51
|
Wang X, Li Y, Xia X, Zhang M, Ge C, Xia X, Xiao H, Xu S. Mutagenicity of 7-ketocholesterol in CHO cells: The role of lipid peroxidation. Toxicology 2020; 446:152587. [PMID: 33017620 DOI: 10.1016/j.tox.2020.152587] [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: 06/23/2020] [Revised: 09/10/2020] [Accepted: 09/14/2020] [Indexed: 02/07/2023]
Abstract
As an important cholesterol oxide, 7-ketocholesterol plays a deleterious role in the occurrence of cancer. Although the fact had been proved that 7-ketocholesterol could induce several biological phenomena, including apoptosis, DNA damage, et al., this issue whether 7-ketocholesterol led to mutagenesis in mammalian cells remains largely unexplored. Here, we investigated the major role of lipid peroxidation in the genotoxic response to 7-ketocholesterol in chinese hamster ovary (CHO) cells. The results showed that 7-ketocholesterol induced gene mutation and DNA double-strand breaks (DSBs) in concentration- and time-dependent manner. After CHO cells were treated with 25 μM 7-ketocholesterol for 48 h, the mutation frequency at hprt gene loci and the level of γ-H2AX protein were both significantly increased. Exposure to 7-ketocholesterol resulted in a concentration-dependent increase in the apoptotic rate and the protein expression of cleaved caspase-3 and -7 in CHO cells. Moreover, a significant increase of superoxide dismutase (SOD) activity and content of malondialdehyde (MDA) was also observed. Using a inhibitor of lipid peroxidation (butylated hydroxytoluene), it was found to remarkably inhibit the genotoxicity and MDA levels caused by 7-ketocholesterol. These findings indicated that lipid peroxidation was involved in the mutagenic process of 7-ketocholesterol in CHO cells.
Collapse
Affiliation(s)
- Xiaofei Wang
- School of Biology, Food and Environment, Hefei University, Hefei, 230601, PR China
| | - Yintao Li
- School of Biology, Food and Environment, Hefei University, Hefei, 230601, PR China; Anhui Province Key Laboratory of Environmental Toxicology and Pollution Control Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, 230031, PR China
| | - Xuanyi Xia
- School of Biology, Food and Environment, Hefei University, Hefei, 230601, PR China
| | - Min Zhang
- School of Biology, Food and Environment, Hefei University, Hefei, 230601, PR China
| | - Chunmei Ge
- School of Biology, Food and Environment, Hefei University, Hefei, 230601, PR China
| | - Xiaoxiao Xia
- School of Biology, Food and Environment, Hefei University, Hefei, 230601, PR China
| | - Hourong Xiao
- School of Biology, Food and Environment, Hefei University, Hefei, 230601, PR China.
| | - Shengmin Xu
- Institute of Physical Science and Information Technology, Anhui University, Hefei, 230601, PR China.
| |
Collapse
|
52
|
De Lira MN, Raman SJ, Schulze A, Schneider-Schaulies S, Avota E. Neutral Sphingomyelinase-2 (NSM 2) Controls T Cell Metabolic Homeostasis and Reprogramming During Activation. Front Mol Biosci 2020; 7:217. [PMID: 33088808 PMCID: PMC7498697 DOI: 10.3389/fmolb.2020.00217] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2020] [Accepted: 08/04/2020] [Indexed: 12/15/2022] Open
Abstract
Neutral sphingomyelinase-2 (NSM2) is a member of a superfamily of enzymes responsible for conversion of sphingomyelin into phosphocholine and ceramide at the cytosolic leaflet of the plasma membrane. Upon specific ablation of NSM2, T cells proved to be hyper-responsive to CD3/CD28 co-stimulation, indicating that the enzyme acts to dampen early overshooting activation of these cells. It remained unclear whether hyper-reactivity of NSM2-deficient T cells is supported by a deregulated metabolic activity in these cells. Here, we demonstrate that ablation of NSM2 activity affects metabolism of the quiescent CD4+ T cells which accumulate ATP in mitochondria and increase basal glycolytic activity. This supports enhanced production of total ATP and metabolic switch early after TCR/CD28 stimulation. Most interestingly, increased metabolic activity in resting NSM2-deficient T cells does not support sustained response upon stimulation. While elevated under steady-state conditions in NSM2-deficient CD4+ T cells, the mTORC1 pathway regulating mitochondria size, oxidative phosphorylation, and ATP production is impaired after 24 h of stimulation. Taken together, the absence of NSM2 promotes a hyperactive metabolic state in unstimulated CD4+ T cells yet fails to support sustained T cell responses upon antigenic stimulation.
Collapse
Affiliation(s)
| | | | - Almut Schulze
- Division of Tumor Metabolism and Microenvironment, German Cancer Research Center, Heidelberg, Germany
| | | | - Elita Avota
- Institute for Virology and Immunobiology, University of Würzburg, Würzburg, Germany
| |
Collapse
|
53
|
Iwahori A, Maekawa M, Narita A, Kato A, Sato T, Ogura J, Sato Y, Kikuchi M, Noguchi A, Higaki K, Okuyama T, Takahashi T, Eto Y, Mano N. Development of a Diagnostic Screening Strategy for Niemann-Pick Diseases Based on Simultaneous Liquid Chromatography-Tandem Mass Spectrometry Analyses of N-Palmitoyl-O-phosphocholine-serine and Sphingosylphosphorylcholine. Biol Pharm Bull 2020; 43:1398-1406. [PMID: 32581190 DOI: 10.1248/bpb.b20-00400] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Early diagnosis of Niemann-Pick diseases (NPDs) is important for better prognosis of such diseases. N-Palmitoyl-O-phosphocholine-serine (PPCS) is a new NPD biomarker possessing high sensitivity, and with its combination with sphingosylphosphocholine (SPC) it may be possible to distinguish NPD-C from NPD-A/B. In this study, a rapid liquid chromatography-tandem mass spectrometry (LC-MS/MS) method (method 1) and a validated LC-MS/MS analysis (method 2) of PPCS and SPC were developed, and we have proposed a diagnostic screening strategy for NPDs using a combination of serum PPCS and SPC concentrations. Nexera and API 5000 were used as LC-MS/MS systems. C18 columns with lengths of 10 and 50 mm were used for method 1 and 2, respectively. 2H3-Labeled PPCS and nor-SPC were used as internal standards. Selective reaction monitoring in positive-ion mode was used for MS/MS. Run times of 1.2 and 8 min were set for methods 1 and 2, respectively. In both methods 1 and 2, two analytes showed high linearity in the range of 1-4000 ng/mL. Method 2 provided high accuracy and precision in method validation. Serum concentrations of both analytes were significantly higher in NPD-C patients than those of healthy subjects in both methods. Serum PPCS correlated between methods 1 and 2; however, it was different in the case of SPC. The serum PPCS/SPC ratio was different in healthy subjects, NPD-C, and NPD-A/B. These results suggest that using a combination of the two LC-MS/MS analytical methods for PPCS and SPC is useful for diagnostic screening of NPDs.
Collapse
Affiliation(s)
- Anna Iwahori
- Faculty of Pharmaceutical Sciences, Tohoku University
| | - Masamitsu Maekawa
- Faculty of Pharmaceutical Sciences, Tohoku University.,Department of Pharmaceutical Sciences, Tohoku University Hospital
| | - Aya Narita
- Division of Child Neurology, Tottori University Hospital
| | - Akie Kato
- Department of Pediatrics, Akita University Graduate School of Medicine
| | - Toshihiro Sato
- Department of Pharmaceutical Sciences, Tohoku University Hospital
| | - Jiro Ogura
- Department of Pharmaceutical Sciences, Tohoku University Hospital
| | - Yu Sato
- Department of Pharmaceutical Sciences, Tohoku University Hospital
| | - Masafumi Kikuchi
- Faculty of Pharmaceutical Sciences, Tohoku University.,Department of Pharmaceutical Sciences, Tohoku University Hospital
| | - Atsuko Noguchi
- Department of Pediatrics, Akita University Graduate School of Medicine
| | - Katsumi Higaki
- Division of Functional Genomics, Research Centre for Bioscience and Technology, Faculty of Medicine, Tottori University
| | - Torayuki Okuyama
- Department of Clinical Laboratory Medicine, National Center for Child Health and Development
| | - Tsutomu Takahashi
- Department of Pediatrics, Akita University Graduate School of Medicine
| | - Yoshikatsu Eto
- Advanced Clinical Research Center, Institute for Neurological Disorders
| | - Nariyasu Mano
- Faculty of Pharmaceutical Sciences, Tohoku University.,Department of Pharmaceutical Sciences, Tohoku University Hospital
| |
Collapse
|
54
|
de Medina P, Diallo K, Huc-Claustre E, Attia M, Soulès R, Silvente-Poirot S, Poirot M. The 5,6-epoxycholesterol metabolic pathway in breast cancer: Emergence of new pharmacological targets. Br J Pharmacol 2020; 178:3248-3260. [PMID: 32696532 DOI: 10.1111/bph.15205] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2020] [Revised: 07/06/2020] [Accepted: 07/10/2020] [Indexed: 12/12/2022] Open
Abstract
Metabolic pathways have emerged as cornerstones in carcinogenic deregulation providing new therapeutic strategies for cancer management. Recently, a new branch of cholesterol metabolism has been discovered involving the biochemical transformation of 5,6-epoxycholesterols (5,6-ECs). The 5,6-ECs are metabolized in breast cancers to the tumour promoter oncosterone whereas, in normal breast tissue, they are metabolized to the tumour suppressor metabolite, dendrogenin A (DDA). Blocking the mitogenic and invasive potential of oncosterone will present new opportunities for breast cancer treatment. The reactivation of DDA biosynthesis, or its use as a drug, represents promising therapeutic approaches such as DDA-deficiency complementation, activation of breast cancer cell re-differentiation and breast cancer chemoprevention. This review presents current knowledge of the 5,6-EC metabolic pathway in breast cancer, focusing on the 5,6-EC metabolic enzymes ChEH and HSD11B2 and on 5,6-EC metabolite targets, the oxysterol receptor (LXRβ) and the glucocorticoid receptor. LINKED ARTICLES: This article is part of a themed issue on Oxysterols, Lifelong Health and Therapeutics. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v178.16/issuetoc.
