchester Centre for Genomic Medicine, Manchester University NHS Foundation Trust, Oxford Road, Manchester, UK
| | - Adam Hodgkinson
- Department of Paediatric Histopathology, Royal Manchester Children's Hospital, Oxford Road, Manchester, UK
| | - Kathryn Brammeier
- Manchester Centre for Genomic Medicine, Manchester University NHS Foundation Trust, Oxford Road, Manchester, UK
| | - Heather Church
- Manchester Centre for Genomic Medicine, Manchester University NHS Foundation Trust, Oxford Road, Manchester, UK
| | - Christine Merrigan
- National Centre for Inherited Metabolic Disorders, Children's Health Ireland at Temple Street, Dublin, Ireland
| | - Joanne Hughes
- National Centre for Inherited Metabolic Disorders, Children's Health Ireland at Temple Street, Dublin, Ireland
| | - Pamela Evans
- Department of Haematology and Oncology, Children's Health Ireland at Crumlin, Dublin, Ireland
| | - Helen Campbell
- Department of Blood and Marrow Transplantation, Royal Manchester Children's Hospital, Oxford Road, Manchester, UK
| | - Denise Bonney
- Department of Blood and Marrow Transplantation, Royal Manchester Children's Hospital, Oxford Road, Manchester, UK
| | - William G Newman
- Manchester Centre for Genomic Medicine, Manchester University NHS Foundation Trust, Oxford Road, Manchester, UK
- Evolution and Genomic Science, School of Biological Sciences, University of Manchester, Manchester, UK
| | - Brian W Bigger
- Stem Cell and Neurotherapies Laboratory, Division of Cell Matrix Biology and Regenerative Medicine, University of Manchester, Manchester, UK
| | - Alexander Broomfield
- Manchester Centre for Genomic Medicine, Manchester University NHS Foundation Trust, Oxford Road, Manchester, UK
| | - Simon A Jones
- Manchester Centre for Genomic Medicine, Manchester University NHS Foundation Trust, Oxford Road, Manchester, UK
| | - Robert F Wynn
- Department of Blood and Marrow Transplantation, Royal Manchester Children's Hospital, Oxford Road, Manchester, UK
| |
Collapse
|
21
|
Kilvington A, Barnaba C, Rajasekaran S, Laurens Leimanis ML, Medina-Meza IG. Lipid profiling and dietary assessment of infant formulas reveal high intakes of major cholesterol oxidative product (7-ketocholesterol). Food Chem 2021; 354:129529. [PMID: 33761334 DOI: 10.1016/j.foodchem.2021.129529] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2020] [Revised: 02/03/2021] [Accepted: 03/02/2021] [Indexed: 12/17/2022]
Abstract
Approximately two-thirds of US infants receive infant formula (IF) as a primary or sole nutritional source during the first six months of life. IF is available in a variety of commercial presentations; from a manufacturing standpoint, they can be categorized as powder- (PIF) or liquid- (LIF) based formulations. Thirty commercial IFs were analyzed in their oxidative and non-oxidative lipid profiles. We identified 7-ketocholesterol - a major end-product of cholesterol oxidation - as a potential biomarker of IF manufacturing. The statistical analysis allowed a re-classification of IF based on their metabolomic fingerprint, resulting in three groups assigned with low-to-high oxidative status. Finally, we modeled the dietary intake of cholesterol, sterols, and 7-ketocholesterol in the first year of life. The database provided in this study will be instrumental for scientists interested in infant nutrition, to establish bases for epidemiological studies aimed to find connections between nutrition and diet-associated diseases, such as sitosterolemia.
Collapse
Affiliation(s)
- Alice Kilvington
- Department of Chemical Engineering and Materials Science, Michigan State University, East Lansing, MI, USA.
| | - Carlo Barnaba
- Institute of Quantitative Health Science and Engineering, Michigan State University, East Lansing, MI, USA.
| | - Surender Rajasekaran
- Pediatric Intensive Care Unit, Helen DeVos Children's Hospital, 100 Michigan Street NE, Grand Rapids, MI, USA; Department of Pediatric and Human Development, College of Human Medicine, Michigan State University, East Lansing, MI, USA.
| | - Mara L Laurens Leimanis
- Pediatric Intensive Care Unit, Helen DeVos Children's Hospital, 100 Michigan Street NE, Grand Rapids, MI, USA; Department of Pediatric and Human Development, College of Human Medicine, Michigan State University, East Lansing, MI, USA.
| | - Ilce G Medina-Meza
- Department of Chemical Engineering and Materials Science, Michigan State University, East Lansing, MI, USA; Department of Biosystems and Agricultural Engineering, Michigan State University, East Lansing, MI, USA.
| |
Collapse
|
22
|
McComb M, Browne RW, Bhattacharya S, Bodziak ML, Jakimovski D, Weinstock-Guttman B, Kuhle J, Zivadinov R, Ramanathan M. The cholesterol autoxidation products, 7-ketocholesterol and 7β-hydroxycholesterol are associated with serum neurofilaments in multiple sclerosis. Mult Scler Relat Disord 2021; 50:102864. [PMID: 33677412 DOI: 10.1016/j.msard.2021.102864] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2020] [Revised: 02/17/2021] [Accepted: 02/22/2021] [Indexed: 12/22/2022]
Abstract
BACKGROUND Serum neurofilament light chain (sNfL) is an established marker of neuroaxonal injury in multiple sclerosis (MS). OBJECTIVES To investigate if oxysterols produced from non-enzymatic and enzymatic cholesterol oxidation are differentially associated with sNfL measurements in MS. METHODS This longitudinal study included 62 relapsing-remitting (RR-MS) and 36 progressive MS (PMS) patients with baseline and 5-year follow-up measures of serum levels of 6 oxysterols, sNfL and lipids. The oxysterols, 24-hydroxycholesterol (24HC), 25HC, 27HC, 7αHC, 7βHC and 7-ketocholesterol (7KC), were measured using liquid chromatography-mass spectrometry. sNfL was measured using single molecular array assay. Serum high-density lipoprotein cholesterol (HDL-C) and low-density lipoprotein cholesterol (LDL-C) levels were obtained from a lipid profile. RESULTS The enzymatically produced oxysterols 24HC, 25HC, 27HC and 7αHC were not associated with sNfL. However, baseline levels of reactive oxygen species (ROS) produced oxysterols, 7KC (p = 0.032) and 7βHC (p = 0.0025), were positively associated with sNfL levels at follow-up. Follow-up 7KC (p = 0.038) levels were also associated with follow-up sNfL levels. The associations of 7KC or 7βHC with sNfL remained significant after adjusting for LDL-C or HDL-C. CONCLUSIONS 7KC and 7βHC, produced by ROS-mediated cholesterol oxidation are associated with neuroaxonal injury as assessed by sNfL in MS.
Collapse
Affiliation(s)
- Mason McComb
- Department of Pharmaceutical Sciences, State University of New York, Buffalo, NY, United States
| | - Richard W Browne
- Department of Biotechnical and Clinical Laboratory Sciences, State University of New York, Buffalo, NY, United States
| | - Sonia Bhattacharya
- Department of Biotechnical and Clinical Laboratory Sciences, State University of New York, Buffalo, NY, United States
| | - Mary Lou Bodziak
- Department of Biotechnical and Clinical Laboratory Sciences, State University of New York, Buffalo, NY, United States
| | - Dejan Jakimovski
- Buffalo Neuroimaging Analysis Center, Department of Neurology, University at Buffalo, State University of New York, Buffalo, NY, United States
| | - Bianca Weinstock-Guttman
- Jacobs Comprehensive MS Treatment and Research Center, Department of Neurology, University at Buffalo, State University of New York, Buffalo, NY, United States
| | - Jens Kuhle
- Neurologic Clinic and Policlinic, Departments of Medicine, Biomedicine and Clinical Research, University Hospital Basel, University of Basel, Basel, Switzerland
| | - Robert Zivadinov
- Jacobs Comprehensive MS Treatment and Research Center, Department of Neurology, University at Buffalo, State University of New York, Buffalo, NY, United States; Center for Biomedical Imaging at Clinical Translational Science Institute, University at Buffalo, State University of New York, Buffalo, NY, United States
| | - Murali Ramanathan
- Department of Pharmaceutical Sciences, State University of New York, Buffalo, NY, United States; Jacobs Comprehensive MS Treatment and Research Center, Department of Neurology, University at Buffalo, State University of New York, Buffalo, NY, United States.
| |
Collapse
|
23
|
ELMostafi H, Bahbiti Y, Elhessni A, Bousalham R, Doumar H, Ouichou A, Benmhammed H, Touil T, Mesfioui A. Neuroprotective potential of Argan oil in neuropsychiatric disorders in rats: A review. J Funct Foods 2020. [DOI: 10.1016/j.jff.2020.104233] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
|
24
|
Sidhu R, Kell P, Dietzen DJ, Farhat NY, Do AND, Porter FD, Berry-Kravis E, Reunert J, Marquardt T, Giugliani R, Lourenço CM, Wang RY, Movsesyan N, Plummer E, Schaffer JE, Ory DS, Jiang X. Application of a glycinated bile acid biomarker for diagnosis and assessment of response to treatment in Niemann-pick disease type C1. Mol Genet Metab 2020; 131:405-417. [PMID: 33257258 PMCID: PMC8139135 DOI: 10.1016/j.ymgme.2020.11.005] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/10/2020] [Revised: 11/12/2020] [Accepted: 11/12/2020] [Indexed: 01/05/2023]
Abstract
Niemann-Pick disease type C (NPC) is a neurodegenerative disease in which mutation of NPC1 or NPC2 gene leads to lysosomal accumulation of unesterified cholesterol and sphingolipids. Diagnosis of NPC disease is challenging due to non-specific early symptoms. Biomarker and genetic tests are used as first-line diagnostic tests for NPC. In this study, we developed a plasma test based on N-(3β,5α,6β-trihydroxy-cholan-24-oyl)glycine (TCG) that was markedly increased in the plasma of human NPC1 subjects. The test showed sensitivity of 0.9945 and specificity of 0.9982 to differentiate individuals with NPC1 from NPC1 carriers and controls. Compared to other commonly used biomarkers, cholestane-3β,5α,6β-triol (C-triol) and N-palmitoyl-O-phosphocholine (PPCS, also referred to as lysoSM-509), TCG was equally sensitive for identifying NPC1 but more specific. Unlike C-triol and PPCS, TCG showed excellent stability and no spurious generation of marker in the sample preparation or aging of samples. TCG was also elevated in lysosomal acid lipase deficiency (LALD) and acid sphingomyelinase deficiency (ASMD). Plasma TCG was significantly reduced after intravenous (IV) 2-hydroxypropyl-β-cyclodextrin (HPβCD) treatment. These results demonstrate that plasma TCG was superior to C-triol and PPCS as NPC1 diagnostic biomarker and was able to evaluate the peripheral treatment efficacy of IV HPβCD treatment.
Collapse
Affiliation(s)
- Rohini Sidhu
- Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Pamela Kell
- Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Dennis J Dietzen
- Department of Pathology & Immunology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Nicole Y Farhat
- Section on Molecular Dysmorphology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, NIH, DHHS, Bethesda, MD 20892, USA
| | - An Ngoc Dang Do
- Section on Molecular Dysmorphology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, NIH, DHHS, Bethesda, MD 20892, USA
| | - Forbes D Porter
- Section on Molecular Dysmorphology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, NIH, DHHS, Bethesda, MD 20892, USA
| | | | - Janine Reunert
- Klinik und Poliklinik für Kinder- und Jugendmedizin - Allgemeine Pädiatrie, Universitätsklinikum Münster, Albert-Schweitzer-Campus 1, Gebäude A1, 48149 Münster, Germany
| | - Thorsten Marquardt
- Klinik und Poliklinik für Kinder- und Jugendmedizin - Allgemeine Pädiatrie, Universitätsklinikum Münster, Albert-Schweitzer-Campus 1, Gebäude A1, 48149 Münster, Germany
| | - Roberto Giugliani
- Department of Genetics, UFRGS, Medical Genetics Service, HCPA, BioDiscovery Laboratory, HCPA, Hospital de Clínicas de Porto Alegre, National Institute of Population Medical Genetics - INAGEMP, Porto Alegre, RS 90035-903, Brazil
| | - Charles M Lourenço
- Faculdade de Medicina - Centro Universitario Estácio de Ribeirão Preto, Rua Abrahão Issa Halach, 980 - Ribeirânia, Ribeirão Preto, - SP, Brazil
| | - Raymond Y Wang
- Division of Metabolic Disorders, CHOC Children's Specialists, Orange, CA 92868, USA; Department of Pediatrics, University of California-Irvine School of Medicine, Orange, CA 92868, USA
| | - Nina Movsesyan
- Research Institute, CHOC Children's Hospital, Orange, CA 92868, USA
| | - Ellen Plummer
- Asante Pediatric Hematology and Oncology, Medford, OR, 97504, USA
| | - Jean E Schaffer
- Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Daniel S Ory
- Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Xuntian Jiang
- Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA.
| |
Collapse
|
25
|
Wu C, Iwamoto T, Hossain MA, Akiyama K, Igarashi J, Miyajima T, Eto Y. A combination of 7-ketocholesterol, lysosphingomyelin and bile acid-408 to diagnose Niemann-Pick disease type C using LC-MS/MS. PLoS One 2020; 15:e0238624. [PMID: 32898135 PMCID: PMC7478639 DOI: 10.1371/journal.pone.0238624] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2019] [Accepted: 08/20/2020] [Indexed: 11/18/2022] Open
Abstract
BACKGROUND Niemann-Pick disease type C (NPC) is an autosomal recessive disorder caused by mutations of NPC1 or NPC2, which encode the proteins that are responsible for intracellular cholesterol trafficking. Loss of this function results in the accumulation of cholesterol-related products, such as oxysterols, sphingolipids, and NPC-related bile acids, which were recently used as biochemical biomarkers for the diagnosis of NPC. Bile acid-408 is a new significant compound we found in Japanese NPC patients, and it likely belongs to the category of bile acids. However, the diagnosis of NPC using a single biomarker is not satisfactory for clinical application because of the high instance of false negatives or positives. Therefore, we proposed an application of NPC diagnosis using a combination of 7-ketocholesterol (7-KC), lysosphingomyelin (lysoSM), bile acid-408 and/or glucosylsphingosine (lysoGL-1). METHODS AND FINDINGS 7-KC, lysoSM and lysoGL-1 in sera and bile acid-408 in dried blood spots (DBS) were quantified within 17 minutes using tandem mass spectrometry and high-resolution mass spectrometry, respectively. We measured these biomarkers in NPC patients (n = 19), X-linked adrenoleukodystrophy (X-ALD) patients (n = 5), patients with other lysosomal diseases (n = 300), newborns (n = 124) and healthy people (n = 74). Our results showed a promising accuracy (97%) for NPC diagnosis using the combination of 7-KC, lysoSM and bile acid-408. However, contrary to our expectations, lysoGL-1 levels did not present at a significantly greater amount in NPC patients than other patients and negative controls. CONCLUSIONS The combination of 7-KC, lysoSM and bile acid-408 improves the accuracy of NPC diagnosis and is feasible for mass screening due to its simple sample preparation and measurement. Future research should investigate the chemical structure of bile acid-408 to further facilitate its advantage in diagnosis.