Collapse
Affiliation(s)
- Philippe de Medina
- UMR-1037, Cancer Research Center of Toulouse (CRCT), Team "Cholesterol Metabolism and Therapeutic Innovations"; Equipe labellisée par la Ligue Nationale Contre le Cancer, The French Network for Nutrition and Cancer Research (NACRe Network), INSERM-Université de Toulouse, Toulouse, France
| | - Khadijetou Diallo
- UMR-1037, Cancer Research Center of Toulouse (CRCT), Team "Cholesterol Metabolism and Therapeutic Innovations"; Equipe labellisée par la Ligue Nationale Contre le Cancer, The French Network for Nutrition and Cancer Research (NACRe Network), INSERM-Université de Toulouse, Toulouse, France
| | - Emilie Huc-Claustre
- UMR-1037, Cancer Research Center of Toulouse (CRCT), Team "Cholesterol Metabolism and Therapeutic Innovations"; Equipe labellisée par la Ligue Nationale Contre le Cancer, The French Network for Nutrition and Cancer Research (NACRe Network), INSERM-Université de Toulouse, Toulouse, France
| | - Mehdi Attia
- UMR-1037, Cancer Research Center of Toulouse (CRCT), Team "Cholesterol Metabolism and Therapeutic Innovations"; Equipe labellisée par la Ligue Nationale Contre le Cancer, The French Network for Nutrition and Cancer Research (NACRe Network), INSERM-Université de Toulouse, Toulouse, France
| | - Régis Soulès
- UMR-1037, Cancer Research Center of Toulouse (CRCT), Team "Cholesterol Metabolism and Therapeutic Innovations"; Equipe labellisée par la Ligue Nationale Contre le Cancer, The French Network for Nutrition and Cancer Research (NACRe Network), INSERM-Université de Toulouse, Toulouse, France
| | - Sandrine Silvente-Poirot
- UMR-1037, Cancer Research Center of Toulouse (CRCT), Team "Cholesterol Metabolism and Therapeutic Innovations"; Equipe labellisée par la Ligue Nationale Contre le Cancer, The French Network for Nutrition and Cancer Research (NACRe Network), INSERM-Université de Toulouse, Toulouse, France
| | - Marc Poirot
- UMR-1037, Cancer Research Center of Toulouse (CRCT), Team "Cholesterol Metabolism and Therapeutic Innovations"; Equipe labellisée par la Ligue Nationale Contre le Cancer, The French Network for Nutrition and Cancer Research (NACRe Network), INSERM-Université de Toulouse, Toulouse, France
| |
Collapse
|
55
|
High diagnostic value of plasma Niemann-Pick type C biomarkers in adults with selected neurological and/or psychiatric disorders. J Neurol 2020; 267:3371-3377. [PMID: 32592146 DOI: 10.1007/s00415-020-10020-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2020] [Revised: 06/20/2020] [Accepted: 06/22/2020] [Indexed: 01/02/2023]
Abstract
Late-onset Niemann-Pick type C (NP-C) is a rare, underdiagnosed lysosomal disease with neurological manifestations. A specific treatment, miglustat, can stabilize the disease if given early. Recently, three plasma screening biomarkers (PSBs) were developed [cholestane3β,5α,6βtriol (C-triol), 7-ketocholesterol (7-KC), and lysosphingomyelin-509 (LSM-509)], allowing a simpler and quite robust screening of patients suitable for genetic testing. The objective of our study was to evaluate practical utility and feasibility of large-scale PSB screening for NP-C in selected adult patients. Patients were prospectively enrolled if they showed, starting from 12 years of age, at least one of the three initial neuro-psychiatric manifestations described in NP-C: (1) gait disorder (cerebellar and/or dystonic); (2) cognitive decline with frontal lobe syndrome; (3) atypical psychosis. PSBs were measured in plasma of all patients and, if positive (LSM-509 and/or C-triol + 7-KC elevated), sequencing of NPC1 and NPC2 genes was performed. A total of 251 patients [136 males, 115 females; median age 42.1 (range 12.2-85.6) years] were screened. Six patients had positive PSBs. Two were confirmed to have NP-C (0.8% diagnostic yield, both with all three PSBs highly increased, especially LSM-509). False-positive rate was 1.2%, which was identical if only considering LSM-509. By contrast, false-positive rates were 8.1% and 5.7% for 7-KC and C-triol, respectively. We showed that selecting patients with neurologic and/or psychiatric symptoms consistent with NP-C for large-scale PSB screening is a simple and valid strategy to identify new adult NP-C patients, and would probably lead to earlier diagnosis and treatment administration if widely applied.
Collapse
|
56
|
Yañez MJ, Marín T, Balboa E, Klein AD, Alvarez AR, Zanlungo S. Finding pathogenic commonalities between Niemann-Pick type C and other lysosomal storage disorders: Opportunities for shared therapeutic interventions. Biochim Biophys Acta Mol Basis Dis 2020; 1866:165875. [PMID: 32522631 DOI: 10.1016/j.bbadis.2020.165875] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2020] [Revised: 05/06/2020] [Accepted: 06/03/2020] [Indexed: 12/12/2022]
Abstract
Lysosomal storage disorders (LSDs) are diseases characterized by the accumulation of macromolecules in the late endocytic system and are caused by inherited defects in genes that encode mainly lysosomal enzymes or transmembrane lysosomal proteins. Niemann-Pick type C disease (NPCD), a LSD characterized by liver damage and progressive neurodegeneration that leads to early death, is caused by mutations in the genes encoding the NPC1 or NPC2 proteins. Both proteins are involved in the transport of cholesterol from the late endosomal compartment to the rest of the cell. Loss of function of these proteins causes primary cholesterol accumulation, and secondary accumulation of other lipids, such as sphingolipids, in lysosomes. Despite years of studying the genetic and molecular bases of NPCD and related-lysosomal disorders, the pathogenic mechanisms involved in these diseases are not fully understood. In this review we will summarize the pathogenic mechanisms described for NPCD and we will discuss their relevance for other LSDs with neurological components such as Niemann- Pick type A and Gaucher diseases. We will particularly focus on the activation of signaling pathways that may be common to these three pathologies with emphasis on how the intra-lysosomal accumulation of lipids leads to pathology, specifically to neurological impairments. We will show that although the primary lipid storage defect is different in these three LSDs, there is a similar secondary accumulation of metabolites and activation of signaling pathways that can lead to common pathogenic mechanisms. This analysis might help to delineate common pathological mechanisms and therapeutic targets for lysosomal storage diseases.
Collapse
Affiliation(s)
- M J Yañez
- Department of Gastroenterology, Faculty of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - T Marín
- Department of Gastroenterology, Faculty of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - E Balboa
- Department of Gastroenterology, Faculty of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - A D Klein
- Centro de Genética y Genómica, Facultad de Medicina, Clínica Alemana Universidad del Desarrollo, Santiago, Chile
| | - A R Alvarez
- Laboratory of Cell Signaling, Department of Cellular and Molecular Biology, Biological Sciences Faculty, Pontificia Universidad Católica de Chile, Santiago, Chile; CARE UC, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - S Zanlungo
- Department of Gastroenterology, Faculty of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile.
| |
Collapse
|
57
|
Magro Dos Reis I, Houben T, Oligschläger Y, Bücken L, Steinbusch H, Cassiman D, Lütjohann D, Westerterp M, Prickaerts J, Plat J, Shiri-Sverdlov R. Dietary plant stanol ester supplementation reduces peripheral symptoms in a mouse model of Niemann-Pick type C1 disease. J Lipid Res 2020; 61:830-839. [PMID: 32291331 PMCID: PMC7269767 DOI: 10.1194/jlr.ra120000632] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2020] [Revised: 03/23/2020] [Indexed: 11/20/2022] Open
Abstract
Niemann-Pick type C (NPC)1 disease is a rare genetic condition in which the function of the lysosomal cholesterol transporter NPC1 protein is impaired. Consequently, sphingolipids and cholesterol accumulate in lysosomes of all tissues, triggering a cascade of pathological events that culminate in severe systemic and neurological symptoms. Lysosomal cholesterol accumulation is also a key factor in the development of atherosclerosis and NASH. In these two metabolic diseases, the administration of plant stanol esters has been shown to ameliorate cellular cholesterol accumulation and inflammation. Given the overlap of pathological mechanisms among atherosclerosis, NASH, and NPC1 disease, we sought to investigate whether dietary supplementation with plant stanol esters improves the peripheral features of NPC1 disease. To this end, we used an NPC1 murine model featuring a Npc1-null allele (Npc1nih ), creating a dysfunctional NPC1 protein. Npc1nih mice were fed a 2% or 6% plant stanol ester-enriched diet over the course of 5 weeks. During this period, hepatic and blood lipid and inflammatory profiles were assessed. Npc1nih mice fed the plant stanol-enriched diet exhibited lower hepatic cholesterol accumulation, damage, and inflammation than regular chow-fed Npc1nih mice. Moreover, plant stanol consumption shifted circulating T-cells and monocytes in particular toward an anti-inflammatory profile. Overall, these effects were stronger following dietary supplementation with 6% stanols, suggesting a dose-dependent effect. The findings of our study highlight the potential use of plant stanols as an affordable complementary means to ameliorate disorders in hepatic and blood lipid metabolism and reduce inflammation in NPC1 disease.