Collapse
Affiliation(s)
- Chen Wu
- Advanced Clinical Research Center, Institute of Neurological Disorders, Shin-Yurigaoka General Hospital, Kawasaki, Kanagawa, Japan
- Rare Disease Research Center, AnGes, Kawasaki, Kanagawa, Japan
| | - Takeo Iwamoto
- Core Research Facilities for Basic Science, The Jikei University School of Medicine, Tokyo, Japan
| | - Mohammad Arif Hossain
- Advanced Clinical Research Center, Institute of Neurological Disorders, Shin-Yurigaoka General Hospital, Kawasaki, Kanagawa, Japan
| | - Keiko Akiyama
- Advanced Clinical Research Center, Institute of Neurological Disorders, Shin-Yurigaoka General Hospital, Kawasaki, Kanagawa, Japan
| | - Junko Igarashi
- Rare Disease Research Center, AnGes, Kawasaki, Kanagawa, Japan
| | - Takashi Miyajima
- Advanced Clinical Research Center, Institute of Neurological Disorders, Shin-Yurigaoka General Hospital, Kawasaki, Kanagawa, Japan
- Rare Disease Research Center, AnGes, Kawasaki, Kanagawa, Japan
| | - Yoshikatsu Eto
- Advanced Clinical Research Center, Institute of Neurological Disorders, Shin-Yurigaoka General Hospital, Kawasaki, Kanagawa, Japan
| |
Collapse
|
26
|
Pietrobattista A, Veraldi S, Candusso M, Basso MS, Liccardo D, Della Corte C, Mosca A, Alterio T, Sacchetti E, Catesini G, Deodato F, Boenzi S, Dionisi-Vici C. The contribution of plasma oxysterols in the challenging diagnostic work-up of infantile cholestasis. Clin Chim Acta 2020; 507:181-186. [PMID: 32353361 DOI: 10.1016/j.cca.2020.04.028] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2020] [Revised: 04/22/2020] [Accepted: 04/24/2020] [Indexed: 12/28/2022]
|
27
|
Novel biomarkers for lysosomal storage disorders: Metabolomic and proteomic approaches. Clin Chim Acta 2020; 509:195-209. [PMID: 32561345 DOI: 10.1016/j.cca.2020.06.028] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2020] [Revised: 06/13/2020] [Accepted: 06/15/2020] [Indexed: 12/20/2022]
Abstract
Lysosomal storage disorders (LSDs) are characterized by the accumulation of specific disease substrates inside the lysosomes of various cells, eventually leading to the deterioration of cellular function and multisystem organ damage. With the continuous discovery and validation of novel and advanced therapies for most LSDs, there is an urgent need to discover more versatile and clinically relevant biomarkers. The utility of these biomarkers should ideally extend beyond the screening and diagnosis of LSDs to the evaluation of disease severity and monitoring of therapy. Metabolomic and proteomic approaches provide the means to the discovery and validation of such novel biomarkers. This is achieved mainly through the application of various mass spectrometric techniques to common and easily accessible biological samples, such as plasma, urine and dried blood spots. In this review, we tried to summarize the complexity of the lysosomal disorders phenotypes, their current diagnostic and therapeutic approaches, the various techniques supporting metabolomic and proteomic studies and finally we tried to explore the newly discovered biomarkers for most LSDs and their reported clinical values.
Collapse
|
28
|
Eskes ECB, Sjouke B, Vaz FM, Goorden SMI, van Kuilenburg ABP, Aerts JMFG, Hollak CEM. Biochemical and imaging parameters in acid sphingomyelinase deficiency: Potential utility as biomarkers. Mol Genet Metab 2020; 130:16-26. [PMID: 32088119 DOI: 10.1016/j.ymgme.2020.02.002] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/12/2019] [Revised: 02/03/2020] [Accepted: 02/04/2020] [Indexed: 12/19/2022]
Abstract
Acid Sphingomyelinase Deficiency (ASMD), or Niemann-Pick type A/B disease, is a rare lipid storage disorder leading to accumulation of sphingomyelin and its precursors primarily in macrophages. The disease has a broad phenotypic spectrum ranging from a fatal infantile form with severe neurological involvement (the infantile neurovisceral type) to a primarily visceral form with different degrees of pulmonary, liver, spleen and skeletal involvement (the chronic visceral type). With the upcoming possibility of treatment with enzyme replacement therapy, the need for biomarkers that predict or reflect disease progression has increased. Biomarkers should be validated for their use as surrogate markers of clinically relevant endpoints. In this review, clinically important endpoints as well as biochemical and imaging markers of ASMD are discussed and potential new biomarkers are identified. We suggest as the most promising biomarkers that may function as surrogate endpoints in the future: diffusion capacity measured by spirometry, spleen volume, platelet count, low-density lipoprotein cholesterol, liver fibrosis measured with a fibroscan, lysosphingomyelin and walked distance in six minutes. Currently, no biomarkers have been validated. Several plasma markers of lipid-laden cells, fibrosis or inflammation are of high potential as biomarkers and deserve further study. Based upon current guidelines for biomarkers, recommendations for the validation process are provided.
Collapse
Affiliation(s)
- Eline C B Eskes
- Amsterdam UMC, University of Amsterdam, Department of Endocrinology and Metabolism, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands
| | - Barbara Sjouke
- Amsterdam UMC, University of Amsterdam, Department of Endocrinology and Metabolism, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands
| | - Frédéric M Vaz
- Amsterdam UMC, University of Amsterdam, Department of Clinical Chemistry, Laboratory Genetic Metabolic Diseases, Gastroenterology & Metabolism, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands
| | - Susan M I Goorden
- Amsterdam UMC, University of Amsterdam, Department of Clinical Chemistry, Laboratory Genetic Metabolic Diseases, Gastroenterology & Metabolism, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands
| | - André B P van Kuilenburg
- Amsterdam UMC, University of Amsterdam, Department of Clinical Chemistry, Laboratory Genetic Metabolic Diseases, Gastroenterology & Metabolism, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands
| | - Johannes M F G Aerts
- Leiden Institute of Chemistry, University of Leiden, Department of Medical Biochemistry, Einsteinweg 55, 2333 CC Leiden, The Netherlands
| | - Carla E M Hollak
- Amsterdam UMC, University of Amsterdam, Department of Endocrinology and Metabolism, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands.
| |
Collapse
|
29
|
Cooper JA, Church HJ, Wu HY. Cholestane-3β, 5α, 6β-triol: Further insights into the performance of this oxysterol in diagnosis of Niemann-Pick disease type C. Mol Genet Metab 2020; 130:77-86. [PMID: 32178982 DOI: 10.1016/j.ymgme.2020.02.008] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/22/2019] [Revised: 02/21/2020] [Accepted: 02/25/2020] [Indexed: 11/20/2022]
Abstract
In recent years the oxysterol species cholestane-3β, 5α, 6β-triol (C-triol) has found application as a diagnostic biomarker for Niemann-Pick disease type C. Other studies have described increased C-triol in patients with Niemann-Pick disease type A/B and milder increases in lysosomal acid lipase deficiency (LALD), whereas they note normal C-triol levels in Smith-Lemli-Opitz syndrome (SLOS) and familial hypercholesterolaemia (FH) patients. Herein, we review data collected in our laboratory during method evaluation along with 5 years of routine analysis and present findings which differ from those reported by other groups with respect to LALD, SLOS and FH in particular, whilst providing further evidence regarding the clinical sensitivity and specificity of this biomarker, which are difficult to accurately assess. All of our Wolman disease (severe LALD) patients have demonstrated gross elevations of C-triol at diagnosis, with reduction to normal levels after induction of enzyme replacement therapy. In diagnostic specimens from SLOS patients we observed very low or undetectable C-triol levels whereas in post-therapeutic SLOS patients demonstrated normalised levels; we also describe a homozygous FH patient in which C-triol is significantly elevated. Upon investigation, we found that C-triol was formed artefactually from cholesterol during our sample preparation, i.e. this is a false positive of analytical origin; at present it is unclear whether similar effects occur during sample preparation in other laboratories. Our data demonstrates clinical sensitivity of 100% during routine application to diagnostic specimens; this is in keeping with other estimates, yet in a small proportion of patients diagnosed prior to C-triol measurement, either by Filipin staining of fibroblasts or molecular genetics, we have observed normal C-triol concentrations. Clinical specificity of C-triol alone is 93.4% and 95.3% when performed in conjunction with lysosomal enzymology. These performance statistics are very similar to those achieved with Filipin staining of cultured fibroblasts in the 5 years preceding introduction of C-triol to routine use in our laboratory. It is increasingly apparent to us that although this analyte is a very useful addition to the diagnostic tools available for NPC, with considerable advantages over more invasive and time-consuming methods, the interpretation of results is complex and should be undertaken only in light of clinical details and results of other analyses including enzymology for lysosomal acid lipase and acid sphingomyelinase.
Collapse
Affiliation(s)
- J A Cooper
- Willink Biochemical Genetics Laboratory, Manchester University NHS Foundation Trust, United Kingdom
| | - H J Church
- Willink Biochemical Genetics Laboratory, Manchester University NHS Foundation Trust, United Kingdom
| | - H Y Wu
- Willink Biochemical Genetics Laboratory, Manchester University NHS Foundation Trust, United Kingdom.
| |
Collapse
|
30
|
Nascimbeni F, Dionisi Vici C, Vespasiani Gentilucci U, Angelico F, Nobili V, Petta S, Valenti L. AISF update on the diagnosis and management of adult-onset lysosomal storage diseases with hepatic involvement. Dig Liver Dis 2020; 52:359-367. [PMID: 31902560 DOI: 10.1016/j.dld.2019.12.005] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/19/2019] [Revised: 12/07/2019] [Accepted: 12/10/2019] [Indexed: 12/11/2022]
Abstract
Lysosomal storage diseases (LSDs) are a heterogeneous group of inherited disorders caused by loss-of-function mutations in genes encoding for lysosomal enzymes/proteins. The consequence is a progressive accumulation of substrates in these intracellular organelles, resulting in cellular and tissue damage. The overall incidence is about 1/8000 live births, but is likely underestimated. LSDs are chronic progressive multi-systemic disorders, generally presenting with visceromegaly, and involvement of the central nervous system, eyes, the skeleton, and the respiratory and cardiovascular systems. The age at onset and phenotypic expression are highly variable, according to the specific enzymatic defect and tissues involved, the residual activity, and the disease-causing genotype. Enzyme-replacement therapies and substrate-reduction therapies have recently become available, leading to the improvement in symptoms, disease progression and quality of life of affected individuals. Liver involvement and hepatosplenomegaly are frequent features of LSDs and a hallmark of adult-onset forms, frequently leading to medical attention. LSDs should therefore be considered in the differential diagnosis of liver disease with organomegaly. The present document will provide a short overview of adult-onset LSDs with hepatic involvement, highlighting the specificities and systemic manifestations of the ones most frequently encountered in clinical practice, which may hint at the correct diagnosis and the appropriate treatment.
Collapse
Affiliation(s)
- Fabio Nascimbeni
- Regional Referral Centre for Lysosomal Storage Diseases, Division of Internal Medicine and Metabolism, Civil Hospital, AOU of Modena, University of Modena and Reggio Emilia, Modena, Italy
| | - Carlo Dionisi Vici
- Division of Metabolism, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | | | - Francesco Angelico
- Department of Public Health and Infective Diseases, Università Sapienza, Roma, Italy
| | - Valerio Nobili
- Division of Hepatology and Gastroenterology, Bambino Gesù Children's Hospital IRCCS, Rome, Italy
| | - Salvatore Petta
- Gastroenterology and Hepatology, PROMISE, Palermo University, Italy
| | - Luca Valenti
- Department of Pathophysiology and Transplantation, Università degli Studi di Milano, and Translational Medicine, Department of Transfusion Medicine and Hematology, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy.
| |
Collapse
|
31
|
Anderson A, Campo A, Fulton E, Corwin A, Jerome WG, O'Connor MS. 7-Ketocholesterol in disease and aging. Redox Biol 2020; 29:101380. [PMID: 31926618 PMCID: PMC6926354 DOI: 10.1016/j.redox.2019.101380] [Citation(s) in RCA: 109] [Impact Index Per Article: 21.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2019] [Revised: 11/06/2019] [Accepted: 11/10/2019] [Indexed: 02/08/2023] Open
Abstract
7-Ketocholesterol (7KC) is a toxic oxysterol that is associated with many diseases and disabilities of aging, as well as several orphan diseases. 7KC is the most common product of a reaction between cholesterol and oxygen radicals and is the most concentrated oxysterol found in the blood and arterial plaques of coronary artery disease patients as well as various other disease tissues and cell types. Unlike cholesterol, 7KC consistently shows cytotoxicity to cells and its physiological function in humans or other complex organisms is unknown. Oxysterols, particularly 7KC, have also been shown to diffuse through membranes where they affect receptor and enzymatic function. Here, we will explore the known and proposed mechanisms of pathologies that are associated with 7KC, as well speculate about the future of 7KC as a diagnostic and therapeutic target in medicine.