Collapse
Affiliation(s)
- Inês Magro Dos Reis
- Department of Molecular Genetics, School of Nutrition and Translational Research in Metabolism (NUTRIM), Maastricht University, Maastricht, The Netherlands
| | - Tom Houben
- Department of Molecular Genetics, School of Nutrition and Translational Research in Metabolism (NUTRIM), Maastricht University, Maastricht, The Netherlands
| | - Yvonne Oligschläger
- Department of Molecular Genetics, School of Nutrition and Translational Research in Metabolism (NUTRIM), Maastricht University, Maastricht, The Netherlands
| | - Leoni Bücken
- Department of Molecular Genetics, School of Nutrition and Translational Research in Metabolism (NUTRIM), Maastricht University, Maastricht, The Netherlands
| | - Hellen Steinbusch
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience, Maastricht University, Maastricht, The Netherlands
| | - David Cassiman
- Liver Research Unit University of Leuven, Leuven, Belgium; Department of Gastroenterology-Hepatology and Metabolic Center, University Hospitals Leuven, Leuven, Belgium
| | - Dieter Lütjohann
- Institute of Clinical Chemistry and Clinical Pharmacology, Medical Faculty, University of Bonn, Bonn, Germany
| | - Marit Westerterp
- Department of Pediatrics, Section Molecular Genetics, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Jos Prickaerts
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience, Maastricht University, Maastricht, The Netherlands
| | - Jogchum Plat
- Department of Nutrition and Movement Sciences, School for Nutrition, Toxicology, and Metabolism, Maastricht University, Maastricht, The Netherlands
| | - Ronit Shiri-Sverdlov
- Department of Molecular Genetics, School of Nutrition and Translational Research in Metabolism (NUTRIM), Maastricht University, Maastricht, The Netherlands. mailto:
| |
Collapse
|
58
|
Ballout RA, Sviridov D, Bukrinsky MI, Remaley AT. The lysosome: A potential juncture between SARS-CoV-2 infectivity and Niemann-Pick disease type C, with therapeutic implications. FASEB J 2020; 34:7253-7264. [PMID: 32367579 PMCID: PMC7383733 DOI: 10.1096/fj.202000654r] [Citation(s) in RCA: 75] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2020] [Revised: 04/21/2020] [Accepted: 04/21/2020] [Indexed: 02/06/2023]
Abstract
Drug repurposing is potentially the fastest available option in the race to identify safe and efficacious drugs that can be used to prevent and/or treat COVID‐19. By describing the life cycle of the newly emergent coronavirus, SARS‐CoV‐2, in light of emerging data on the therapeutic efficacy of various repurposed antimicrobials undergoing testing against the virus, we highlight in this review a possible mechanistic convergence between some of these tested compounds. Specifically, we propose that the lysosomotropic effects of hydroxychloroquine and several other drugs undergoing testing may be responsible for their demonstrated in vitro antiviral activities against COVID‐19. Moreover, we propose that Niemann‐Pick disease type C (NPC), a lysosomal storage disorder, may provide new insights into potential future therapeutic targets for SARS‐CoV‐2, by highlighting key established features of the disorder that together result in an “unfavorable” host cellular environment that may interfere with viral propagation. Our reasoning evolves from previous biochemical and cell biology findings related to NPC, coupled with the rapidly evolving data on COVID‐19. Our overall aim is to suggest that pharmacological interventions targeting lysosomal function in general, and those particularly capable of reversibly inducing transient NPC‐like cellular and biochemical phenotypes, constitute plausible mechanisms that could be used to therapeutically target COVID‐19.
Collapse
Affiliation(s)
- Rami A Ballout
- Lipoprotein Metabolism Section, Translational Vascular Medicine Branch, National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Dmitri Sviridov
- Lipoproteins and Atherosclerosis Laboratory, Baker Heart and Diabetes Institute, Melbourne, VIC, Australia
| | - Michael I Bukrinsky
- Department of Microbiology, Immunology and Tropical Medicine, School of Medicine and Health Sciences, The George Washington University, Washington, DC, USA
| | - Alan T Remaley
- Lipoprotein Metabolism Section, Translational Vascular Medicine Branch, National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
59
|
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: 2.3] [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
|
60
|
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: 0.8] [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
|
61
|
Metabolomic Studies of Lipid Storage Disorders, with Special Reference to Niemann-Pick Type C Disease: A Critical Review with Future Perspectives. Int J Mol Sci 2020; 21:ijms21072533. [PMID: 32260582 PMCID: PMC7178094 DOI: 10.3390/ijms21072533] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2020] [Revised: 03/27/2020] [Accepted: 04/01/2020] [Indexed: 01/18/2023] Open
Abstract
Lysosomal storage disorders (LSDs) are predominantly very rare recessive autosomal neurodegenerative diseases.Sphingolipidoses, a sub-group of LSDs, result from defects in lysosomal enzymes involved in sphingolipid catabolism, and feature disrupted storage systems which trigger complex pathogenic cascades with other organelles collaterally affected. This process leads to cell dysfunction and death, particularly in the central nervous system. One valuable approach to gaining insights into the global impact of lysosomal dysfunction is through metabolomics, which represents a discovery tool for investigating disease-induced modifications in the patterns of large numbers of simultaneously-analysed metabolites, which also features the identification of biomarkers Here, the scope and applications of metabolomics strategies to the investigation of sphingolipidoses is explored in order to facilitate our understanding of the biomolecular basis of these conditions. This review therefore surveys the benefits of applying ’state-of-the-art’ metabolomics strategies, both univariate and multivariate, to sphingolipidoses, particularly Niemann-Pick type C disease. Relevant limitations of these techniques are also discussed, along with the latest advances and developments. We conclude that metabolomics strategies are highly valuable, distinctive bioanalytical techniques for probing LSDs, most especially for the detection and validation of potential biomarkers. They also show much promise for monitoring disease progression and the evaluation of therapeutic strategies and targets.
Collapse
|
62
|
Sidhu R, Kell P, Dietzen DJ, Farhat NY, Do AND, Porter FD, Berry-Kravis E, Vite CH, Reunert J, Marquardt T, Giugliani R, Lourenço CM, Bodamer O, Wang RY, Plummer E, Schaffer JE, Ory DS, Jiang X. Application of N-palmitoyl-O-phosphocholineserine for diagnosis and assessment of response to treatment in Niemann-Pick type C disease. Mol Genet Metab 2020; 129:292-302. [PMID: 32033912 PMCID: PMC7145728 DOI: 10.1016/j.ymgme.2020.01.007] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/15/2019] [Revised: 01/09/2020] [Accepted: 01/13/2020] [Indexed: 12/12/2022]
Abstract
Niemann-Pick type C (NPC) disease is a rare lysosomal storage disorder caused by mutations in either the NPC1 or the NPC2 gene. A new class of lipids, N-acyl-O-phosphocholineserines were recently identified as NPC biomarkers. The most abundant species in this class of lipid, N-palmitoyl-O-phosphocholineserine (PPCS), was evaluated for diagnosis of NPC disease and treatment efficacy assessment with 2-hydroxypropyl-β-cyclodextrin (HPβCD) in NPC. Liquid chromatography-tandem mass spectrometry (LC-MS/MS) methods were developed and validated to measure PPCS in human plasma and cerebrospinal fluid (CSF). A cutoff of 248 ng/mL in plasma provided a sensitivity of 100.0% and specificity of 96.6% in identifying NPC1 patients from control and NPC1 carrier subjects. PPCS was significantly elevated in CSF from NPC1 patients, and CSF PPCS levels were significantly correlated with NPC neurological disease severity scores. Plasma and CSF PPCS did not change significantly in response to intrathetical (IT) HPβCD treatment. In an intravenous (IV) HPβCD trial, plasma PPCS in all patients was significantly reduced. These results demonstrate that plasma PPCS was able to diagnose NPC1 patients with high sensitivity and specificity, and 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 Pediatrics, 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
| | | | - Charles H Vite
- Department of Clinical Studies, University of Pennsylvania School of Veterinary Medicine, PA 19104, 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, Universidade Federal do Rio Grande do Sul, Medical Genetics Service, 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
| | - Olaf Bodamer
- Division of Genetics and Genomics, Boston Children's Hospital, Boston, MA 02115, USA
| | - 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
| | - Ellen Plummer
- Asante Pediatric Hematology and Oncology - Medford, 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
|
63
|
Li D, Shao R, Wang N, Zhou N, Du K, Shi J, Wang Y, Zhao Z, Ye X, Zhang X, Xu H. Sulforaphane Activates a lysosome-dependent transcriptional program to mitigate oxidative stress. Autophagy 2020; 17:872-887. [PMID: 32138578 DOI: 10.1080/15548627.2020.1739442] [Citation(s) in RCA: 71] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Oxidative stress underlies a number of pathological conditions, including cancer, neurodegeneration, and aging. Antioxidant-rich foods help maintain cellular redox homeostasis and mitigate oxidative stress, but the underlying mechanisms are not clear. For example, sulforaphane (SFN), an electrophilic compound that is enriched in cruciferous vegetables such as broccoli, is a potent inducer of cellular antioxidant responses. NFE2L2/NRF2 (nuclear factor, erythroid 2 like 2), a transcriptional factor that controls the expression of multiple detoxifying enzymes through antioxidant response elements (AREs), is a proposed target of SFN. NFE2L2/NRF2 is a target gene of TFEB (transcription factor EB), a master regulator of autophagic and lysosomal functions, which we show here to be potently activated by SFN. SFN induces TFEB nuclear translocation via a Ca2+-dependent but MTOR (mechanistic target of rapamycin kinase)-independent mechanism through a moderate increase in reactive oxygen species (ROS). Activated TFEB then boosts the expression of genes required for autophagosome and lysosome biogenesis, which are known to facilitate the clearance of damaged mitochondria. Notably, TFEB activity is required for SFN-induced protection against both acute oxidant bursts and chronic oxidative stress. Hence, by simultaneously activating macroautophagy/autophagy and detoxifying pathways, natural compound SFN may trigger a self-defense cellular mechanism that can effectively mitigate oxidative stress commonly associated with many metabolic and age-related diseases.Abbreviations: ANOVA: analyzes of variance; AREs: antioxidant response elements; Baf-A1: bafilomycin A1; BHA: butylhydroxyanisole; CAT: catechin hydrate; CCCP: carbonyl cyanide m- chlorophenylhydrazone; CLEAR: coordinated lysosomal expression and regulation; DCFH-DA: 2',7'-dichlorofluorescin diacetate; FBS: fetal bovine serum; GFP: green fluorescent protein; HMOX1/HO-1: heme oxygenase 1; KD: knockdown; KEAP1: kelch like ECH associated protein 1; KO: knockout; LAMP1: lysosomal associated membrane protein 1; MCOLN1/TRPML1: mucolipin 1; ML-SA1: mucolipin-specific synthetic agonist 1; ML-SI3: mucolipin-specific synthetic inhibitor 3; MTOR: mechanistic target of rapamycin kinase; MTORC1: mechanistic target of rapamycin kinase complex 1; NAC: N-acetylcysteine; NFE2L2/NRF2: nuclear factor: erythroid 2 like 2; NPC: Niemann-Pick type C; PBS: phosphate-buffered saline; PPP2/PP2A: protein phosphatase 2; Q-PCR: real time polymerase chain reaction; ROS: reactive oxygen species; RPS6KB1/S6K1/p70S6K: ribosomal protein S6 kinase B1; SFN: sulforaphane; TFEB: transcription factor EB; WT, wild-type.