Collapse
|
32
|
Griffiths WJ, Yutuc E, Abdel-Khalik J, Crick PJ, Hearn T, Dickson A, Bigger BW, Hoi-Yee Wu T, Goenka A, Ghosh A, Jones SA, Covey DF, Ory DS, Wang Y. Metabolism of Non-Enzymatically Derived Oxysterols: Clues from sterol metabolic disorders. Free Radic Biol Med 2019; 144:124-133. [PMID: 31009661 PMCID: PMC6863434 DOI: 10.1016/j.freeradbiomed.2019.04.020] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/28/2019] [Revised: 04/12/2019] [Accepted: 04/16/2019] [Indexed: 12/18/2022]
Abstract
Cholestane-3β,5α,6β-triol (3β,5α,6β-triol) is formed from cholestan-5,6-epoxide (5,6-EC) in a reaction catalysed by cholesterol epoxide hydrolase, following formation of 5,6-EC through free radical oxidation of cholesterol. 7-Oxocholesterol (7-OC) and 7β-hydroxycholesterol (7β-HC) can also be formed by free radical oxidation of cholesterol. Here we investigate how 3β,5α,6β-triol, 7-OC and 7β-HC are metabolised to bile acids. We show, by monitoring oxysterol metabolites in plasma samples rich in 3β,5α,6β-triol, 7-OC and 7β-HC, that these three oxysterols fall into novel branches of the acidic pathway of bile acid biosynthesis becoming (25R)26-hydroxylated then carboxylated, 24-hydroxylated and side-chain shortened to give the final products 3β,5α,6β-trihydroxycholanoic, 3β-hydroxy-7-oxochol-5-enoic and 3β,7β-dihydroxychol-5-enoic acids, respectively. The intermediates in these pathways may be causative of some phenotypical features of, and/or have diagnostic value for, the lysosomal storage diseases, Niemann Pick types C and B and lysosomal acid lipase deficiency. Free radical derived oxysterols are metabolised in human to unusual bile acids via novel branches of the acidic pathway, intermediates in these pathways are observed in plasma.
Collapse
Affiliation(s)
- William J Griffiths
- Institute of Life Science, Swansea University Medical School, Singleton Park, Swansea, SA2 8PP, UK.
| | - Eylan Yutuc
- Institute of Life Science, Swansea University Medical School, Singleton Park, Swansea, SA2 8PP, UK
| | - Jonas Abdel-Khalik
- Institute of Life Science, Swansea University Medical School, Singleton Park, Swansea, SA2 8PP, UK
| | - Peter J Crick
- Institute of Life Science, Swansea University Medical School, Singleton Park, Swansea, SA2 8PP, UK
| | - Thomas Hearn
- Institute of Life Science, Swansea University Medical School, Singleton Park, Swansea, SA2 8PP, UK
| | - Alison Dickson
- Institute of Life Science, Swansea University Medical School, Singleton Park, Swansea, SA2 8PP, UK
| | - Brian W Bigger
- Stem Cell & Neurotherapies, Division of Cell Matrix Biology and Regenerative Medicine, Stopford Building, Oxford Road, University of Manchester, Manchester, M13 9PT, UK
| | - Teresa Hoi-Yee Wu
- Manchester Centre for Genomic Medicine, 6th Floor, St Mary's Hospital, Central Manchester Foundation Trust, University of Manchester, Oxford Road, Manchester, M13 9WL, UK
| | - Anu Goenka
- Manchester Centre for Genomic Medicine, 6th Floor, St Mary's Hospital, Central Manchester Foundation Trust, University of Manchester, Oxford Road, Manchester, M13 9WL, UK
| | - Arunabha Ghosh
- Manchester Centre for Genomic Medicine, 6th Floor, St Mary's Hospital, Central Manchester Foundation Trust, University of Manchester, Oxford Road, Manchester, M13 9WL, UK
| | - Simon A Jones
- Manchester Centre for Genomic Medicine, 6th Floor, St Mary's Hospital, Central Manchester Foundation Trust, University of Manchester, Oxford Road, Manchester, M13 9WL, UK
| | - Douglas F Covey
- Department of Developmental Biology, Washington University School of Medicine, St Louis, MO, 63110, USA
| | - Daniel S Ory
- Diabetic Cardiovascular Disease Center, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Yuqin Wang
- Institute of Life Science, Swansea University Medical School, Singleton Park, Swansea, SA2 8PP, UK.
| |
Collapse
|
33
|
Pergande MR, Serna‐Perez F, Mohsin SB, Hanek J, Cologna SM. Lipidomic Analysis Reveals Altered Fatty Acid Metabolism in the Liver of the Symptomatic Niemann–Pick, Type C1 Mouse Model. Proteomics 2019; 19:e1800285. [DOI: 10.1002/pmic.201800285] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2019] [Revised: 07/22/2019] [Indexed: 12/23/2022]
Affiliation(s)
- Melissa R. Pergande
- Department of ChemistryUniversity of Illinois at Chicago Chicago IL 60607 USA
| | - Fidel Serna‐Perez
- Department of ChemistryUniversity of Illinois at Chicago Chicago IL 60607 USA
| | | | - Jonathon Hanek
- Department of ChemistryUniversity of Illinois at Chicago Chicago IL 60607 USA
| | - Stephanie M. Cologna
- Department of ChemistryUniversity of Illinois at Chicago Chicago IL 60607 USA
- Department of ChemistryLaboratory for Integrative NeuroscienceUniversity of Illinois at Chicago Chicago IL 60607 USA
| |
Collapse
|
34
|
Screening for Niemann-Pick type C disease in neurodegenerative diseases. J Clin Neurosci 2019; 68:266-267. [PMID: 31221578 DOI: 10.1016/j.jocn.2019.06.025] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2019] [Accepted: 06/10/2019] [Indexed: 02/06/2023]
Abstract
Niemann Pick type C (NP-C) is an autosomal recessive neurovisceral lysosomal storage disorder caused by NPC1 and NPC2 gene mutations. We screened for NP-C 24 patients with Progressive Supranuclear Palsy and 10 with Multiple System Atrophy cerebellar type (MSA-C). Among PSP patients, no NPC1 or NPC2 gene variants were detected. One patient with MSA-C (10%) resulted to carry a pathogenic missense NPC1 gene mutation (p.C184Y) in heterozygous state. NPC1 genes variants might represent a risk or susceptibility factor in the development of α-synucleinopathies such as MSA. The common pattern of lysosomal dysfunction might explain the pathophysiological link between these disorders.
Collapse
|
35
|
Beck KR, Kanagaratnam S, Kratschmar DV, Birk J, Yamaguchi H, Sailer AW, Seuwen K, Odermatt A. Enzymatic interconversion of the oxysterols 7β,25-dihydroxycholesterol and 7-keto,25-hydroxycholesterol by 11β-hydroxysteroid dehydrogenase type 1 and 2. J Steroid Biochem Mol Biol 2019; 190:19-28. [PMID: 30902677 DOI: 10.1016/j.jsbmb.2019.03.011] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/29/2019] [Revised: 03/15/2019] [Accepted: 03/18/2019] [Indexed: 02/06/2023]
Abstract
Oxysterols are cholesterol metabolites derived through either autoxidation or enzymatic processes. They consist of a large family of bioactive lipids that have been associated with the progression of multiple pathologies. In order to unravel (patho-)physiological mechanisms involving oxysterols, it is crucial to elucidate the underlying formation and degradation of oxysterols. A role of 11β-hydroxysteroid dehydrogenases (11β-HSDs) in oxysterol metabolism by catalyzing the interconversion of 7-ketocholesterol (7kC) and 7β-hydroxycholesterol (7βOHC) has already been reported. The present study addresses a function of 11β-HSD1 in the enzymatic generation of 7β,25-dihydroxycholesterol (7β25OHC) from 7-keto,25-hydroxycholesterol (7k25OHC) and tested whether 11β-HSD2 is able to catalyze the reverse reaction. For the first time, using recombinant enzymes, the formation of 7k25OHC from 7kC by cholesterol 25-hydroxylase (CH25H) and further stereospecific oxoreduction to 7β25OHC by human and mouse 11β-HSD1 could be demonstrated. Additionally, experiments using human 11β-HSD2 showed the oxidation of 7β25OHC to 7k25OHC. Molecular modeling provided an explanation for the stereospecific interconversion of 7β25OHC and 7k25OHC. Production of the Epstein-Barr virus-induced gene 2 (EBI2) ligand 7β25OHC from 7k25OHC in challenged tissue by 11β-HSD1 may be important in inflammation. In conclusion, these results demonstrate a novel glucocorticoid-independent pre-receptor regulation mediated by 11β-HSDs.
Collapse
Affiliation(s)
- Katharina R Beck
- Division of Molecular and Systems Toxicology, Department of Pharmaceutical Sciences, University of Basel, Klingelbergstrasse 50, 4056 Basel, Switzerland
| | - Sharavan Kanagaratnam
- Division of Molecular and Systems Toxicology, Department of Pharmaceutical Sciences, University of Basel, Klingelbergstrasse 50, 4056 Basel, Switzerland
| | - Denise V Kratschmar
- Division of Molecular and Systems Toxicology, Department of Pharmaceutical Sciences, University of Basel, Klingelbergstrasse 50, 4056 Basel, Switzerland
| | - Julia Birk
- Division of Molecular and Systems Toxicology, Department of Pharmaceutical Sciences, University of Basel, Klingelbergstrasse 50, 4056 Basel, Switzerland
| | - Hideaki Yamaguchi
- Department of Applied Biological Chemistry, Meijo University, Nagoya 468-8502, Japan
| | - Andreas W Sailer
- Disease Area X, Novartis Institutes for BioMedical Research, Basel, Switzerland
| | - Klaus Seuwen
- Disease Area X, Novartis Institutes for BioMedical Research, Basel, Switzerland
| | - Alex Odermatt
- Division of Molecular and Systems Toxicology, Department of Pharmaceutical Sciences, University of Basel, Klingelbergstrasse 50, 4056 Basel, Switzerland.
| |
Collapse
|
36
|
Cebolla JJ, Irún P, Mozas P, Giraldo P. Evaluation of two approaches to lysosomal acid lipase deficiency patient identification: An observational retrospective study. Atherosclerosis 2019; 285:49-54. [PMID: 31004967 DOI: 10.1016/j.atherosclerosis.2019.03.013] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/23/2018] [Revised: 03/06/2019] [Accepted: 03/14/2019] [Indexed: 11/23/2022]
Abstract
BACKGROUND AND AIMS Lysosomal acid lipase deficiency (LALD) leads to the accumulation of cholesteryl esters and/or triglycerides (TG) in lysosomes due to the lack of the enzyme codified by the LIPA gene. The most common symptoms are dyslipidaemia and hypertransaminasemia, together with manifestations common to other lysosomal storage disorders (LSDs), including visceromegalies and elevated plasma biomarkers. Alteration of the lipid-liver profile (LLP) has been widely applied as a criterion for LALD screening, but the usefulness of biomarkers has not yet been explored. Our purpose was to explore the utility of plasma chitotriosidase activity (ChT) and CCL18/PARC concentration in addition to LLP to identify LALD patients in an observational retrospective study of two different sample collections. METHODS Biological samples refining: Collection 1 (primary hypercholesterolemia suspected) included unrelated individuals with hyperlipidaemia and without LDLR, APOB and PCSK9 gene mutations (Set 1), and Collection 2 (LSD suspected) included individuals without definitive LSD diagnosis (Set 2). We assessed plasma LLP (total cholesterol and its fractions, TG concentration and transaminases activities), as well as plasma ChT and CCL18/PARC. All subjects with anomalous LLP and/or biomarker levels were LIPA sequenced. RESULTS Twenty-four subjects showed altered LLP and/or biomarkers. We identified two LALD patients (one homozygous and one compound heterozygous) and one carrier of a novel LIPA variant. CONCLUSIONS The measurement of plasma ChT and CCL18/PARC combined with LLP will be a useful approach to identifying LALD patients in retrospective LALD patient studies.
Collapse
Affiliation(s)
- Jorge J Cebolla
- Instituto de Investigación Sanitaria Aragón (IIS Aragón), GIIS-012, Zaragoza, 50009, Spain; Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), CB/06/07/1036, Instituto de Salud Carlos III (ISCIII), Hospital Universitario Miguel Servet, Zaragoza, 50009, Spain; Fundación Española para el Estudio y Terapéutica de la Enfermedad de Gaucher y Otras Lisosomales (FEETEG), Zaragoza, 50008, Spain; Departamento de Bioquímica, Biología Molecular y Celular, Facultad de Ciencias, Universidad de Zaragoza, Zaragoza, 50009, Spain.
| | - Pilar Irún
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), CB/06/07/1036, Instituto de Salud Carlos III (ISCIII), Hospital Universitario Miguel Servet, Zaragoza, 50009, Spain
| | - Pilar Mozas
- Departamento de Bioquímica, Biología Molecular y Celular, Facultad de Ciencias, Universidad de Zaragoza, Zaragoza, 50009, Spain
| | - Pilar Giraldo
- Instituto de Investigación Sanitaria Aragón (IIS Aragón), GIIS-012, Zaragoza, 50009, Spain; Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), CB/06/07/1036, Instituto de Salud Carlos III (ISCIII), Hospital Universitario Miguel Servet, Zaragoza, 50009, Spain; Fundación Española para el Estudio y Terapéutica de la Enfermedad de Gaucher y Otras Lisosomales (FEETEG), Zaragoza, 50008, Spain
| |
Collapse
|
37
|
Brahmi F, Vejux A, Sghaier R, Zarrouk A, Nury T, Meddeb W, Rezig L, Namsi A, Sassi K, Yammine A, Badreddine I, Vervandier-Fasseur D, Madani K, Boulekbache-Makhlouf L, Nasser B, Lizard G. Prevention of 7-ketocholesterol-induced side effects by natural compounds. Crit Rev Food Sci Nutr 2018; 59:3179-3198. [DOI: 10.1080/10408398.2018.1491828] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Affiliation(s)
- Fatiha Brahmi
- Team ‘Biochemistry of the Peroxisome, Inflammation and Lipid Metabolism’, Lab. Bio-PeroxIL, Université de Bourgogne Franche-Comté, Dijon, France
- Lab. Biomathématique, Biochimie, Biophysique et Scientométrie, Faculté des Sciences de la Nature et de la Vie, Université de Bejaia, Bejaia, Algeria
| | - Anne Vejux
- Team ‘Biochemistry of the Peroxisome, Inflammation and Lipid Metabolism’, Lab. Bio-PeroxIL, Université de Bourgogne Franche-Comté, Dijon, France
| | - Randa Sghaier
- Team ‘Biochemistry of the Peroxisome, Inflammation and Lipid Metabolism’, Lab. Bio-PeroxIL, Université de Bourgogne Franche-Comté, Dijon, France
- Lab-NAFS ‘Nutrition - Functional Food & Vascular Health’, LR12ES05, Université de Monastir, Monastir, Tunisia
- Faculty of Medicine, Lab. Biochemistry, Sousse, Tunisia
| | - Amira Zarrouk
- Lab-NAFS ‘Nutrition - Functional Food & Vascular Health’, LR12ES05, Université de Monastir, Monastir, Tunisia
- Faculty of Medicine, Lab. Biochemistry, Sousse, Tunisia
| | - Thomas Nury
- Team ‘Biochemistry of the Peroxisome, Inflammation and Lipid Metabolism’, Lab. Bio-PeroxIL, Université de Bourgogne Franche-Comté, Dijon, France
| | - Wiem Meddeb
- Team ‘Biochemistry of the Peroxisome, Inflammation and Lipid Metabolism’, Lab. Bio-PeroxIL, Université de Bourgogne Franche-Comté, Dijon, France
- LMMA/IPEST, Faculty of Science, University of Carthage, Bizerte, Tunisia
| | - Leila Rezig
- Team ‘Biochemistry of the Peroxisome, Inflammation and Lipid Metabolism’, Lab. Bio-PeroxIL, Université de Bourgogne Franche-Comté, Dijon, France
- ESIAT, Lab. Conservation et Valorisation des Aliments, Tunis, Tunisia
| | - Amira Namsi
- Team ‘Biochemistry of the Peroxisome, Inflammation and Lipid Metabolism’, Lab. Bio-PeroxIL, Université de Bourgogne Franche-Comté, Dijon, France
- University Tunis El Manar, Faculty of Science of Tunis, Laboratory of Functional Neurophysiology and Pathology, Tunis, Tunisia
| | - Khouloud Sassi
- Team ‘Biochemistry of the Peroxisome, Inflammation and Lipid Metabolism’, Lab. Bio-PeroxIL, Université de Bourgogne Franche-Comté, Dijon, France
- Lab. Onco-Hematology, Faculty de Medicine of Tunis, Université de Tunis El Manar, Tunis, Tunisia
| | - Aline Yammine
- Team ‘Biochemistry of the Peroxisome, Inflammation and Lipid Metabolism’, Lab. Bio-PeroxIL, Université de Bourgogne Franche-Comté, Dijon, France
- Bioactive Molecules Research Lab, Faculty of Sciences, Lebanese University, Beirut, Lebanon
| | - Iham Badreddine
- Team ‘Biochemistry of the Peroxisome, Inflammation and Lipid Metabolism’, Lab. Bio-PeroxIL, Université de Bourgogne Franche-Comté, Dijon, France
- Lab. ‘Valorisation des Ressources Naturelles et Environnement’, Université Ibn Zohr, Taroudant, Morocco
| | | | - Khodir Madani
- Lab. Biomathématique, Biochimie, Biophysique et Scientométrie, Faculté des Sciences de la Nature et de la Vie, Université de Bejaia, Bejaia, Algeria
| | - Lila Boulekbache-Makhlouf
- Lab. Biomathématique, Biochimie, Biophysique et Scientométrie, Faculté des Sciences de la Nature et de la Vie, Université de Bejaia, Bejaia, Algeria
| | - Boubker Nasser
- Lab. Neuroscience and Biochemistry, Université Hassan 1er, Settat, Morocco
| | - Gérard Lizard
- Team ‘Biochemistry of the Peroxisome, Inflammation and Lipid Metabolism’, Lab. Bio-PeroxIL, Université de Bourgogne Franche-Comté, Dijon, France
| |
Collapse
|
38
|
Bezine M, Maatoug S, Ben Khalifa R, Debbabi M, Zarrouk A, Wang Y, Griffiths WJ, Nury T, Samadi M, Vejux A, de Sèze J, Moreau T, Kharrat R, El Ayeb M, Lizard G. Modulation of Kv3.1b potassium channel level and intracellular potassium concentration in 158N murine oligodendrocytes and BV-2 murine microglial cells treated with 7-ketocholesterol, 24S-hydroxycholesterol or tetracosanoic acid (C24:0). Biochimie 2018; 153:56-69. [DOI: 10.1016/j.biochi.2018.02.008] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2017] [Accepted: 02/14/2018] [Indexed: 01/19/2023]
|
39
|
Deodato F, Boenzi S, Taurisano R, Semeraro M, Sacchetti E, Carrozzo R, Dionisi-Vici C. The impact of biomarkers analysis in the diagnosis of Niemann-Pick C disease and acid sphingomyelinase deficiency. Clin Chim Acta 2018; 486:387-394. [PMID: 30153451 DOI: 10.1016/j.cca.2018.08.039] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2018] [Revised: 08/24/2018] [Accepted: 08/24/2018] [Indexed: 11/18/2022]
Abstract
BACKGROUND Although representing two distinct disease entities, Niemann-Pick disease type C (NP-C) disease and acid sphingomyelinase deficiency (ASMD) share several phenotypic features. The lack of biomarkers was responsible in the past of diagnostic delay. Recently, plasma oxysterols, cholestan-3β,5α,6β-triol (Triol) and 7-ketocholesterol (7-KC) and lysosphingolipids, Lyso-sphingomyelin (Lyso-SM) and Lysosphingomyelin-509 (Lyso-SM-509), have been proposed as diagnostic biomarkers. We aimed to assess the diagnostic power of the two biomarkers categories and to evaluate possible correlations with patients' age and clinical phenotypes. PATIENTS AND METHODS We analyzed plasma oxysterols and lysosphingolipids in patients affected by NP-C and ASMD, and compared with healthy controls. RESULTS Oxysterols were always increased in both NP-C and ASMD. In NP-C, Lyso-SM and Lyso-SM-509 were increased in 70%, and 100% of patients, respectively. Biomarkers negatively correlated with patients' age, with highest levels in early-infantile, intermediate in the late-infantile and lowest in the juvenile phenotype. In ASMD, lysosphingolipids were both increased, with a greater order of magnitude than in NP-C, with highest levels in chronic-neurovisceral vs visceral phenotype. CONCLUSIONS Lysosphingolipids are useful biomarkers for a rapid and precise diagnosis, allowing clear distinction between NP-C and ASMD. They are more reliable biomarkers than oxysterols and correlate with patients' age and clinical phenotype.
Collapse
Affiliation(s)
- Federica Deodato
- Clinical Division and Research Unit of Metabolic Diseases, Bambino Gesù Children's Hospital, IRCCS, Piazza S. Onofrio 4, 00165 Rome, Italy.
| | - Sara Boenzi
- Clinical Division and Research Unit of Metabolic Diseases, Bambino Gesù Children's Hospital, IRCCS, Piazza S. Onofrio 4, 00165 Rome, Italy.
| | - Roberta Taurisano
- Clinical Division and Research Unit of Metabolic Diseases, Bambino Gesù Children's Hospital, IRCCS, Piazza S. Onofrio 4, 00165 Rome, Italy.
| | - Michela Semeraro
- Clinical Division and Research Unit of Metabolic Diseases, Bambino Gesù Children's Hospital, IRCCS, Piazza S. Onofrio 4, 00165 Rome, Italy.
| | - Elisa Sacchetti
- Clinical Division and Research Unit of Metabolic Diseases, Bambino Gesù Children's Hospital, IRCCS, Piazza S. Onofrio 4, 00165 Rome, Italy.
| | - Rosalba Carrozzo
- Unit of Neuromuscular Diseases, Bambino Gesù Children's Hospital, IRCCS, Piazza S. Onofrio 4, 00165 Rome, Italy.
| | - Carlo Dionisi-Vici
- Clinical Division and Research Unit of Metabolic Diseases, Bambino Gesù Children's Hospital, IRCCS, Piazza S. Onofrio 4, 00165 Rome, Italy.
| |
Collapse
|
40
|
Piraud M, Pettazzoni M, Lavoie P, Ruet S, Pagan C, Cheillan D, Latour P, Vianey-Saban C, Auray-Blais C, Froissart R. Contribution of tandem mass spectrometry to the diagnosis of lysosomal storage disorders. J Inherit Metab Dis 2018; 41:457-477. [PMID: 29556840 DOI: 10.1007/s10545-017-0126-3] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/30/2017] [Revised: 11/25/2017] [Accepted: 12/11/2017] [Indexed: 02/07/2023]
Abstract
Tandem mass spectrometry (MS/MS) is a highly sensitive and specific technique. Thanks to the development of triple quadrupole analyzers, it is becoming more widely used in laboratories working in the field of inborn errors of metabolism. We review here the state of the art of this technique applied to the diagnosis of lysosomal storage disorders (LSDs) and how MS/MS has changed the diagnostic rationale in recent years. This fine technology brings more sensitive, specific, and reliable methods than the previous biochemical ones for the analysis of urinary glycosaminoglycans, oligosaccharides, and sialic acid. In sphingolipidoses, the quantification of urinary sphingolipids (globotriaosylceramide, sulfatides) is possible. The measurement of new plasmatic biomarkers such as oxysterols, bile acids, and lysosphingolipids allows the screening of many sphingolipidoses and related disorders (Niemann-Pick type C), replacing tedious biochemical techniques. Applied to amniotic fluid, a more reliable prenatal diagnosis or screening of LSDs is now available for fetuses presenting with antenatal manifestations. Applied to enzyme measurements, it allows high throughput assays for the screening of large populations, even newborn screening. The advent of this new method can modify the diagnostic rationale behind LSDs.
Collapse
Affiliation(s)
- Monique Piraud
- Unité Maladies Héréditaires du Métabolisme, Service de Biochimie et Biologie Moléculaire Grand Est, Centre de Biologie et de Pathologie Est, Hospices Civils de Lyon, 59 boulevard Pinel, 69677, Bron cedex, France.
| | - Magali Pettazzoni
- Unité Maladies Héréditaires du Métabolisme, Service de Biochimie et Biologie Moléculaire Grand Est, Centre de Biologie et de Pathologie Est, Hospices Civils de Lyon, 59 boulevard Pinel, 69677, Bron cedex, France
| | - Pamela Lavoie
- Service de Génétique Médicale, Département de Pédiatrie, Faculté de médecine et des sciences de la santé, Université de Sherbrooke, Sherbrooke, QC, Canada
| | - Séverine Ruet
- Unité Maladies Héréditaires du Métabolisme, Service de Biochimie et Biologie Moléculaire Grand Est, Centre de Biologie et de Pathologie Est, Hospices Civils de Lyon, 59 boulevard Pinel, 69677, Bron cedex, France
| | - Cécile Pagan
- Unité Maladies Héréditaires du Métabolisme, Service de Biochimie et Biologie Moléculaire Grand Est, Centre de Biologie et de Pathologie Est, Hospices Civils de Lyon, 59 boulevard Pinel, 69677, Bron cedex, France
| | - David Cheillan
- Unité Maladies Héréditaires du Métabolisme, Service de Biochimie et Biologie Moléculaire Grand Est, Centre de Biologie et de Pathologie Est, Hospices Civils de Lyon, 59 boulevard Pinel, 69677, Bron cedex, France
| | - Philippe Latour
- Unité de Neurogénétique Moléculaire, Service de Biochimie et Biologie Moléculaire Grand Est, Centre de Biologie et de Pathologie Est, Hospices Civils de Lyon, Lyon, France
| | - Christine Vianey-Saban
- Unité Maladies Héréditaires du Métabolisme, Service de Biochimie et Biologie Moléculaire Grand Est, Centre de Biologie et de Pathologie Est, Hospices Civils de Lyon, 59 boulevard Pinel, 69677, Bron cedex, France
| | - Christiane Auray-Blais
- Service de Génétique Médicale, Département de Pédiatrie, Faculté de médecine et des sciences de la santé, Université de Sherbrooke, Sherbrooke, QC, Canada
| | - Roseline Froissart
- Unité Maladies Héréditaires du Métabolisme, Service de Biochimie et Biologie Moléculaire Grand Est, Centre de Biologie et de Pathologie Est, Hospices Civils de Lyon, 59 boulevard Pinel, 69677, Bron cedex, France
| |
Collapse
|
41
|
Griffiths WJ, Gilmore I, Yutuc E, Abdel-Khalik J, Crick PJ, Hearn T, Dickson A, Bigger BW, Wu THY, Goenka A, Ghosh A, Jones SA, Wang Y. Identification of unusual oxysterols and bile acids with 7-oxo or 3β,5α,6β-trihydroxy functions in human plasma by charge-tagging mass spectrometry with multistage fragmentation. J Lipid Res 2018; 59:1058-1070. [PMID: 29626102 PMCID: PMC5983402 DOI: 10.1194/jlr.d083246] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2018] [Revised: 03/26/2018] [Indexed: 12/29/2022] Open
Abstract
7-Oxocholesterol (7-OC), 5,6-epoxycholesterol (5,6-EC), and its hydrolysis product cholestane-3β,5α,6β-triol (3β,5α,6β-triol) are normally minor oxysterols in human samples; however, in disease, their levels may be greatly elevated. This is the case in plasma from patients suffering from some lysosomal storage disorders, e.g., Niemann-Pick disease type C, or the inborn errors of sterol metabolism, e.g., Smith-Lemli-Opitz syndrome and cerebrotendinous xanthomatosis. A complication in the analysis of 7-OC and 5,6-EC is that they can also be formed ex vivo from cholesterol during sample handling in air, causing confusion with molecules formed in vivo. When formed endogenously, 7-OC, 5,6-EC, and 3β,5α,6β-triol can be converted to bile acids. Here, we describe methodology based on chemical derivatization and LC/MS with multistage fragmentation (MSn) to identify the necessary intermediates in the conversion of 7-OC to 3β-hydroxy-7-oxochol-5-enoic acid and 5,6-EC and 3β,5α,6β-triol to 3β,5α,6β-trihydroxycholanoic acid. Identification of intermediate metabolites is facilitated by their unusual MSn fragmentation patterns. Semiquantitative measurements are possible, but absolute values await the synthesis of isotope-labeled standards.