Collapse
Affiliation(s)
- Dan Li
- Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, College of Pharmaceutical Sciences, Zhejiang University of Technology, Hangzhou, China.,Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, MI, USA
| | - Rong Shao
- Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, College of Pharmaceutical Sciences, Zhejiang University of Technology, Hangzhou, China
| | - Na Wang
- Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, College of Pharmaceutical Sciences, Zhejiang University of Technology, Hangzhou, China.,Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, MI, USA
| | - Nan Zhou
- Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, College of Pharmaceutical Sciences, Zhejiang University of Technology, Hangzhou, China.,Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, MI, USA
| | - Kaili Du
- Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, College of Pharmaceutical Sciences, Zhejiang University of Technology, Hangzhou, China.,Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, MI, USA
| | - Jiahui Shi
- Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, College of Pharmaceutical Sciences, Zhejiang University of Technology, Hangzhou, China
| | - Yihan Wang
- Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, College of Pharmaceutical Sciences, Zhejiang University of Technology, Hangzhou, China.,Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, MI, USA
| | - Zhuangzhuang Zhao
- Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, College of Pharmaceutical Sciences, Zhejiang University of Technology, Hangzhou, China.,Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, MI, USA
| | - Xin Ye
- Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, College of Pharmaceutical Sciences, Zhejiang University of Technology, Hangzhou, China
| | - Xiaoli Zhang
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, MI, USA
| | - Haoxing Xu
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, MI, USA
| |
Collapse
|
64
|
Iron chelation by deferiprone does not rescue the Niemann-Pick Disease Type C1 mouse model. Biometals 2020; 33:87-95. [PMID: 32100150 DOI: 10.1007/s10534-020-00233-5] [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: 02/02/2020] [Accepted: 02/19/2020] [Indexed: 02/08/2023]
Abstract
Niemann-Pick Disease Type C (NP-C) is a fatal lysosomal storage disorder with progressive neurodegeneration. In addition to the characteristic cholesterol and lipid overload phenotype, we previously found that altered metal homeostasis is also a pathological feature. Increased brain iron in the Npc1-/- mouse model of NP-C may potentially contribute to neurodegeneration, similar to neurodegenerative diseases such as Alzheimer's and Parkinson's diseases. Deferiprone (DFP) is a brain penetrating iron chelator that has demonstrated effectiveness in preventing neurological deterioration in Parkinson's disease clinical trials. Therefore, we hypothesized that DFP treatment, targeting brain iron overload, may have therapeutic benefits for NP-C. Npc1-/- mice were assigned to four experimental groups: (1) pre-symptomatic (P15) + 75 mg/kg DFP; (2) pre-symptomatic (P15) + 150 mg/kg DFP; (3) symptomatic (P49) + 75 mg/kg DFP; (4) symptomatic (P49) + 150 mg/kg DFP. Our study found that in Npc1-/- mice, DFP treatment did not offer any improvement over the expected disease trajectory and median lifespan. Moreover, earlier treatment and higher dose of DFP resulted in adverse effects on body weight and onset of ataxia. The outcome of our study indicated that, despite increased brain iron, Npc1-/- mice were vulnerable to pharmacological iron depletion, especially in early life. Therefore, based on the current model, iron chelation therapy is not a suitable treatment option for NP-C.
Collapse
|
65
|
MAEKAWA M, MANO N. Identification and Evaluation of Biomarkers for Niemann-Pick Disease Type C Based on Chemical Analysis Techniques. CHROMATOGRAPHY 2020. [DOI: 10.15583/jpchrom.2020.001] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Affiliation(s)
| | - Nariyasu MANO
- Department of Pharmaceutical Sciences, Tohoku University Hospital
| |
Collapse
|
66
|
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: 102] [Impact Index Per Article: 25.5] [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
|
67
|
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: 30] [Impact Index Per Article: 6.0] [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
|
68
|
The psychopharmacology of Wilson disease and other metabolic disorders. HANDBOOK OF CLINICAL NEUROLOGY 2019. [PMID: 31727212 DOI: 10.1016/b978-0-444-64012-3.00011-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/29/2023]
Abstract
Wilson disease (WD) is a hereditary metabolic disorder (HMD) caused by a mutation in the copper-transporting gene ATP7B affecting the liver and central nervous system. About 30% of patients with WD may initially present with psychiatric symptoms, and management can be difficult. More generally, HMDs are a rare but important cause of psychiatric disorders in adolescents and adults. Main signs of HMDs may remain isolated for years before the appearance of hepatic or neurologic signs. The incidence of HMDs has been estimated at approximately 40 cases per 100,000 live births. Some of them are treatable and new diagnostic methods and therapies have become available. HMDs that present purely with psychiatric symptoms are very difficult to diagnose due to low awareness of these rare diseases among psychiatrists and neurologists. However, it is important to identify HMDs in order to provide disease-specific treatment and possible prevention of irreversible physical and neurologic complications. Genetic counseling can also be provided. Psychotropic medications should be prescribed carefully in that indication. This chapter focuses on three HMD categories: chronic, treatable HMDs (e.g., WD); acute, treatable HMDs; and chronic HMDs that are difficult to treat. In this review we focus on the psychopharmacology of WD and other chronic and difficult-to-treat HMDs. We provide some keys to take into account the main side effects associated with common psychotropic medications.
Collapse
|
69
|
Maekawa M, Jinnoh I, Matsumoto Y, Narita A, Mashima R, Takahashi H, Iwahori A, Saigusa D, Fujii K, Abe A, Higaki K, Yamauchi S, Ozeki Y, Shimoda K, Tomioka Y, Okuyama T, Eto Y, Ohno K, T Clayton P, Yamaguchi H, Mano N. Structural Determination of Lysosphingomyelin-509 and Discovery of Novel Class Lipids from Patients with Niemann-Pick Disease Type C. Int J Mol Sci 2019; 20:ijms20205018. [PMID: 31658747 PMCID: PMC6829288 DOI: 10.3390/ijms20205018] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2019] [Revised: 10/06/2019] [Accepted: 10/08/2019] [Indexed: 02/02/2023] Open
Abstract
Niemann-Pick disease type C (NPC) is an autosomal recessive disorder caused by the mutation of cholesterol-transporting proteins. In addition, early treatment is important for good prognosis of this disease because of the progressive neurodegeneration. However, the diagnosis of this disease is difficult due to a variety of clinical spectrum. Lysosphingomyelin-509, which is one of the most useful biomarkers for NPC, was applied for the rapid and easy detection of NPC. The fact that its chemical structure was unknown until recently implicates the unrevealed pathophysiology and molecular mechanisms of NPC. In this study, we aimed to elucidate the structure of lysosphingomyelin-509 by various mass spectrometric techniques. As our identification strategy, we adopted analytical and organic chemistry approaches to the serum of patients with NPC. Chemical derivatization and hydrogen abstraction dissociation-tandem mass spectrometry were used for the determination of function groups and partial structure, respectively. As a result, we revealed the exact structure of lysosphingomyelin-509 as N-acylated and O-phosphocholine adducted serine. Additionally, we found that a group of metabolites with N-acyl groups were increased considerably in the serum/plasma of patients with NPC as compared to that of other groups using targeted lipidomics analysis. Our techniques were useful for the identification of lysosphingomyelin-509.
Collapse
Affiliation(s)
- Masamitsu Maekawa
- Department of Pharmaceutical Sciences, Tohoku University Hospital, 1-1 Seiryo-machi, Aoba-ku, Sendai, Miyagi 980-8574, Japan.
| | - Isamu Jinnoh
- Laboratory of Clinical Pharmacy, Faculty of Pharmaceutical Sciences, Tohoku University, 1-1 Seiryo-machi, Aoba-Ku, Sendai, Miyagi 980-8574, Japan.
| | - Yotaro Matsumoto
- Department of Pharmaceutical Sciences, Tohoku University Hospital, 1-1 Seiryo-machi, Aoba-ku, Sendai, Miyagi 980-8574, Japan.
- Laboratory of Oncology, Pharmacy Practice and Sciences, Graduate School of Pharmaceutical Sciences, Tohoku University, 6-3 Aoba, Aoba-Ku, Sendai, Miyagi 980-8578, Japan.
| | - Aya Narita
- Division of Child Neurology, Tottori University Hospital, 86 Nishi-machi, Yonago, Tottori 683-8503, Japan.
| | - Ryuichi Mashima
- Department of Clinical Laboratory Medicine, National Center for Child Health and Development, 2-10-1 Okura, Setagaya-ku, Tokyo 157-8535, Japan.
| | - Hidenori Takahashi
- Koichi Tanaka Mass Spectrometry Research Laboratory, Shimadzu Corporation, 1 Nishinokyo-Kuwabaracho Nakagyo-ku, Kyoto 604-8511, Japan.
| | - Anna Iwahori
- Laboratory of Clinical Pharmacy, Faculty of Pharmaceutical Sciences, Tohoku University, 1-1 Seiryo-machi, Aoba-Ku, Sendai, Miyagi 980-8574, Japan.
| | - Daisuke Saigusa
- Department of Pharmaceutical Sciences, Tohoku University Hospital, 1-1 Seiryo-machi, Aoba-ku, Sendai, Miyagi 980-8574, Japan.
- Department of Integrative Genomics, Tohoku Medical Megabank Organization, Tohoku University, 2-1 Seiryo-machi, Aoba-ku, Sendai, Miyagi 980-8575, Japan.
| | - Kumiko Fujii
- Department of Psychiatry, Dokkyo Medical University School of Medicine, 880 Kitakobayashi, Mibu, Tochigi 321-0293, Japan.
| | - Ai Abe
- Laboratory of Clinical Pharmacy, Faculty of Pharmaceutical Sciences, Tohoku University, 1-1 Seiryo-machi, Aoba-Ku, Sendai, Miyagi 980-8574, Japan.
| | - Katsumi Higaki
- Division of Functional Genomics, Research Centre for Bioscience and Technology, Faculty of Medicine, Tottori University, 86 Nishi-cho, Yonago 683-8503, Japan.
| | - Shosei Yamauchi
- Koichi Tanaka Mass Spectrometry Research Laboratory, Shimadzu Corporation, 1 Nishinokyo-Kuwabaracho Nakagyo-ku, Kyoto 604-8511, Japan.
| | - Yuji Ozeki
- Department of Psychiatry, Shiga University of Medical Science, Setatsukiwacho, Otsu, Shiga 520-2192 Japan.
| | - Kazutaka Shimoda
- Department of Psychiatry, Dokkyo Medical University School of Medicine, 880 Kitakobayashi, Mibu, Tochigi 321-0293, Japan.
| | - Yoshihisa Tomioka
- Department of Pharmaceutical Sciences, Tohoku University Hospital, 1-1 Seiryo-machi, Aoba-ku, Sendai, Miyagi 980-8574, Japan.
- Laboratory of Oncology, Pharmacy Practice and Sciences, Graduate School of Pharmaceutical Sciences, Tohoku University, 6-3 Aoba, Aoba-Ku, Sendai, Miyagi 980-8578, Japan.
| | - Torayuki Okuyama
- Department of Clinical Laboratory Medicine, National Center for Child Health and Development, 2-10-1 Okura, Setagaya-ku, Tokyo 157-8535, Japan.
| | - Yoshikatsu Eto
- Advanced Clinical Research Center, Institute for Neurological Disorders, Furusawa-Miyako 255, Asou-ku, Kawasaki, Kanagawa 215-0026, Japan.
| | - Kousaku Ohno
- Division of Child Neurology, Tottori University Hospital, 86 Nishi-machi, Yonago, Tottori 683-8503, Japan.
| | - Peter T Clayton
- Inborn Errors of Metabolism, Clinical and Molecular Genetics Unit, UCL Great Ormond Street Institute of Child Health. 30 Guilford Street, University College London, WC1N 1EH London, UK.
| | - Hiroaki Yamaguchi
- Department of Pharmaceutical Sciences, Tohoku University Hospital, 1-1 Seiryo-machi, Aoba-ku, Sendai, Miyagi 980-8574, Japan.
- Laboratory of Clinical Pharmacy, Faculty of Pharmaceutical Sciences, Tohoku University, 1-1 Seiryo-machi, Aoba-Ku, Sendai, Miyagi 980-8574, Japan.
| | - Nariyasu Mano
- Department of Pharmaceutical Sciences, Tohoku University Hospital, 1-1 Seiryo-machi, Aoba-ku, Sendai, Miyagi 980-8574, Japan.
- Laboratory of Clinical Pharmacy, Faculty of Pharmaceutical Sciences, Tohoku University, 1-1 Seiryo-machi, Aoba-Ku, Sendai, Miyagi 980-8574, Japan.
| |
Collapse
|
70
|
Maekawa M, Jinnoh I, Narita A, Iida T, Saigusa D, Iwahori A, Nittono H, Okuyama T, Eto Y, Ohno K, Clayton PT, Yamaguchi H, Mano N. Investigation of diagnostic performance of five urinary cholesterol metabolites for Niemann-Pick disease type C. J Lipid Res 2019; 60:2074-2081. [PMID: 31586016 DOI: 10.1194/jlr.m093971] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2019] [Revised: 09/30/2019] [Indexed: 11/20/2022] Open
Abstract
Niemann-Pick disease type C (NPC) is an autosomal recessive disorder characterized by progressive nervous degeneration. Because of the diversity of clinical symptoms and onset age, the diagnosis of this disease is difficult. Therefore, biomarker tests have attracted significant attention for earlier diagnostics. In this study, we developed a simultaneous analysis method for five urinary conjugated cholesterol metabolites, which are potential diagnostic biomarkers for a rapid, convenient, and noninvasive chemical diagnosis, using LC/MS/MS. By the method, their urinary concentrations were quantified and the NPC diagnostic performances were evaluated. The developed LC/MS/MS method showed high accuracy and satisfied all analytical method validation criteria. When the urine of healthy controls and patients with NPC was analyzed, three of five urinary conjugated cholesterol metabolite concentrations corrected by urinary creatinine were significantly higher in the patients with NPC. As a result of receiver operating characteristics analysis, these urinary metabolites might have excellent diagnostic marker performance. 3β-Sulfooxy-7β-hydroxy-5-cholenoic acid showed particularly excellent diagnostic performance with both 100% clinical sensitivity and specificity, suggesting that it is a useful NPC diagnostic marker. The urinary conjugated cholesterol metabolites exhibited high NPC diagnostic marker performance and could be used for NPC diagnosis.
Collapse
Affiliation(s)
- Masamitsu Maekawa
- Department of Pharmaceutical Sciences, Tohoku University Hospital, Aoba-ku, Sendai 980-8574, Japan
| | - Isamu Jinnoh
- Faculty of Pharmaceutical Sciences, Tohoku University, Aoba-Ku, Sendai 980-8574, Japan
| | - Aya Narita
- Division of Child Neurology, Tottori University Hospital, Yonago, Tottori 683-8503, Japan
| | - Takashi Iida
- College of Humanities and Sciences, Nihon University, Setagaya-ku, Tokyo 156-8550, Japan
| | - Daisuke Saigusa
- Department of Pharmaceutical Sciences, Tohoku University Hospital, Aoba-ku, Sendai 980-8574, Japan.,Department of Integrative Genomics, Tohoku Medical Megabank Organization, Tohoku University, Aoba-ku, Sendai 980-8575, Japan
| | - Anna Iwahori
- Faculty of Pharmaceutical Sciences, Tohoku University, Aoba-Ku, Sendai 980-8574, Japan
| | - Hiroshi Nittono
- Junshin Clinic Bile Acid Institute, Meguro-ku, Tokyo 152-0011, Japan
| | - Torayuki Okuyama
- Department of Clinical Laboratory Medicine, National Center for Child Health and Development, Setagaya-ku, Tokyo 157-8535, Japan
| | - Yoshikatsu Eto
- Advanced Clinical Research Center, Institute for Neurological Disorders, Asou-ku, Kawasaki, Kanagawa 215-0026, Japan
| | - Kousaku Ohno
- Division of Child Neurology, Tottori University Hospital, Yonago, Tottori 683-8503, Japan
| | - Peter T Clayton
- Biochemistry Research Group, Clinical and Molecular Genetics Unit, UCL Institute of Child Health, London WC1N 1EH, United Kingdom
| | - Hiroaki Yamaguchi
- Department of Pharmaceutical Sciences, Tohoku University Hospital, Aoba-ku, Sendai 980-8574, Japan.,Faculty of Pharmaceutical Sciences, Tohoku University, Aoba-Ku, Sendai 980-8574, Japan
| | - Nariyasu Mano
- Department of Pharmaceutical Sciences, Tohoku University Hospital, Aoba-ku, Sendai 980-8574, Japan.,Faculty of Pharmaceutical Sciences, Tohoku University, Aoba-Ku, Sendai 980-8574, Japan
| |
Collapse
|
71
|
Sitarska D, Ługowska A. Laboratory diagnosis of the Niemann-Pick type C disease: an inherited neurodegenerative disorder of cholesterol metabolism. Metab Brain Dis 2019; 34:1253-1260. [PMID: 31197681 PMCID: PMC6744384 DOI: 10.1007/s11011-019-00445-w] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/21/2019] [Accepted: 05/28/2019] [Indexed: 01/08/2023]
Abstract
Niemann-Pick type C disease (NPC) is a genetically determined neurodegenerative metabolic disease resulting from the mutations in the NPC1 or NPC2 genes. It belongs to the lysosomal storage diseases and its main cause is impaired cholesterol transport in late endosomes or lysosomes. NPC is inherited in an autosomal recessive trait. Due to the wide range in age of onset, often unspecific clinical picture and varying dynamics of disease progression, the diagnosis is very difficult and long-lasting. The most characteristic visceral symptoms are hepato- or hepatosplenomegaly, which may appear independently of neurological or psychiatric symptoms at various stages of the disease. Available biochemical biomarkers should be tested as early as possible in patients presenting with hepato- or hepatosplenomegaly, long-lasting cholestatic jaundice in neonates or infantile patients, as well as in individuals at any age with: vertical supranuclear gaze palsy (VSGP), ataxia, dystonia, frontotemporal dementia and untreatable schizophrenia or psychosis. Research on biomarkers which can detect NPC patients (Cholestan-3β, 5α, 6β-triol, 7-ketocholesterol, lysosphingomyelin isoforms and bile acid metabolites) is still ongoing, although they are not specific for the NPC disease only. This mini review describes currently used diagnostic methods.