Collapse
Affiliation(s)
| | - Ian Gilmore
- Swansea University Medical School, Swansea SA2 8PP, Wales, United Kingdom
| | - Eylan Yutuc
- Swansea University Medical School, Swansea SA2 8PP, Wales, United Kingdom
| | - Jonas Abdel-Khalik
- Swansea University Medical School, Swansea SA2 8PP, Wales, United Kingdom
| | - Peter J Crick
- Swansea University Medical School, Swansea SA2 8PP, Wales, United Kingdom
| | - Thomas Hearn
- Swansea University Medical School, Swansea SA2 8PP, Wales, United Kingdom
| | - Alison Dickson
- Swansea University Medical School, Swansea SA2 8PP, Wales, United Kingdom
| | - Brian W Bigger
- Stem Cell and Neurotherapies, Division of Cell Matrix Biology and Regenerative Medicine, University of Manchester, Manchester M13 9PT, United Kingdom
| | - Teresa Hoi-Yee Wu
- Manchester Centre for Genomic Medicine, St. Mary's Hospital, Central Manchester Foundation Trust, University of Manchester, Manchester M13 9WL, United Kingdom
| | - Anu Goenka
- Manchester Centre for Genomic Medicine, St. Mary's Hospital, Central Manchester Foundation Trust, University of Manchester, Manchester M13 9WL, United Kingdom
| | - Arunabha Ghosh
- Manchester Centre for Genomic Medicine, St. Mary's Hospital, Central Manchester Foundation Trust, University of Manchester, Manchester M13 9WL, United Kingdom
| | - Simon A Jones
- Manchester Centre for Genomic Medicine, St. Mary's Hospital, Central Manchester Foundation Trust, University of Manchester, Manchester M13 9WL, United Kingdom
| | - Yuqin Wang
- Swansea University Medical School, Swansea SA2 8PP, Wales, United Kingdom.
| |
Collapse
|
42
|
Abstract
PURPOSE OF REVIEW To update researchers of recently discovered metabolites of cholesterol and of its precursors and to suggest relevant metabolic pathways. RECENT FINDINGS Patients suffering from inborn errors of sterol biosynthesis, transport and metabolism display unusual metabolic pathways, which may be major routes in the diseased state but minor in the healthy individual. Although quantitatively minor, these pathways may still be important in healthy individuals. Four inborn errors of metabolism, Smith-Lemli-Opitz syndrome, cerebrotendinous xanthomatosis and Niemann Pick disease types B (NPB) and C (NPC) result from mutations in different genes but can generate elevated levels of the same sterol metabolite, 7-oxocholesterol, in plasma. How this molecule is metabolized further is of great interest as its metabolites may have an important role in embryonic development. A second metabolite, abundant in NPC and NPB diseases, cholestane-3β,5α,6β-triol (3β,5α,6β-triol), has recently been shown to be metabolized to the corresponding bile acid, 3β,5α,6β-trihydroxycholanoic acid, providing a diagnostic marker in plasma. The origin of cholestane-3β,5α,6β-triol is likely to be 3β-hydroxycholestan-5,6-epoxide, which can alternatively be metabolized to the tumour suppressor dendrogenin A (DDA). In breast tumours, DDA levels are found to be decreased compared with normal tissues linking sterol metabolism to cancer. SUMMARY Unusual sterol metabolites and pathways may not only provide markers of disease, but also clues towards cause and treatment.
Collapse
Affiliation(s)
- Yuqin Wang
- Swansea University Medical School, ILS1 Building, Singleton Park, Swansea, UK
| | | |
Collapse
|
43
|
López de Frutos L, Cebolla JJ, Irún P, Köhler R, Giraldo P. The erythrocyte osmotic resistance test as screening tool for cholesterol-related lysosomal storage diseases. Clin Chim Acta 2018; 480:161-165. [PMID: 29447902 DOI: 10.1016/j.cca.2018.02.010] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2017] [Revised: 01/15/2018] [Accepted: 02/12/2018] [Indexed: 01/24/2023]
Abstract
BACKGROUND Erythrocyte volume regulation and membrane elasticity are essential for adaptation to osmotic and mechanical stress, and life span. Here, we evaluated whether defective cholesterol trafficking caused by the rare lysosomal storages diseases (LSDs), Niemann-Pick type C (NPC) and Lysosomal acid lipase (LAL) deficiency (LALD) impairs these properties. Moreover, we tested whether measurements of cholesterol membrane content and osmotic resistance serve as a screening test for these LSDs. METHODS Patients were genotyped for mutations in NPC1, NPC2, or LIPA genes. We measured LSD plasma biomarkers and LAL activity. Red blood cells (RBC) membrane cholesterol content was evaluated in 73 subjects. Osmotic resistance tests (ORT) were conducted in 121 blood samples from LSD suspected patients and controls. RESULTS We did not find statistically significant differences between RBC cholesterol content between subjects and controls. However, the ORT, particularly at 0.49% (w/v) hypotonic sodium chloride solution, revealed a significant higher osmotic resistance in LSDs patients than in controls. We established a cut-off value of ≤51% of haemolysis with sensibility and specificity values of 80% and 70%, respectively. CONCLUSIONS NPC and LALD do not alter cholesterol content in the RBC membrane but increase osmotic resistance. Therefore, ORT serves as screening test for the studied LSDs.
Collapse
Affiliation(s)
- Laura López de Frutos
- Instituto de Investigación Sanitaria Aragón (IIS Aragón) GIIS-012, Unidad de Investigación Traslacional, Hospital Universitario Miguel Servet, Zaragoza 50009, Spain; Fundación para el Estudio y la Terapéutica de la Enfermedad de Gaucher y Otras Lisosomales (FEETEG), Zaragoza 50009, Spain.
| | - Jorge J Cebolla
- Instituto de Investigación Sanitaria Aragón (IIS Aragón) GIIS-012, Unidad de Investigación Traslacional, Hospital Universitario Miguel Servet, Zaragoza 50009, Spain; Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER) U-752, Zaragoza 50009, Spain; Universidad de Zaragoza, Departamento de Bioquímica, Biología Molecular y Celular, Zaragoza 50009, Spain
| | - Pilar Irún
- Instituto de Investigación Sanitaria Aragón (IIS Aragón) GIIS-012, Unidad de Investigación Traslacional, Hospital Universitario Miguel Servet, Zaragoza 50009, Spain; Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER) U-752, Zaragoza 50009, Spain
| | - Ralf Köhler
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER) U-752, Zaragoza 50009, Spain; Fundación Agencia Aragonesa para la Investigación y el Desarrollo (ARAID), Zaragoza 50009, Spain
| | - Pilar Giraldo
- Instituto de Investigación Sanitaria Aragón (IIS Aragón) GIIS-012, Unidad de Investigación Traslacional, Hospital Universitario Miguel Servet, Zaragoza 50009, Spain; Fundación para el Estudio y la Terapéutica de la Enfermedad de Gaucher y Otras Lisosomales (FEETEG), Zaragoza 50009, Spain; Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER) U-752, Zaragoza 50009, Spain
| |
Collapse
|
44
|
Voorink-Moret M, Goorden SMI, van Kuilenburg ABP, Wijburg FA, Ghauharali-van der Vlugt JMM, Beers-Stet FS, Zoetekouw A, Kulik W, Hollak CEM, Vaz FM. Rapid screening for lipid storage disorders using biochemical markers. Expert center data and review of the literature. Mol Genet Metab 2018; 123:76-84. [PMID: 29290526 DOI: 10.1016/j.ymgme.2017.12.431] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/10/2017] [Revised: 12/17/2017] [Accepted: 12/17/2017] [Indexed: 12/26/2022]
Abstract
BACKGROUND In patients suspected of a lipid storage disorder (sphingolipidoses, lipidoses), confirmation of the diagnosis relies predominantly on the measurement of specific enzymatic activities and genetic studies. New UPLC-MS/MS methods have been developed to measure lysosphingolipids and oxysterols, which, combined with chitotriosidase activity may represent a rapid first tier screening for lipid storage disorders. MATERIAL AND METHODS A lysosphingolipid panel consisting of lysoglobotriaosylceramide (LysoGb3), lysohexosylceramide (LysoHexCer: both lysoglucosylceramide and lysogalactosylceramide), lysosphingomyelin (LysoSM) and its carboxylated analogue lysosphingomyelin-509 (LysoSM-509) was measured in control subjects and plasma samples of predominantly untreated patients affected with lipid storage disorders (n=74). In addition, the oxysterols cholestane-3β,5α,6β-triol and 7-ketocholesterol were measured in a subset of these patients (n=36) as well as chitotriosidase activity (n=43). A systematic review of the literature was performed to assess the usefulness of these biochemical markers. RESULTS Specific elevations of metabolites, i.e. without overlap between controls and other lipid storage disorders, were found for several lysosomal storage diseases: increased LysoSM levels in acid sphingomyelinase deficiency (Niemann-Pick disease type A/B), LysoGb3 levels in males with classical phenotype Fabry disease and LysoHexCer (i.e. lysoglucosylceramide/lysogalactosylceramide) in Gaucher and Krabbe diseases. While elevated levels of LysoSM-509 and cholestane-3β,5α,6β-triol did not discriminate between Niemann Pick disease type C and acid sphingomyelinase deficiency, LysoSM-509/LysoSM ratio was specifically elevated in Niemann-Pick disease type C. In Gaucher disease type I, mild increases in several lysosphingolipids were found including LysoGb3 with levels in the range of non-classical Fabry males and females. Chitotriosidase showed specific elevations in symptomatic Gaucher disease, and was mildly elevated in all other lipid storage disorders. Review of the literature identified 44 publications. Most findings were in line with our cohort. Several moderate elevations of biochemical markers were found across a wide range of other, mainly inherited metabolic, diseases. CONCLUSION Measurement in plasma of LysoSLs and oxysterols by UPLC-MS/MS in combination with activity of chitotriosidase provides a useful first tier screening of patients suspected of lipid storage disease. The LysoSM-509/LysoSM ratio is a promising parameter in Niemann-Pick disease type C. Further studies in larger groups of untreated patients and controls are needed to improve the specificity of the findings.
Collapse
Affiliation(s)
- M Voorink-Moret
- Department of Endocrinology and Metabolism, Academic Medical Center, University of Amsterdam, The Netherlands.
| | - S M I Goorden
- Laboratory Genetic Metabolic Diseases, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands.
| | - A B P van Kuilenburg
- Laboratory Genetic Metabolic Diseases, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands.
| | - F A Wijburg
- Department of Pediatrics, Academic Medical Center, University of Amsterdam, The Netherlands.
| | | | - F S Beers-Stet
- Laboratory Genetic Metabolic Diseases, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands.
| | - A Zoetekouw
- Laboratory Genetic Metabolic Diseases, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands.
| | - W Kulik
- Laboratory Genetic Metabolic Diseases, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands.
| | - C E M Hollak
- Department of Endocrinology and Metabolism, Academic Medical Center, University of Amsterdam, The Netherlands.
| | - F M Vaz
- Laboratory Genetic Metabolic Diseases, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands.
| |
Collapse
|
45
|
Hammerschmidt TG, Oliveira Schmitt Ribas G, Saraiva‐Pereira ML, Bonatto MP, Kessler RG, Souza FTS, Trapp F, Michelin‐Tirelli K, Burin MG, Giugliani R, Vargas CR. Molecular and biochemical biomarkers for diagnosis and therapy monitorization of Niemann‐Pick type C patients. Int J Dev Neurosci 2017; 66:18-23. [DOI: 10.1016/j.ijdevneu.2017.11.007] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2017] [Revised: 11/27/2017] [Accepted: 11/28/2017] [Indexed: 11/29/2022] Open
Affiliation(s)
- Tatiane Grazieli Hammerschmidt
- Departamento de Análises, Faculdade de FarmáciaUFRGSAvenida Ipiranga, 2752CEP 90610‐000Porto AlegreRSBrazil
- Programa de Pós‐Graduação em Ciências Farmacêuticas, UFRGSAv. Ipiranga, 2752CEP 90610‐000Porto AlegreRSBrazil
| | - Graziela Oliveira Schmitt Ribas
- Programa de Pós‐Graduação em Ciências Farmacêuticas, UFRGSAv. Ipiranga, 2752CEP 90610‐000Porto AlegreRSBrazil
- Serviço de Genética Médica, HCPARua Ramiro Barcelos, 2350CEP 90035‐003Porto AlegreRSBrazil
| | - Maria Luiza Saraiva‐Pereira
- Programa de Pós‐Graduação em Ciências Biológicas, Bioquímica, UFRGSRua Ramiro Barcelos, 2600CEP 90035‐003Porto AlegreRSBrazil
- Programa de Pós‐Graduação em Ciências Farmacêuticas, UFRGSAv. Ipiranga, 2752CEP 90610‐000Porto AlegreRSBrazil
| | - Márcia Polese Bonatto
- Programa de Pós‐Graduação em Ciências Biológicas, Bioquímica, UFRGSRua Ramiro Barcelos, 2600CEP 90035‐003Porto AlegreRSBrazil
| | - Rejane Gus Kessler
- Programa de Pós‐Graduação em Ciências Biológicas, Bioquímica, UFRGSRua Ramiro Barcelos, 2600CEP 90035‐003Porto AlegreRSBrazil
- Departamento de GenéticaIB, Universidade Federal do Rio Grande do SulPorto AlegreRSBrazil
| | - Fernanda Timm Seabra Souza
- Programa de Pós‐Graduação em Ciências Biológicas, Bioquímica, UFRGSRua Ramiro Barcelos, 2600CEP 90035‐003Porto AlegreRSBrazil
- Serviço de Genética Médica, HCPARua Ramiro Barcelos, 2350CEP 90035‐003Porto AlegreRSBrazil
| | - Franciele Trapp
- Serviço de Genética Médica, HCPARua Ramiro Barcelos, 2350CEP 90035‐003Porto AlegreRSBrazil
| | - Kristiane Michelin‐Tirelli
- Programa de Pós‐Graduação em Ciências Biológicas, Bioquímica, UFRGSRua Ramiro Barcelos, 2600CEP 90035‐003Porto AlegreRSBrazil
- Serviço de Genética Médica, HCPARua Ramiro Barcelos, 2350CEP 90035‐003Porto AlegreRSBrazil
| | - Maira Graeff Burin
- Programa de Pós‐Graduação em Ciências Biológicas, Bioquímica, UFRGSRua Ramiro Barcelos, 2600CEP 90035‐003Porto AlegreRSBrazil
- Serviço de Genética Médica, HCPARua Ramiro Barcelos, 2350CEP 90035‐003Porto AlegreRSBrazil
| | - Roberto Giugliani
- Programa de Pós‐Graduação em Ciências Biológicas, Bioquímica, UFRGSRua Ramiro Barcelos, 2600CEP 90035‐003Porto AlegreRSBrazil
- Serviço de Genética Médica, HCPARua Ramiro Barcelos, 2350CEP 90035‐003Porto AlegreRSBrazil
- Departamento de GenéticaIB, Universidade Federal do Rio Grande do SulPorto AlegreRSBrazil
| | - Carmen Regla Vargas
- Departamento de Análises, Faculdade de FarmáciaUFRGSAvenida Ipiranga, 2752CEP 90610‐000Porto AlegreRSBrazil
- Programa de Pós‐Graduação em Ciências Biológicas, Bioquímica, UFRGSRua Ramiro Barcelos, 2600CEP 90035‐003Porto AlegreRSBrazil
- Programa de Pós‐Graduação em Ciências Farmacêuticas, UFRGSAv. Ipiranga, 2752CEP 90610‐000Porto AlegreRSBrazil
- Serviço de Genética Médica, HCPARua Ramiro Barcelos, 2350CEP 90035‐003Porto AlegreRSBrazil
| |
Collapse
|
46
|
Patterson MC, Clayton P, Gissen P, Anheim M, Bauer P, Bonnot O, Dardis A, Dionisi-Vici C, Klünemann HH, Latour P, Lourenço CM, Ory DS, Parker A, Pocoví M, Strupp M, Vanier MT, Walterfang M, Marquardt T. Recommendations for the detection and diagnosis of Niemann-Pick disease type C: An update. Neurol Clin Pract 2017; 7:499-511. [PMID: 29431164 PMCID: PMC5800709 DOI: 10.1212/cpj.0000000000000399] [Citation(s) in RCA: 101] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Purpose of review: Niemann-Pick disease type C (NP-C) is a neurovisceral disorder that may be more prevalent than earlier estimates. Diagnosis of NP-C is often delayed; a key aim for clinical practice is to reduce this delay. Recently, substantial progress has been made in the field of NP-C screening and diagnosis, justifying an update to the existing recommendations for clinical practice. Recent findings: New biomarker profiling and genetic analysis technologies are included as first-line diagnostic tests for NP-C. Most diagnoses can now be confirmed by combination of biomarker and genetic analyses. Filipin staining may facilitate diagnosis in uncertain cases. Recommendations are provided for psychiatrists, neuro-ophthalmologists, and radiologists, and on screening within specific at-risk patient cohorts. The NP-C diagnostic algorithm has been updated and simplified. Summary: This publication provides expert recommendations for clinicians who may see patients presenting with the signs and symptoms of NP-C, including general practitioners, pediatricians, neurologists, and psychiatrists.
Collapse
Affiliation(s)
- Marc C Patterson
- Mayo Clinic (MCP), Rochester, MN; UCL Great Ormond Street Institute of Child Health (PC, PG), London, UK; Great Ormond Street Hospital (PG), London, UK; Département de Neurologie (MA), Hôpital de Hautepierre, CHU de Strasbourg; Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC) (MA), INSERM-U964/CNRS-UMR7104/Université de Strasbourg, Illkirch; Fédération de Médecine Translationnelle de Strasbourg (FMTS) (MA), Université de Strasbourg, France; Institute of Medical Genetics and Applied Genomics (PB), University Hospital of Tübingen; Centogene AG (PB), Rostock, Germany; Universitaire de Psychiatrie de l'Enfant et de l'Adolescent (OB), CHU de Nantes, France; Regional Coordinator Centre for Rare Diseases (AD), University Hospital Santa Maria della Misericordia, Udine, Italy; Division of Metabolism, Bambino Gesù Children's Hospital (CD-V), Rome, Italy; Klinik und Poliklinik für Psychiatrie, Psychosomatik und Psychotherapie der Universität Regensburg am Bezirksklinikum (H-HK), Regensburg, Germany; Hospices Civils de Lyon-Centre de Biologie et Pathologie Est (PL), Bron, France; University of São Paulo (HCFMRP-USP) (CML), Ribeirão Preto, SP, Brazil; Department of Medicine (DSO), Washington University, St Louis, MO; Child Development Centre (AP), Addenbrooke's Hospital, Cambridge, UK; University of Zaragoza (MP), IIS Aragon, Spain; Department of Neurology and German Center for Vertigo and Balance Disorders (MS), University Hospital Munich, Germany; Laboratoire Gillet-Mérieux (MTV), Centre de Biologie et Pathologie Est, Hospices Civils de Lyon, Bron, France; Department of Neuropsychiatry (MW), Royal Melbourne Hospital & University of Melbourne, Australia; and Universitätsklinikum Münster (TM), Germany
| | - Peter Clayton
- Mayo Clinic (MCP), Rochester, MN; UCL Great Ormond Street Institute of Child Health (PC, PG), London, UK; Great Ormond Street Hospital (PG), London, UK; Département de Neurologie (MA), Hôpital de Hautepierre, CHU de Strasbourg; Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC) (MA), INSERM-U964/CNRS-UMR7104/Université de Strasbourg, Illkirch; Fédération de Médecine Translationnelle de Strasbourg (FMTS) (MA), Université de Strasbourg, France; Institute of Medical Genetics and Applied Genomics (PB), University Hospital of Tübingen; Centogene AG (PB), Rostock, Germany; Universitaire de Psychiatrie de l'Enfant et de l'Adolescent (OB), CHU de Nantes, France; Regional Coordinator Centre for Rare Diseases (AD), University Hospital Santa Maria della Misericordia, Udine, Italy; Division of Metabolism, Bambino Gesù Children's Hospital (CD-V), Rome, Italy; Klinik und Poliklinik für Psychiatrie, Psychosomatik und Psychotherapie der Universität Regensburg am Bezirksklinikum (H-HK), Regensburg, Germany; Hospices Civils de Lyon-Centre de Biologie et Pathologie Est (PL), Bron, France; University of São Paulo (HCFMRP-USP) (CML), Ribeirão Preto, SP, Brazil; Department of Medicine (DSO), Washington University, St Louis, MO; Child Development Centre (AP), Addenbrooke's Hospital, Cambridge, UK; University of Zaragoza (MP), IIS Aragon, Spain; Department of Neurology and German Center for Vertigo and Balance Disorders (MS), University Hospital Munich, Germany; Laboratoire Gillet-Mérieux (MTV), Centre de Biologie et Pathologie Est, Hospices Civils de Lyon, Bron, France; Department of Neuropsychiatry (MW), Royal Melbourne Hospital & University of Melbourne, Australia; and Universitätsklinikum Münster (TM), Germany
| | - Paul Gissen
- Mayo Clinic (MCP), Rochester, MN; UCL Great Ormond Street Institute of Child Health (PC, PG), London, UK; Great Ormond Street Hospital (PG), London, UK; Département de Neurologie (MA), Hôpital de Hautepierre, CHU de Strasbourg; Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC) (MA), INSERM-U964/CNRS-UMR7104/Université de Strasbourg, Illkirch; Fédération de Médecine Translationnelle de Strasbourg (FMTS) (MA), Université de Strasbourg, France; Institute of Medical Genetics and Applied Genomics (PB), University Hospital of Tübingen; Centogene AG (PB), Rostock, Germany; Universitaire de Psychiatrie de l'Enfant et de l'Adolescent (OB), CHU de Nantes, France; Regional Coordinator Centre for Rare Diseases (AD), University Hospital Santa Maria della Misericordia, Udine, Italy; Division of Metabolism, Bambino Gesù Children's Hospital (CD-V), Rome, Italy; Klinik und Poliklinik für Psychiatrie, Psychosomatik und Psychotherapie der Universität Regensburg am Bezirksklinikum (H-HK), Regensburg, Germany; Hospices Civils de Lyon-Centre de Biologie et Pathologie Est (PL), Bron, France; University of São Paulo (HCFMRP-USP) (CML), Ribeirão Preto, SP, Brazil; Department of Medicine (DSO), Washington University, St Louis, MO; Child Development Centre (AP), Addenbrooke's Hospital, Cambridge, UK; University of Zaragoza (MP), IIS Aragon, Spain; Department of Neurology and German Center for Vertigo and Balance Disorders (MS), University Hospital Munich, Germany; Laboratoire Gillet-Mérieux (MTV), Centre de Biologie et Pathologie Est, Hospices Civils de Lyon, Bron, France; Department of Neuropsychiatry (MW), Royal Melbourne Hospital & University of Melbourne, Australia; and Universitätsklinikum Münster (TM), Germany
| | - Mathieu Anheim
- Mayo Clinic (MCP), Rochester, MN; UCL Great Ormond Street Institute of Child Health (PC, PG), London, UK; Great Ormond Street Hospital (PG), London, UK; Département de Neurologie (MA), Hôpital de Hautepierre, CHU de Strasbourg; Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC) (MA), INSERM-U964/CNRS-UMR7104/Université de Strasbourg, Illkirch; Fédération de Médecine Translationnelle de Strasbourg (FMTS) (MA), Université de Strasbourg, France; Institute of Medical Genetics and Applied Genomics (PB), University Hospital of Tübingen; Centogene AG (PB), Rostock, Germany; Universitaire de Psychiatrie de l'Enfant et de l'Adolescent (OB), CHU de Nantes, France; Regional Coordinator Centre for Rare Diseases (AD), University Hospital Santa Maria della Misericordia, Udine, Italy; Division of Metabolism, Bambino Gesù Children's Hospital (CD-V), Rome, Italy; Klinik und Poliklinik für Psychiatrie, Psychosomatik und Psychotherapie der Universität Regensburg am Bezirksklinikum (H-HK), Regensburg, Germany; Hospices Civils de Lyon-Centre de Biologie et Pathologie Est (PL), Bron, France; University of São Paulo (HCFMRP-USP) (CML), Ribeirão Preto, SP, Brazil; Department of Medicine (DSO), Washington University, St Louis, MO; Child Development Centre (AP), Addenbrooke's Hospital, Cambridge, UK; University of Zaragoza (MP), IIS Aragon, Spain; Department of Neurology and German Center for Vertigo and Balance Disorders (MS), University Hospital Munich, Germany; Laboratoire Gillet-Mérieux (MTV), Centre de Biologie et Pathologie Est, Hospices Civils de Lyon, Bron, France; Department of Neuropsychiatry (MW), Royal Melbourne Hospital & University of Melbourne, Australia; and Universitätsklinikum Münster (TM), Germany
| | - Peter Bauer
- Mayo Clinic (MCP), Rochester, MN; UCL Great Ormond Street Institute of Child Health (PC, PG), London, UK; Great Ormond Street Hospital (PG), London, UK; Département de Neurologie (MA), Hôpital de Hautepierre, CHU de Strasbourg; Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC) (MA), INSERM-U964/CNRS-UMR7104/Université de Strasbourg, Illkirch; Fédération de Médecine Translationnelle de Strasbourg (FMTS) (MA), Université de Strasbourg, France; Institute of Medical Genetics and Applied Genomics (PB), University Hospital of Tübingen; Centogene AG (PB), Rostock, Germany; Universitaire de Psychiatrie de l'Enfant et de l'Adolescent (OB), CHU de Nantes, France; Regional Coordinator Centre for Rare Diseases (AD), University Hospital Santa Maria della Misericordia, Udine, Italy; Division of Metabolism, Bambino Gesù Children's Hospital (CD-V), Rome, Italy; Klinik und Poliklinik für Psychiatrie, Psychosomatik und Psychotherapie der Universität Regensburg am Bezirksklinikum (H-HK), Regensburg, Germany; Hospices Civils de Lyon-Centre de Biologie et Pathologie Est (PL), Bron, France; University of São Paulo (HCFMRP-USP) (CML), Ribeirão Preto, SP, Brazil; Department of Medicine (DSO), Washington University, St Louis, MO; Child Development Centre (AP), Addenbrooke's Hospital, Cambridge, UK; University of Zaragoza (MP), IIS Aragon, Spain; Department of Neurology and German Center for Vertigo and Balance Disorders (MS), University Hospital Munich, Germany; Laboratoire Gillet-Mérieux (MTV), Centre de Biologie et Pathologie Est, Hospices Civils de Lyon, Bron, France; Department of Neuropsychiatry (MW), Royal Melbourne Hospital & University of Melbourne, Australia; and Universitätsklinikum Münster (TM), Germany
| | - Olivier Bonnot
- Mayo Clinic (MCP), Rochester, MN; UCL Great Ormond Street Institute of Child Health (PC, PG), London, UK; Great Ormond Street Hospital (PG), London, UK; Département de Neurologie (MA), Hôpital de Hautepierre, CHU de Strasbourg; Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC) (MA), INSERM-U964/CNRS-UMR7104/Université de Strasbourg, Illkirch; Fédération de Médecine Translationnelle de Strasbourg (FMTS) (MA), Université de Strasbourg, France; Institute of Medical Genetics and Applied Genomics (PB), University Hospital of Tübingen; Centogene AG (PB), Rostock, Germany; Universitaire de Psychiatrie de l'Enfant et de l'Adolescent (OB), CHU de Nantes, France; Regional Coordinator Centre for Rare Diseases (AD), University Hospital Santa Maria della Misericordia, Udine, Italy; Division of Metabolism, Bambino Gesù Children's Hospital (CD-V), Rome, Italy; Klinik und Poliklinik für Psychiatrie, Psychosomatik und Psychotherapie der Universität Regensburg am Bezirksklinikum (H-HK), Regensburg, Germany; Hospices Civils de Lyon-Centre de Biologie et Pathologie Est (PL), Bron, France; University of São Paulo (HCFMRP-USP) (CML), Ribeirão Preto, SP, Brazil; Department of Medicine (DSO), Washington University, St Louis, MO; Child Development Centre (AP), Addenbrooke's Hospital, Cambridge, UK; University of Zaragoza (MP), IIS Aragon, Spain; Department of Neurology and German Center for Vertigo and Balance Disorders (MS), University Hospital Munich, Germany; Laboratoire Gillet-Mérieux (MTV), Centre de Biologie et Pathologie Est, Hospices Civils de Lyon, Bron, France; Department of Neuropsychiatry (MW), Royal Melbourne Hospital & University of Melbourne, Australia; and Universitätsklinikum Münster (TM), Germany
| | - Andrea Dardis
- Mayo Clinic (MCP), Rochester, MN; UCL Great Ormond Street Institute of Child Health (PC, PG), London, UK; Great Ormond Street Hospital (PG), London, UK; Département de Neurologie (MA), Hôpital de Hautepierre, CHU de Strasbourg; Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC) (MA), INSERM-U964/CNRS-UMR7104/Université de Strasbourg, Illkirch; Fédération de Médecine Translationnelle de Strasbourg (FMTS) (MA), Université de Strasbourg, France; Institute of Medical Genetics and Applied Genomics (PB), University Hospital of Tübingen; Centogene AG (PB), Rostock, Germany; Universitaire de Psychiatrie de l'Enfant et de l'Adolescent (OB), CHU de Nantes, France; Regional Coordinator Centre for Rare Diseases (AD), University Hospital Santa Maria della Misericordia, Udine, Italy; Division of Metabolism, Bambino Gesù Children's Hospital (CD-V), Rome, Italy; Klinik und Poliklinik für Psychiatrie, Psychosomatik und Psychotherapie der Universität Regensburg am Bezirksklinikum (H-HK), Regensburg, Germany; Hospices Civils de Lyon-Centre de Biologie et Pathologie Est (PL), Bron, France; University of São Paulo (HCFMRP-USP) (CML), Ribeirão Preto, SP, Brazil; Department of Medicine (DSO), Washington University, St Louis, MO; Child Development Centre (AP), Addenbrooke's Hospital, Cambridge, UK; University of Zaragoza (MP), IIS Aragon, Spain; Department of Neurology and German Center for Vertigo and Balance Disorders (MS), University Hospital Munich, Germany; Laboratoire Gillet-Mérieux (MTV), Centre de Biologie et Pathologie Est, Hospices Civils de Lyon, Bron, France; Department of Neuropsychiatry (MW), Royal Melbourne Hospital & University of Melbourne, Australia; and Universitätsklinikum Münster (TM), Germany
| | - Carlo Dionisi-Vici