Collapse
Affiliation(s)
- Dominika Sitarska
- Department of Genetics, Institute of Psychiatry and Neurology, Al. Sobieskiego 9, 02-957, Warsaw, Poland
| | - Agnieszka Ługowska
- Department of Genetics, Institute of Psychiatry and Neurology, Al. Sobieskiego 9, 02-957, Warsaw, Poland.
| |
Collapse
|
72
|
Bräuer AU, Kuhla A, Holzmann C, Wree A, Witt M. Current Challenges in Understanding the Cellular and Molecular Mechanisms in Niemann-Pick Disease Type C1. Int J Mol Sci 2019; 20:ijms20184392. [PMID: 31500175 PMCID: PMC6771135 DOI: 10.3390/ijms20184392] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2019] [Revised: 09/04/2019] [Accepted: 09/05/2019] [Indexed: 02/06/2023] Open
Abstract
Rare diseases are a heterogeneous group of very different clinical syndromes. Their most common causes are defects in the hereditary material, and they can therefore be passed on to descendants. Rare diseases become manifest in almost all organs and often have a systemic expressivity, i.e., they affect several organs simultaneously. An effective causal therapy is often not available and can only be developed when the underlying causes of the disease are understood. In this review, we focus on Niemann–Pick disease type C1 (NPC1), which is a rare lipid-storage disorder. Lipids, in particular phospholipids, are a major component of the cell membrane and play important roles in cellular functions, such as extracellular receptor signaling, intracellular second messengers and cellular pressure regulation. An excessive storage of fats, as seen in NPC1, can cause permanent damage to cells and tissues in the brain and peripheral nervous system, but also in other parts of the body. Here, we summarize the impact of NPC1 pathology on several organ systems, as revealed in experimental animal models and humans, and give an overview of current available treatment options.
Collapse
Affiliation(s)
- Anja U Bräuer
- Research Group Anatomy, School of Medicine and Health Sciences, Carl von Ossietzky University Oldenburg, D-26129 Oldenburg, Germany.
- Research Center for Neurosensory Science, Carl von Ossietzky University Oldenburg, D-26129 Oldenburg, Germany.
| | - Angela Kuhla
- Institute for Experimental Surgery, Rostock University Medical Center, Schillingallee 69a, 18057 Rostock, Germany.
- Center of Transdisciplinary Neuroscience Rostock, D-18147 Rostock, Germany.
| | - Carsten Holzmann
- Center of Transdisciplinary Neuroscience Rostock, D-18147 Rostock, Germany.
- Institute of Medical Genetics, Rostock University Medical Center, D-18057 Rostock, Germany.
| | - Andreas Wree
- Center of Transdisciplinary Neuroscience Rostock, D-18147 Rostock, Germany.
- Institute of Anatomy, Rostock University Medical Center, D-18057 Rostock, Germany.
| | - Martin Witt
- Center of Transdisciplinary Neuroscience Rostock, D-18147 Rostock, Germany.
- Institute of Anatomy, Rostock University Medical Center, D-18057 Rostock, Germany.
| |
Collapse
|
73
|
Benussi A, Cotelli MS, Cantoni V, Bertasi V, Turla M, Dardis A, Biasizzo J, Manenti R, Cotelli M, Padovani A, Borroni B. Clinical and neurophysiological characteristics of heterozygous NPC1 carriers. JIMD Rep 2019; 49:80-88. [PMID: 31497485 PMCID: PMC6718120 DOI: 10.1002/jmd2.12059] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/19/2019] [Revised: 05/02/2019] [Accepted: 05/27/2019] [Indexed: 12/20/2022] Open
Abstract
Niemann-Pick disease type C (NPC) is an uncommon lysosomal storage disorder, which is characterized neuropathologically by cholinergic dysfunction and presents clinically with a broad series of neurological signs and symptoms. NPC is inherited as an autosomal recessive trait, caused by mutations in the NPC1 or NPC2 genes. However, recent reports have raised concerns on heterozygous NPC1 gene mutation carriers, which historically have been considered as clinically unaffected, occasionally presenting with clinical parkinsonian syndromes or dementia. In the present study, we aimed at comprehensively assessing clinical, biochemical, and neurophysiological features in heterozygous NPC1 gene mutation carriers. We assessed cholinergic intracortical circuits with transcranial magnetic stimulation, executive functions and plasma oxysterol levels in two families comprising two monozygotic twins with a homozygous NPC1 p.P888S mutation, four patients with a compound heterozygous p.E451K and p.G992W mutation, 10 heterozygous NPC1 p.P888S carriers, 1 heterozygous NPC1 p.E451K carrier, and 11 noncarrier family members. We observed a significant impairment in cholinergic circuits, evaluated with short-latency afferent inhibition (SAI), and executive abilities in homozygous/compound heterozygous patients and heterozygous asymptomatic NPC1 carriers, compared to noncarriers. Moreover, we reported a significant correlation between executive functions performances and both plasma oxysterol levels and neurophysiological parameters. These data suggest that heterozygous NPC1 carriers show subclinical deficits in cognition, possibly mediated by an impairment of cholinergic circuits, which in turn may mediate the onset of neurological disorders in a subset of patients.
Collapse
Affiliation(s)
- Alberto Benussi
- Neurology Unit, Department of Clinical and Experimental SciencesUniversity of BresciaBresciaItaly
| | | | - Valentina Cantoni
- Neurology Unit, Department of Clinical and Experimental SciencesUniversity of BresciaBresciaItaly
- Department of Neuroscience, Psychology, Drug Research and Child HealthUniversity of FlorenceFlorenceItaly
| | | | | | - Andrea Dardis
- University Hospital “Santa Maria della Misericordia”UdineItaly
| | | | - Rosa Manenti
- IRCCS Istituto Centro San Giovanni di DioBresciaItaly
| | - Maria Cotelli
- IRCCS Istituto Centro San Giovanni di DioBresciaItaly
| | - Alessandro Padovani
- Neurology Unit, Department of Clinical and Experimental SciencesUniversity of BresciaBresciaItaly
| | - Barbara Borroni
- Neurology Unit, Department of Clinical and Experimental SciencesUniversity of BresciaBresciaItaly
| |
Collapse
|
74
|
Solheim S, Hutchinson SA, Lundanes E, Wilson SR, Thorne JL, Roberg-Larsen H. Fast liquid chromatography-mass spectrometry reveals side chain oxysterol heterogeneity in breast cancer tumour samples. J Steroid Biochem Mol Biol 2019; 192:105309. [PMID: 30779932 DOI: 10.1016/j.jsbmb.2019.02.004] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/11/2018] [Revised: 01/26/2019] [Accepted: 02/12/2019] [Indexed: 01/07/2023]
Abstract
Oxysterols can contribute to proliferation of breast cancer through activation of the Estrogen Receptors, and to metastasis through activation of the Liver X Receptors. Endogenous levels of both esterified and free sidechain-hydroxylated oxysterols were examined in breast cancer tumours from Estrogen Receptor positive and negative breast tumours, using a novel fast liquid chromatography tandem mass spectrometry method. Multiple aliquots of five milligram samples of 22 tumours were analysed for oxysterol content to assess intra- and inter-tumour variation. Derivatization was performed with Girard T reagent (with and without alkaline hydrolysis) and sample clean-up was performed using a robust automatic on-line column switching system ("AFFL"). Oxysterols were separated isocratically on a 2.1 mm inner diameter column packed with ACE SuperPhenylHexyl core shell particles using a mobile phase consisting of 0.1% formic acid in H2O/methanol/acetonitrile (57/10/33, v/v/v) followed by a wash out step (0.1% formic acid in methanol/acetonitrile, 50/50, v/v). The total analysis time, including sample clean-up and column reconditioning, was 8 min (80% time reduction compared to other on-line systems). Analysis revealed large intra-tumour variations of sidechain oxysterols, resulting in no significant differences in endogenous oxysterols levels between Estrogen Receptor positive and Estrogen Receptor negative breast cancers. However, a correlation between esterified and free 27-hydroxycholesterol was observed. The same correlation was not observed for 24S-hydroxycholesterol or 25-hydroxycholesterol. The oxysterol heterogeneity of tumour tissue is a critical factor when assessing the role of these lipids in cancer.