- Mayo Clinic (MCP), Rochester, MN; UCL Great Ormond Street Institute of Child Health (PC, PG), London, UK; Great Ormond Street Hospital (PG), London, UK; Département de Neurologie (MA), Hôpital de Hautepierre, CHU de Strasbourg; Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC) (MA), INSERM-U964/CNRS-UMR7104/Université de Strasbourg, Illkirch; Fédération de Médecine Translationnelle de Strasbourg (FMTS) (MA), Université de Strasbourg, France; Institute of Medical Genetics and Applied Genomics (PB), University Hospital of Tübingen; Centogene AG (PB), Rostock, Germany; Universitaire de Psychiatrie de l'Enfant et de l'Adolescent (OB), CHU de Nantes, France; Regional Coordinator Centre for Rare Diseases (AD), University Hospital Santa Maria della Misericordia, Udine, Italy; Division of Metabolism, Bambino Gesù Children's Hospital (CD-V), Rome, Italy; Klinik und Poliklinik für Psychiatrie, Psychosomatik und Psychotherapie der Universität Regensburg am Bezirksklinikum (H-HK), Regensburg, Germany; Hospices Civils de Lyon-Centre de Biologie et Pathologie Est (PL), Bron, France; University of São Paulo (HCFMRP-USP) (CML), Ribeirão Preto, SP, Brazil; Department of Medicine (DSO), Washington University, St Louis, MO; Child Development Centre (AP), Addenbrooke's Hospital, Cambridge, UK; University of Zaragoza (MP), IIS Aragon, Spain; Department of Neurology and German Center for Vertigo and Balance Disorders (MS), University Hospital Munich, Germany; Laboratoire Gillet-Mérieux (MTV), Centre de Biologie et Pathologie Est, Hospices Civils de Lyon, Bron, France; Department of Neuropsychiatry (MW), Royal Melbourne Hospital & University of Melbourne, Australia; and Universitätsklinikum Münster (TM), Germany
| | - Hans-Hermann Klünemann
- Mayo Clinic (MCP), Rochester, MN; UCL Great Ormond Street Institute of Child Health (PC, PG), London, UK; Great Ormond Street Hospital (PG), London, UK; Département de Neurologie (MA), Hôpital de Hautepierre, CHU de Strasbourg; Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC) (MA), INSERM-U964/CNRS-UMR7104/Université de Strasbourg, Illkirch; Fédération de Médecine Translationnelle de Strasbourg (FMTS) (MA), Université de Strasbourg, France; Institute of Medical Genetics and Applied Genomics (PB), University Hospital of Tübingen; Centogene AG (PB), Rostock, Germany; Universitaire de Psychiatrie de l'Enfant et de l'Adolescent (OB), CHU de Nantes, France; Regional Coordinator Centre for Rare Diseases (AD), University Hospital Santa Maria della Misericordia, Udine, Italy; Division of Metabolism, Bambino Gesù Children's Hospital (CD-V), Rome, Italy; Klinik und Poliklinik für Psychiatrie, Psychosomatik und Psychotherapie der Universität Regensburg am Bezirksklinikum (H-HK), Regensburg, Germany; Hospices Civils de Lyon-Centre de Biologie et Pathologie Est (PL), Bron, France; University of São Paulo (HCFMRP-USP) (CML), Ribeirão Preto, SP, Brazil; Department of Medicine (DSO), Washington University, St Louis, MO; Child Development Centre (AP), Addenbrooke's Hospital, Cambridge, UK; University of Zaragoza (MP), IIS Aragon, Spain; Department of Neurology and German Center for Vertigo and Balance Disorders (MS), University Hospital Munich, Germany; Laboratoire Gillet-Mérieux (MTV), Centre de Biologie et Pathologie Est, Hospices Civils de Lyon, Bron, France; Department of Neuropsychiatry (MW), Royal Melbourne Hospital & University of Melbourne, Australia; and Universitätsklinikum Münster (TM), Germany
| | - Philippe Latour
- Mayo Clinic (MCP), Rochester, MN; UCL Great Ormond Street Institute of Child Health (PC, PG), London, UK; Great Ormond Street Hospital (PG), London, UK; Département de Neurologie (MA), Hôpital de Hautepierre, CHU de Strasbourg; Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC) (MA), INSERM-U964/CNRS-UMR7104/Université de Strasbourg, Illkirch; Fédération de Médecine Translationnelle de Strasbourg (FMTS) (MA), Université de Strasbourg, France; Institute of Medical Genetics and Applied Genomics (PB), University Hospital of Tübingen; Centogene AG (PB), Rostock, Germany; Universitaire de Psychiatrie de l'Enfant et de l'Adolescent (OB), CHU de Nantes, France; Regional Coordinator Centre for Rare Diseases (AD), University Hospital Santa Maria della Misericordia, Udine, Italy; Division of Metabolism, Bambino Gesù Children's Hospital (CD-V), Rome, Italy; Klinik und Poliklinik für Psychiatrie, Psychosomatik und Psychotherapie der Universität Regensburg am Bezirksklinikum (H-HK), Regensburg, Germany; Hospices Civils de Lyon-Centre de Biologie et Pathologie Est (PL), Bron, France; University of São Paulo (HCFMRP-USP) (CML), Ribeirão Preto, SP, Brazil; Department of Medicine (DSO), Washington University, St Louis, MO; Child Development Centre (AP), Addenbrooke's Hospital, Cambridge, UK; University of Zaragoza (MP), IIS Aragon, Spain; Department of Neurology and German Center for Vertigo and Balance Disorders (MS), University Hospital Munich, Germany; Laboratoire Gillet-Mérieux (MTV), Centre de Biologie et Pathologie Est, Hospices Civils de Lyon, Bron, France; Department of Neuropsychiatry (MW), Royal Melbourne Hospital & University of Melbourne, Australia; and Universitätsklinikum Münster (TM), Germany
| | - Charles M Lourenço
- Mayo Clinic (MCP), Rochester, MN; UCL Great Ormond Street Institute of Child Health (PC, PG), London, UK; Great Ormond Street Hospital (PG), London, UK; Département de Neurologie (MA), Hôpital de Hautepierre, CHU de Strasbourg; Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC) (MA), INSERM-U964/CNRS-UMR7104/Université de Strasbourg, Illkirch; Fédération de Médecine Translationnelle de Strasbourg (FMTS) (MA), Université de Strasbourg, France; Institute of Medical Genetics and Applied Genomics (PB), University Hospital of Tübingen; Centogene AG (PB), Rostock, Germany; Universitaire de Psychiatrie de l'Enfant et de l'Adolescent (OB), CHU de Nantes, France; Regional Coordinator Centre for Rare Diseases (AD), University Hospital Santa Maria della Misericordia, Udine, Italy; Division of Metabolism, Bambino Gesù Children's Hospital (CD-V), Rome, Italy; Klinik und Poliklinik für Psychiatrie, Psychosomatik und Psychotherapie der Universität Regensburg am Bezirksklinikum (H-HK), Regensburg, Germany; Hospices Civils de Lyon-Centre de Biologie et Pathologie Est (PL), Bron, France; University of São Paulo (HCFMRP-USP) (CML), Ribeirão Preto, SP, Brazil; Department of Medicine (DSO), Washington University, St Louis, MO; Child Development Centre (AP), Addenbrooke's Hospital, Cambridge, UK; University of Zaragoza (MP), IIS Aragon, Spain; Department of Neurology and German Center for Vertigo and Balance Disorders (MS), University Hospital Munich, Germany; Laboratoire Gillet-Mérieux (MTV), Centre de Biologie et Pathologie Est, Hospices Civils de Lyon, Bron, France; Department of Neuropsychiatry (MW), Royal Melbourne Hospital & University of Melbourne, Australia; and Universitätsklinikum Münster (TM), Germany
| | - Daniel S Ory
- Mayo Clinic (MCP), Rochester, MN; UCL Great Ormond Street Institute of Child Health (PC, PG), London, UK; Great Ormond Street Hospital (PG), London, UK; Département de Neurologie (MA), Hôpital de Hautepierre, CHU de Strasbourg; Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC) (MA), INSERM-U964/CNRS-UMR7104/Université de Strasbourg, Illkirch; Fédération de Médecine Translationnelle de Strasbourg (FMTS) (MA), Université de Strasbourg, France; Institute of Medical Genetics and Applied Genomics (PB), University Hospital of Tübingen; Centogene AG (PB), Rostock, Germany; Universitaire de Psychiatrie de l'Enfant et de l'Adolescent (OB), CHU de Nantes, France; Regional Coordinator Centre for Rare Diseases (AD), University Hospital Santa Maria della Misericordia, Udine, Italy; Division of Metabolism, Bambino Gesù Children's Hospital (CD-V), Rome, Italy; Klinik und Poliklinik für Psychiatrie, Psychosomatik und Psychotherapie der Universität Regensburg am Bezirksklinikum (H-HK), Regensburg, Germany; Hospices Civils de Lyon-Centre de Biologie et Pathologie Est (PL), Bron, France; University of São Paulo (HCFMRP-USP) (CML), Ribeirão Preto, SP, Brazil; Department of Medicine (DSO), Washington University, St Louis, MO; Child Development Centre (AP), Addenbrooke's Hospital, Cambridge, UK; University of Zaragoza (MP), IIS Aragon, Spain; Department of Neurology and German Center for Vertigo and Balance Disorders (MS), University Hospital Munich, Germany; Laboratoire Gillet-Mérieux (MTV), Centre de Biologie et Pathologie Est, Hospices Civils de Lyon, Bron, France; Department of Neuropsychiatry (MW), Royal Melbourne Hospital & University of Melbourne, Australia; and Universitätsklinikum Münster (TM), Germany
| | - Alasdair Parker
- Mayo Clinic (MCP), Rochester, MN; UCL Great Ormond Street Institute of Child Health (PC, PG), London, UK; Great Ormond Street Hospital (PG), London, UK; Département de Neurologie (MA), Hôpital de Hautepierre, CHU de Strasbourg; Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC) (MA), INSERM-U964/CNRS-UMR7104/Université de Strasbourg, Illkirch; Fédération de Médecine Translationnelle de Strasbourg (FMTS) (MA), Université de Strasbourg, France; Institute of Medical Genetics and Applied Genomics (PB), University Hospital of Tübingen; Centogene AG (PB), Rostock, Germany; Universitaire de Psychiatrie de l'Enfant et de l'Adolescent (OB), CHU de Nantes, France; Regional Coordinator Centre for Rare Diseases (AD), University Hospital Santa Maria della Misericordia, Udine, Italy; Division of Metabolism, Bambino Gesù Children's Hospital (CD-V), Rome, Italy; Klinik und Poliklinik für Psychiatrie, Psychosomatik und Psychotherapie der Universität Regensburg am Bezirksklinikum (H-HK), Regensburg, Germany; Hospices Civils de Lyon-Centre de Biologie et Pathologie Est (PL), Bron, France; University of São Paulo (HCFMRP-USP) (CML), Ribeirão Preto, SP, Brazil; Department of Medicine (DSO), Washington University, St Louis, MO; Child Development Centre (AP), Addenbrooke's Hospital, Cambridge, UK; University of Zaragoza (MP), IIS Aragon, Spain; Department of Neurology and German Center for Vertigo and Balance Disorders (MS), University Hospital Munich, Germany; Laboratoire Gillet-Mérieux (MTV), Centre de Biologie et Pathologie Est, Hospices Civils de Lyon, Bron, France; Department of Neuropsychiatry (MW), Royal Melbourne Hospital & University of Melbourne, Australia; and Universitätsklinikum Münster (TM), Germany
| | - Miguel Pocoví
- Mayo Clinic (MCP), Rochester, MN; UCL Great Ormond Street Institute of Child Health (PC, PG), London, UK; Great Ormond Street Hospital (PG), London, UK; Département de Neurologie (MA), Hôpital de Hautepierre, CHU de Strasbourg; Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC) (MA), INSERM-U964/CNRS-UMR7104/Université de Strasbourg, Illkirch; Fédération de Médecine Translationnelle de Strasbourg (FMTS) (MA), Université de Strasbourg, France; Institute of Medical Genetics and Applied Genomics (PB), University Hospital of Tübingen; Centogene AG (PB), Rostock, Germany; Universitaire de Psychiatrie de l'Enfant et de l'Adolescent (OB), CHU de Nantes, France; Regional Coordinator Centre for Rare Diseases (AD), University Hospital Santa Maria della Misericordia, Udine, Italy; Division of Metabolism, Bambino Gesù Children's Hospital (CD-V), Rome, Italy; Klinik und Poliklinik für Psychiatrie, Psychosomatik und Psychotherapie der Universität Regensburg am Bezirksklinikum (H-HK), Regensburg, Germany; Hospices Civils de Lyon-Centre de Biologie et Pathologie Est (PL), Bron, France; University of São Paulo (HCFMRP-USP) (CML), Ribeirão Preto, SP, Brazil; Department of Medicine (DSO), Washington University, St Louis, MO; Child Development Centre (AP), Addenbrooke's Hospital, Cambridge, UK; University of Zaragoza (MP), IIS Aragon, Spain; Department of Neurology and German Center for Vertigo and Balance Disorders (MS), University Hospital Munich, Germany; Laboratoire Gillet-Mérieux (MTV), Centre de Biologie et Pathologie Est, Hospices Civils de Lyon, Bron, France; Department of Neuropsychiatry (MW), Royal Melbourne Hospital & University of Melbourne, Australia; and Universitätsklinikum Münster (TM), Germany
| | - Michael Strupp
- Mayo Clinic (MCP), Rochester, MN; UCL Great Ormond Street Institute of Child Health (PC, PG), London, UK; Great Ormond Street Hospital (PG), London, UK; Département de Neurologie (MA), Hôpital de Hautepierre, CHU de Strasbourg; Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC) (MA), INSERM-U964/CNRS-UMR7104/Université de Strasbourg, Illkirch; Fédération de Médecine Translationnelle de Strasbourg (FMTS) (MA), Université de Strasbourg, France; Institute of Medical Genetics and Applied Genomics (PB), University Hospital of Tübingen; Centogene AG (PB), Rostock, Germany; Universitaire de Psychiatrie de l'Enfant et de l'Adolescent (OB), CHU de Nantes, France; Regional Coordinator Centre for Rare Diseases (AD), University Hospital Santa Maria della Misericordia, Udine, Italy; Division of Metabolism, Bambino Gesù Children's Hospital (CD-V), Rome, Italy; Klinik und Poliklinik für Psychiatrie, Psychosomatik und Psychotherapie der Universität Regensburg am Bezirksklinikum (H-HK), Regensburg, Germany; Hospices Civils de Lyon-Centre de Biologie et Pathologie Est (PL), Bron, France; University of São Paulo (HCFMRP-USP) (CML), Ribeirão Preto, SP, Brazil; Department of Medicine (DSO), Washington University, St Louis, MO; Child Development Centre (AP), Addenbrooke's Hospital, Cambridge, UK; University of Zaragoza (MP), IIS Aragon, Spain; Department of Neurology and German Center for Vertigo and Balance Disorders (MS), University Hospital Munich, Germany; Laboratoire Gillet-Mérieux (MTV), Centre de Biologie et Pathologie Est, Hospices Civils de Lyon, Bron, France; Department of Neuropsychiatry (MW), Royal Melbourne Hospital & University of Melbourne, Australia; and Universitätsklinikum Münster (TM), Germany