Collapse
Affiliation(s)
| | | | | | | | - James L Thorne
- School of Food Science and Nutrition, University of Leeds, United Kingdom.
| | | |
Collapse
|
75
|
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: 2.6] [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
|
76
|
McGill JB, Johnson M, Hurst S, Cade WT, Yarasheski KE, Ostlund RE, Schechtman KB, Razani B, Kastan MB, McClain DA, de las Fuentes L, Davila-Roman VG, Ory DS, Wickline SA, Semenkovich CF. Low dose chloroquine decreases insulin resistance in human metabolic syndrome but does not reduce carotid intima-media thickness. Diabetol Metab Syndr 2019; 11:61. [PMID: 31384309 PMCID: PMC6664523 DOI: 10.1186/s13098-019-0456-4] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/05/2019] [Accepted: 07/20/2019] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Metabolic syndrome, an obesity-related condition associated with insulin resistance and low-grade inflammation, leads to diabetes, cardiovascular diseases, cancer, osteoarthritis, and other disorders. Optimal therapy is unknown. The antimalarial drug chloroquine activates the kinase ataxia telangiectasia mutated (ATM), improves metabolic syndrome and reduces atherosclerosis in mice. To translate this observation to humans, we conducted two clinical trials of chloroquine in people with the metabolic syndrome. METHODS Eligibility included adults with at least 3 criteria of metabolic syndrome but who did not have diabetes. Subjects were studied in the setting of a single academic health center. The specific hypothesis: chloroquine improves insulin sensitivity and decreases atherosclerosis. In Trial 1, the intervention was chloroquine dose escalations in 3-week intervals followed by hyperinsulinemic euglycemic clamps. Trial 2 was a parallel design randomized clinical trial, and the intervention was chloroquine, 80 mg/day, or placebo for 1 year. The primary outcomes were clamp determined-insulin sensitivity for Trial 1, and carotid intima-media thickness (CIMT) for Trial 2. For Trial 2, subjects were allocated based on a randomization sequence using a protocol in blocks of 8. Participants, care givers, and those assessing outcomes were blinded to group assignment. RESULTS For Trial 1, 25 patients were studied. Chloroquine increased hepatic insulin sensitivity without affecting glucose disposal, and improved serum lipids. For Trial 2, 116 patients were randomized, 59 to chloroquine (56 analyzed) and 57 to placebo (51 analyzed). Chloroquine had no effect on CIMT or carotid contrast enhancement by MRI, a pre-specified secondary outcome. The pre-specified secondary outcomes of blood pressure, lipids, and activation of JNK (a stress kinase implicated in diabetes and atherosclerosis) were decreased by chloroquine. Adverse events were similar between groups. CONCLUSIONS These findings suggest that low dose chloroquine, which improves the metabolic syndrome through ATM-dependent mechanisms in mice, modestly improves components of the metabolic syndrome in humans but is unlikely to be clinically useful in this setting.Trial registration ClinicalTrials.gov (NCT00455325, NCT00455403), both posted 03 April 2007.
Collapse
Affiliation(s)
- Janet B. McGill
- Division of Endocrinology, Metabolism & Lipid Research, Department of Medicine, Washington University School of Medicine, 660 South Euclid Avenue, Box 8127, St. Louis, MO 63110 USA
| | - Mariko Johnson
- Division of Endocrinology, Metabolism & Lipid Research, Department of Medicine, Washington University School of Medicine, 660 South Euclid Avenue, Box 8127, St. Louis, MO 63110 USA
| | - Stacy Hurst
- Division of Endocrinology, Metabolism & Lipid Research, Department of Medicine, Washington University School of Medicine, 660 South Euclid Avenue, Box 8127, St. Louis, MO 63110 USA
| | - William T. Cade
- Program in Physical Therapy, Washington University, St. Louis, MO USA
| | - Kevin E. Yarasheski
- Division of Endocrinology, Metabolism & Lipid Research, Department of Medicine, Washington University School of Medicine, 660 South Euclid Avenue, Box 8127, St. Louis, MO 63110 USA
| | - Richard E. Ostlund
- Division of Endocrinology, Metabolism & Lipid Research, Department of Medicine, Washington University School of Medicine, 660 South Euclid Avenue, Box 8127, St. Louis, MO 63110 USA
| | | | - Babak Razani
- Cardiovascular Division, Washington University, St. Louis, MO USA
| | - Michael B. Kastan
- Department of Pharmacology & Cancer Biology, Duke University, Durham, NC USA
| | - Donald A. McClain
- Department of Internal Medicine, Wake Forest School of Medicine, Winston-Salem, NC USA
| | | | | | - Daniel S. Ory
- Cardiovascular Division, Washington University, St. Louis, MO USA
| | | | - Clay F. Semenkovich
- Division of Endocrinology, Metabolism & Lipid Research, Department of Medicine, Washington University School of Medicine, 660 South Euclid Avenue, Box 8127, St. Louis, MO 63110 USA
- Department of Cell Biology & Physiology, Washington University, St. Louis, MO USA
| |
Collapse
|
77
|
Degtyareva AV, Proshlyakova TY, Gautier MS, Degtyarev DN, Kamenets EA, Baydakova GV, Rebrikov DV, Zakharova EY. Oxysterol/chitotriosidase based selective screening for Niemann-Pick type C in infantile cholestasis syndrome patients. BMC MEDICAL GENETICS 2019; 20:123. [PMID: 31296176 PMCID: PMC6625024 DOI: 10.1186/s12881-019-0857-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/16/2019] [Accepted: 07/02/2019] [Indexed: 01/11/2023]
Abstract
Background Niemann-Pick disease type C (NP-C) is an inherited neurodegenerative disease (1 per 100 000 newborns) caused by NPC proteins impairment that leads to unesterified cholesterol accumulation in late endosomal/lysosomal compartments. To date the NP-C diagnostics is usually based on cholesterol detection in fibroblasts using an invasive and time-consuming Filipin staining and we need more arguments to widely introduce oxysterols as a biomarkers in NP-C. Methods Insofar as NP-C represents about 8% of all infant cholestases, in this prospective observational study we tried to re-assess the specificity plasma oxysterol and chitotriosidase as a biochemical screening markers of NP-C in children with cholestasis syndrome of unknown origin. For 108 patients (aged from 2 weeks to 7 years) the levels of cholestane-3β,5α,6β-triol (C-triol) and chitotriosidase (ChT) were measured. For patients with elevated C-triol and/or ChT the NPC1 and NPC2 genes were Sanger-sequenced and 47 additional genes (from the custom liver damage panel) were NGS-sequenced. Results Increased C-triol level (> 50 ng/ml) was detected in 4 (of 108) infants with cholestasis syndrome of unknown origin, with following molecular genetic NP-C diagnosis for one patient. Plasma cholesterol significantly correlates with C-triol (p < 0.05). NGS of high C-triol infants identified three patients with mutations in JAG1 (Alagille syndrome) and ABCB11 (Byler disease) genes. Increased ChT activity was detected in 8 (of 108) patients with various aetiologies, including NP-C, Byler disease and biliary atresia. Conclusion Combined analysis of ChT activity and C-triol levels is an effective method for identifying NP-C.
Collapse
Affiliation(s)
- Anna V Degtyareva
- Kulakov National Medical Research Center for Obstetrics, Gynecology and Perinatology, Moscow, Russia.,Sechenov First Moscow State Medical University, Moscow, Russia
| | | | - Marina S Gautier
- Kulakov National Medical Research Center for Obstetrics, Gynecology and Perinatology, Moscow, Russia
| | - Dmitry N Degtyarev
- Kulakov National Medical Research Center for Obstetrics, Gynecology and Perinatology, Moscow, Russia.,Sechenov First Moscow State Medical University, Moscow, Russia
| | | | | | - Denis V Rebrikov
- Kulakov National Medical Research Center for Obstetrics, Gynecology and Perinatology, Moscow, Russia. .,Pirogov Russian National Research Medical University, Moscow, Russia.
| | | |
Collapse
|
78
|
Sidhu R, Mondjinou Y, Qian M, Song H, Kumar AB, Hong X, Hsu FF, Dietzen DJ, Yanjanin NM, Porter FD, Berry-Kravis E, Vite CH, Gelb MH, Schaffer JE, Ory DS, Jiang X. N-acyl- O-phosphocholineserines: structures of a novel class of lipids that are biomarkers for Niemann-Pick C1 disease. J Lipid Res 2019; 60:1410-1424. [PMID: 31201291 DOI: 10.1194/jlr.ra119000157] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2019] [Revised: 06/13/2019] [Indexed: 01/29/2023] Open
Abstract
Niemann-Pick disease type C1 (NPC1) is a fatal, neurodegenerative, cholesterol storage disorder. With new therapeutics in clinical trials, there is an urgency to improve diagnostics and monitor therapeutic efficacy with biomarkers. In this study, we sought to define the structure of an unknown lipid biomarker for NPC1 with [M + H]+ ion at m/z 509.3351, previously designated as lysoSM-509. The structure of N-palmitoyl-O-phosphocholineserine (PPCS) was proposed for the lipid biomarker based on the results from mass spectrometric analyses and chemical derivatizations. As no commercial standard is available, authentic PPCS was chemically synthesized, and the structure was confirmed by comparison of endogenous and synthetic compounds as well as their derivatives using liquid chromatography-tandem mass spectrometry (LC-MS/MS). PPCS is the most abundant species among N-acyl-O-phosphocholineserines (APCS), a class of lipids that have not been previously detected in biological samples. Further analysis demonstrated that all APCS species with acyl groups ranging from C14 to C24 were elevated in NPC1 plasma. PPCS is also elevated in both central and peripheral tissues of the NPC1 cat model. Identification of APCS structures provide an opportunity for broader exploration of the roles of these novel lipids in NPC1 disease pathology and diagnosis.