| | - Marie T Vanier
- Mayo Clinic (MCP), Rochester, MN; UCL Great Ormond Street Institute of Child Health (PC, PG), London, UK; Great Ormond Street Hospital (PG), London, UK; Département de Neurologie (MA), Hôpital de Hautepierre, CHU de Strasbourg; Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC) (MA), INSERM-U964/CNRS-UMR7104/Université de Strasbourg, Illkirch; Fédération de Médecine Translationnelle de Strasbourg (FMTS) (MA), Université de Strasbourg, France; Institute of Medical Genetics and Applied Genomics (PB), University Hospital of Tübingen; Centogene AG (PB), Rostock, Germany; Universitaire de Psychiatrie de l'Enfant et de l'Adolescent (OB), CHU de Nantes, France; Regional Coordinator Centre for Rare Diseases (AD), University Hospital Santa Maria della Misericordia, Udine, Italy; Division of Metabolism, Bambino Gesù Children's Hospital (CD-V), Rome, Italy; Klinik und Poliklinik für Psychiatrie, Psychosomatik und Psychotherapie der Universität Regensburg am Bezirksklinikum (H-HK), Regensburg, Germany; Hospices Civils de Lyon-Centre de Biologie et Pathologie Est (PL), Bron, France; University of São Paulo (HCFMRP-USP) (CML), Ribeirão Preto, SP, Brazil; Department of Medicine (DSO), Washington University, St Louis, MO; Child Development Centre (AP), Addenbrooke's Hospital, Cambridge, UK; University of Zaragoza (MP), IIS Aragon, Spain; Department of Neurology and German Center for Vertigo and Balance Disorders (MS), University Hospital Munich, Germany; Laboratoire Gillet-Mérieux (MTV), Centre de Biologie et Pathologie Est, Hospices Civils de Lyon, Bron, France; Department of Neuropsychiatry (MW), Royal Melbourne Hospital & University of Melbourne, Australia; and Universitätsklinikum Münster (TM), Germany
| | - Mark Walterfang
- Mayo Clinic (MCP), Rochester, MN; UCL Great Ormond Street Institute of Child Health (PC, PG), London, UK; Great Ormond Street Hospital (PG), London, UK; Département de Neurologie (MA), Hôpital de Hautepierre, CHU de Strasbourg; Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC) (MA), INSERM-U964/CNRS-UMR7104/Université de Strasbourg, Illkirch; Fédération de Médecine Translationnelle de Strasbourg (FMTS) (MA), Université de Strasbourg, France; Institute of Medical Genetics and Applied Genomics (PB), University Hospital of Tübingen; Centogene AG (PB), Rostock, Germany; Universitaire de Psychiatrie de l'Enfant et de l'Adolescent (OB), CHU de Nantes, France; Regional Coordinator Centre for Rare Diseases (AD), University Hospital Santa Maria della Misericordia, Udine, Italy; Division of Metabolism, Bambino Gesù Children's Hospital (CD-V), Rome, Italy; Klinik und Poliklinik für Psychiatrie, Psychosomatik und Psychotherapie der Universität Regensburg am Bezirksklinikum (H-HK), Regensburg, Germany; Hospices Civils de Lyon-Centre de Biologie et Pathologie Est (PL), Bron, France; University of São Paulo (HCFMRP-USP) (CML), Ribeirão Preto, SP, Brazil; Department of Medicine (DSO), Washington University, St Louis, MO; Child Development Centre (AP), Addenbrooke's Hospital, Cambridge, UK; University of Zaragoza (MP), IIS Aragon, Spain; Department of Neurology and German Center for Vertigo and Balance Disorders (MS), University Hospital Munich, Germany; Laboratoire Gillet-Mérieux (MTV), Centre de Biologie et Pathologie Est, Hospices Civils de Lyon, Bron, France; Department of Neuropsychiatry (MW), Royal Melbourne Hospital & University of Melbourne, Australia; and Universitätsklinikum Münster (TM), Germany
| | - Thorsten Marquardt
- Mayo Clinic (MCP), Rochester, MN; UCL Great Ormond Street Institute of Child Health (PC, PG), London, UK; Great Ormond Street Hospital (PG), London, UK; Département de Neurologie (MA), Hôpital de Hautepierre, CHU de Strasbourg; Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC) (MA), INSERM-U964/CNRS-UMR7104/Université de Strasbourg, Illkirch; Fédération de Médecine Translationnelle de Strasbourg (FMTS) (MA), Université de Strasbourg, France; Institute of Medical Genetics and Applied Genomics (PB), University Hospital of Tübingen; Centogene AG (PB), Rostock, Germany; Universitaire de Psychiatrie de l'Enfant et de l'Adolescent (OB), CHU de Nantes, France; Regional Coordinator Centre for Rare Diseases (AD), University Hospital Santa Maria della Misericordia, Udine, Italy; Division of Metabolism, Bambino Gesù Children's Hospital (CD-V), Rome, Italy; Klinik und Poliklinik für Psychiatrie, Psychosomatik und Psychotherapie der Universität Regensburg am Bezirksklinikum (H-HK), Regensburg, Germany; Hospices Civils de Lyon-Centre de Biologie et Pathologie Est (PL), Bron, France; University of São Paulo (HCFMRP-USP) (CML), Ribeirão Preto, SP, Brazil; Department of Medicine (DSO), Washington University, St Louis, MO; Child Development Centre (AP), Addenbrooke's Hospital, Cambridge, UK; University of Zaragoza (MP), IIS Aragon, Spain; Department of Neurology and German Center for Vertigo and Balance Disorders (MS), University Hospital Munich, Germany; Laboratoire Gillet-Mérieux (MTV), Centre de Biologie et Pathologie Est, Hospices Civils de Lyon, Bron, France; Department of Neuropsychiatry (MW), Royal Melbourne Hospital & University of Melbourne, Australia; and Universitätsklinikum Münster (TM), Germany
| |
Collapse
|
47
|
Argan Oil-Mediated Attenuation of Organelle Dysfunction, Oxidative Stress and Cell Death Induced by 7-Ketocholesterol in Murine Oligodendrocytes 158N. Int J Mol Sci 2017; 18:ijms18102220. [PMID: 29065513 PMCID: PMC5666899 DOI: 10.3390/ijms18102220] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2017] [Revised: 10/18/2017] [Accepted: 10/20/2017] [Indexed: 02/07/2023] Open
Abstract
Argan oil is widely used in Morocco in traditional medicine. Its ability to treat cardiovascular diseases is well-established. However, nothing is known about its effects on neurodegenerative diseases, which are often associated with increased oxidative stress leading to lipid peroxidation and the formation of 7-ketocholesterol (7KC) resulting from cholesterol auto-oxidation. As 7KC induces oxidative stress, inflammation and cell death, it is important to identify compounds able to impair its harmful effects. These compounds may be either natural or synthetic molecules or mixtures of molecules such as oils. In this context: (i) the lipid profiles of dietary argan oils from Berkane and Agadir (Morocco) in fatty acids, phytosterols, tocopherols and polyphenols were determined by different chromatographic techniques; and (ii) their anti-oxidant and cytoprotective effects in 158N murine oligodendrocytes cultured with 7KC (25-50 µM; 24 h) without and with argan oil (0.1% v/v) or α-tocopherol (400 µM, positive control) were evaluated with complementary techniques of cellular and molecular biology. Among the unsaturated fatty acids present in argan oils, oleate (C18:1 n-9) and linoleate (C18:1 n-6) were the most abundant; the highest quantities of saturated fatty acids were palmitate (C16:0) and stearate (C18:0). Several phytosterols were found, mainly schottenol and spinasterol (specific to argan oil), cycloartenol, β-amyrin and citrostadienol. α- and γ-tocopherols were also present. Tyrosol and protocatechic acid were the only polyphenols detected. Argan and extra virgin olive oils have many compounds in common, principally oleate and linoleate, and tocopherols. Kit Radicaux Libres (KRL) and ferric reducing antioxidant power (FRAP) tests showed that argan and extra virgin olive oils have anti-oxidant properties. Argan oils were able to attenuate the cytotoxic effects of 7KC on 158N cells: loss of cell adhesion, cell growth inhibition, increased plasma membrane permeability, mitochondrial, peroxisomal and lysosomal dysfunction, and the induction of oxiapoptophagy (OXIdation + APOPTOsis + autoPHAGY). Altogether, our data obtained in 158N oligodendrocytes provide evidence that argan oil is able to counteract the toxic effects of 7KC on nerve cells, thus suggesting that some of its compounds could prevent or mitigate neurodegenerative diseases to the extent that they are able to cross the blood-brain barrier.
Collapse
|
48
|
Bezine M, Debbabi M, Nury T, Ben-Khalifa R, Samadi M, Cherkaoui-Malki M, Vejux A, Raas Q, de Sèze J, Moreau T, El-Ayeb M, Lizard G. Evidence of K+ homeostasis disruption in cellular dysfunction triggered by 7-ketocholesterol, 24S-hydroxycholesterol, and tetracosanoic acid (C24:0) in 158N murine oligodendrocytes. Chem Phys Lipids 2017; 207:135-150. [DOI: 10.1016/j.chemphyslip.2017.03.006] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2017] [Revised: 03/06/2017] [Accepted: 03/07/2017] [Indexed: 12/12/2022]
|
49
|
Kloudova A, Guengerich FP, Soucek P. The Role of Oxysterols in Human Cancer. Trends Endocrinol Metab 2017; 28:485-496. [PMID: 28410994 PMCID: PMC5474130 DOI: 10.1016/j.tem.2017.03.002] [Citation(s) in RCA: 141] [Impact Index Per Article: 17.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/06/2017] [Accepted: 03/20/2017] [Indexed: 12/12/2022]
Abstract
Oxysterols are oxygenated derivatives of cholesterol formed in the human body or ingested in the diet. By modulating the activity of many proteins [e.g., liver X receptors (LXRs), oxysterol-binding proteins (OSBPs), some ATP-binding cassette (ABC) transporters], oxysterols can affect many cellular functions and influence various physiological processes (e.g., cholesterol metabolism, membrane fluidity regulation, intracellular signaling pathways). Therefore, the role of oxysterols is also important in pathological conditions (e.g., atherosclerosis, diabetes mellitus type 2, neurodegenerative disorders). Finally, current evidence suggests that oxysterols play a role in malignancies such as breast, prostate, colon, and bile duct cancer. This review summarizes the physiological importance of oxysterols in the human body with a special emphasis on their roles in various tumors.
Collapse
Affiliation(s)
- Alzbeta Kloudova
- Department of Toxicogenomics, National Institute of Public Health, Prague 100 42, Czech Republic; Third Faculty of Medicine, Charles University, Prague 100 00, Czech Republic
| | - F Peter Guengerich
- Department of Biochemistry, Vanderbilt University School of Medicine, Nashville, TN 37232, USA
| | - Pavel Soucek
- Biomedical Center, Faculty of Medicine in Pilsen, Charles University, Pilsen 323 00, Czech Republic.
| |
Collapse
|
50
|
Improvement of 5,6α-epoxycholesterol, 5,6β-epoxycholesterol, cholestane-3β,5α,6β-triol and 6-oxo-cholestan-3β,5α-diol recovery for quantification by GC/MS. Chem Phys Lipids 2017; 207:92-98. [PMID: 28554594 DOI: 10.1016/j.chemphyslip.2017.05.006] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2017] [Revised: 04/21/2017] [Accepted: 05/23/2017] [Indexed: 01/19/2023]
Abstract
5,6α-epoxycholesterol (5,6α-EC) and 5,6β-epoxycholesterol (5,6β-EC) are oxysterols involved in the anticancer pharmacology of the widely used antitumor drug tamoxifen. They are both metabolized into cholestane-3β,5α,6β-triol (CT) by the cholesterol-5,6-epoxide hydrolase (ChEH) enzyme, and CT is metabolized by an as-yet uncharacterized enzyme into 6-oxo-cholestan-3β,5α-diol (OCDO). A recent feasibility study showed that the 5,6-ECs may represent surrogate markers of tamoxifen activity in breast cancer patients undergoing endocrine therapy, thus there is a growing interest in their accurate quantification. These oxysterols are usually quantified by gas-liquid chromatography coupled to mass spectrometry (GC/MS), using an isotope dilution methodology with the corresponding deuterated oxysterol. This method is considered to be relative quantitative since all of the standards used are deuterated oxysterols, however it is not known whether the preparation of each oxysterol is affected in the same way by the extraction, pre-purification by solid phase extraction (SPE) and trimethylsilylation steps, particularly when using biological samples that contain many other reactive compounds. Thus, in this study we investigated the yield of the 5,6-ECs, CT and OCDO recovery from patient serum samples at different stages of their work-up and trimethylsilylation prior to GC/MS analysis, using [14C]-labeled analogs to follow these oxysterols at each step. We measured a 40 to 60% loss of material for the 5,6-ECs and OCDO, however we also describe the conditions that improved their recovery. Our data also show that the use of deuterated 5,6α-EC, 5,6β-EC, CT and OCDO is an absolute requirement for their accurate quantification.
Collapse
|