Collapse
Affiliation(s)
- Rohini Sidhu
- Departments of Medicine, Washington University School of Medicine, St. Louis, MO 63110
| | - Yawo Mondjinou
- Departments of Medicine, Washington University School of Medicine, St. Louis, MO 63110
| | - Mingxing Qian
- Developmental Biology, Washington University School of Medicine, St. Louis, MO 63110
| | - Haowei Song
- Process and Analytical Development, MilliporeSigma, St. Louis, MO 63118
| | - Arun Babu Kumar
- Department of Chemistry, University of Washington, Seattle, WA 98195
| | - Xinying Hong
- Department of Chemistry, University of Washington, Seattle, WA 98195
| | - Fong-Fu Hsu
- Departments of Medicine, Washington University School of Medicine, St. Louis, MO 63110
| | - Dennis J Dietzen
- Pediatrics, Washington University School of Medicine, St. Louis, MO 63110
| | - Nicole M Yanjanin
- Section on Molecular Dysmorphology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Department of Health and Human Services, Bethesda, MD 20892
| | - Forbes D Porter
- Section on Molecular Dysmorphology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Department of Health and Human Services, Bethesda, MD 20892
| | - Elizabeth Berry-Kravis
- Departments of Pediatrics, Neurological Sciences, and Biochemistry, Rush University Medical Center, Chicago, IL 60612
| | - Charles H Vite
- Department of Clinical Studies and Advanced Medicine, University of Pennsylvania School of Veterinary Medicine, Philadelphia, PA 70736
| | - Michael H Gelb
- Process and Analytical Development, MilliporeSigma, St. Louis, MO 63118
| | - Jean E Schaffer
- Departments of Medicine, Washington University School of Medicine, St. Louis, MO 63110
| | - Daniel S Ory
- Departments of Medicine, Washington University School of Medicine, St. Louis, MO 63110
| | - Xuntian Jiang
- Departments of Medicine, Washington University School of Medicine, St. Louis, MO 63110
| |
Collapse
|
79
|
Kilk K. Metabolomics for Animal Models of Rare Human Diseases: An Expert Review and Lessons Learned. OMICS-A JOURNAL OF INTEGRATIVE BIOLOGY 2019; 23:300-307. [PMID: 31120384 DOI: 10.1089/omi.2019.0065] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Rare diseases occur with a frequency ≤1:1500-1:2500 depending on the location and applicable definitions across countries. Although individually rare, they collectively affect as much as 4-8% of population imposing a large burden on public health. Rarity in prevalence means prolonged path to accurate diagnosis, lack of treatment options, and also limited chances for preclinical studies of pathogenesis. I discuss in this expert review (1) what metabolomics, as a high throughput systems sciences technology, offers for rare disease studies, (2) why animal models are important for the study of rare human diseases and what should be kept in mind while using animal models, and finally, (3) provide examples of recent research to highlight how metabolomics on animal models of rare diseases perform, and how these results can lead to the knowhow, which raises genome, metabolome, and phenotype integration to a whole new level. In sum, metabolomics has been for years in clinical use for diagnosis of certain types of rare diseases. Determination of pathogenesis of more complex diseases and testing of treatment strategies is where animal models and systems biology analytical approaches are necessary. From gathered data, it is possible to go back to diagnostic and prognostic markers for rare diseases, which so far lack reliable and robust diagnosis and therapeutic options. In the future, a major challenge is to reveal the links between genotype, metabolism, and phenotype. Rare diseases could be the key in that process.
Collapse
Affiliation(s)
- Kalle Kilk
- Department of Biochemistry, Institute of Biomedicine and Translational Medicine, University of Tartu, Tartu, Estonia
| |
Collapse
|
80
|
Bonnot O, Gama CS, Mengel E, Pineda M, Vanier MT, Watson L, Watissée M, Schwierin B, Patterson MC. Psychiatric and neurological symptoms in patients with Niemann-Pick disease type C (NP-C): Findings from the International NPC Registry. World J Biol Psychiatry 2019; 20:310-319. [PMID: 28914127 DOI: 10.1080/15622975.2017.1379610] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
Objectives: Niemann-Pick disease type C (NP-C) is a rare inherited neurovisceral disease that should be recognised by psychiatrists as a possible underlying cause of psychiatric abnormalities. This study describes NP-C patients who had psychiatric manifestations at enrolment in the international NPC Registry, a unique multicentre, prospective, observational disease registry. Methods: Treating physicians' data entries describing psychiatric manifestations in NPC patients were coded and grouped by expert psychiatrists. Results: Out of 386 NP-C patients included in the registry as of October 2015, psychiatric abnormalities were reported to be present in 34% (94/280) of those with available data. Forty-four patients were confirmed to have identifiable psychiatric manifestations, with text describing these psychiatric manifestations. In these 44 patients, the median (range) age at onset of psychiatric manifestations was 17.9 years (2.5-67.9; n = 15), while the median (range) age at NP-C diagnosis was 23.7 years (0.2-69.8; n = 34). Almost all patients (43/44; 98%) had an occurrence of ≥1 neurological manifestation at enrolment. Conclusions: These data show that substantial delays in diagnosis of NP-C are long among patients with psychiatric symptoms and, moreover, patients presenting with psychiatric features and at least one of cognitive impairment, neurological manifestations, and/or visceral symptoms should be screened for NP-C.
Collapse
Affiliation(s)
- Olivier Bonnot
- a Department of Child and Adolescent Psychiatry , University and CHU of Nantes , Nantes , France
| | - Clarissa S Gama
- b Laboratory of Molecular Psychiatry , Hospital de Clínicas de Porto Alegre, Federal University of Rio Grande do Sul , Porto Alegre , Brazil
| | - Eugen Mengel
- c Paediatric and Adolescent Medical Centre , Johannes Gutenberg University , Mainz , Germany
| | - Mercè Pineda
- d Department of Neuropediatrica , Fundacio Hospital Sant Joan de Déu , Barcelona , Spain
| | - Marie T Vanier
- e Metabolomic and Metabolic Diseases , INSERM Unit 820 , Lyon , France
| | | | - Marie Watissée
- g Actelion Pharmaceuticals Ltd , Allschwil , Switzerland
| | | | - Marc C Patterson
- h Pediatric and Adolescent Medicine , Mayo Clinic , Rochester , MN , USA
| |
Collapse
|
81
|
Bonnot O, Klünemann HH, Velten C, Torres Martin JV, Walterfang M. Systematic review of psychiatric signs in Niemann-Pick disease type C. World J Biol Psychiatry 2019; 20:320-332. [PMID: 29457916 DOI: 10.1080/15622975.2018.1441548] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Objectives: We conducted the first systematic literature review and analysis of psychiatric manifestations in Niemann-Pick disease type C (NPC) to describe: (1) time of occurrence of psychiatric manifestations relative to other disease manifestations; and (2) frequent combinations of psychiatric, neurological and visceral disease manifestations. Methods: A systematic EMBase literature search was conducted to identify, collate and analyze published data from patients with NPC associated with psychiatric symptoms, published between January 1967 and November 2015. Results: Of 152 identified publications 40 were included after screening that contained useable data from 58 NPC patients (mean [SD] age at diagnosis of NPC 27.8 [15.1] years). Among patients with available data, cognitive, memory and instrumental impairments were most frequent (90% of patients), followed by psychosis (62%), altered behavior (52%) and mood disorders (38%). Psychiatric manifestations were reported before or at neurological disease onset in 41 (76%) patients; organic signs (e.g., hepatosplenomegaly, hearing problems) were reported before psychiatric manifestations in 12 (22%). Substantial delays to diagnosis were observed (5-6 years between psychiatric presentation and NPC diagnosis). Conclusions: NPC should be considered as a possible cause of psychiatric manifestations in patients with an atypical disease course, acute-onset psychosis, treatment failure, and/or certain combinations of psychiatric/neurological/visceral symptoms.
Collapse
Affiliation(s)
- Olivier Bonnot
- a Child and Adolescent Psychiatry Department , CHU and University of Nantes , Nantes , France
| | - Hans-Hermann Klünemann
- b University Clinic for Psychiatry and Psychotherapy, Regensburg University , Regensburg , Germany
| | | | | | | |
Collapse
|
82
|
Baguña Torres J, Yu Z, Bordoloi J, Sunassee K, Smith D, Smith C, Chen O, Purchase R, Tuschl K, Spencer J, Platt F, Blower PJ. Imaging of changes in copper trafficking and redistribution in a mouse model of Niemann-Pick C disease using positron emission tomography. Biometals 2019; 32:293-306. [PMID: 30847690 PMCID: PMC6437134 DOI: 10.1007/s10534-019-00185-5] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2019] [Accepted: 02/25/2019] [Indexed: 01/13/2023]
Abstract
Niemann-Pick C disease (NPC) is an autosomal recessive lysosomal storage disorder resulting from mutations in the NPC1 (95% of cases) or NPC2 genes. Disturbance of copper homeostasis has been reported in NPC1 disease. In this study we have used whole-body positron emission tomography (PET) and brain electronic autoradiography with copper-64 (64Cu), in the form of the copper(II) bis(thiosemicarbazonato) complex 64Cu-GTSM, to image short-term changes in copper trafficking after intravenous injection in a transgenic mouse model of NPC1 disease. 64Cu-GTSM is taken up in all tissues and dissociates rapidly inside cells, allowing monitoring of the subsequent efflux and redistribution of 64Cu from all tissues. Significantly enhanced retention of 64Cu radioactivity was observed in brain, lungs and blood at 15 h post-injection in symptomatic Npc1-/- transgenic mice compared to wildtype controls. The enhanced retention of 64Cu in brain was confirmed by electronic autoradiography, particularly in the midbrain, thalamus, medulla and pons regions. Positron emission tomography imaging with 64Cu in selected chemical forms could be a useful diagnostic and research tool for the management and understanding of NPC1 disease.
Collapse
Affiliation(s)
- Julia Baguña Torres
- School of Biomedical Engineering and Imaging Sciences, King's College London, St Thomas' Hospital, London, SE1 7EH, UK
| | - Zilin Yu
- School of Biomedical Engineering and Imaging Sciences, King's College London, St Thomas' Hospital, London, SE1 7EH, UK
| | - Jayanta Bordoloi
- School of Biomedical Engineering and Imaging Sciences, King's College London, St Thomas' Hospital, London, SE1 7EH, UK
| | - Kavitha Sunassee
- School of Biomedical Engineering and Imaging Sciences, King's College London, St Thomas' Hospital, London, SE1 7EH, UK
| | - David Smith
- Department of Pharmacology, University of Oxford, Mansfield Road, Oxford, OX1 3QT, UK
| | - Claire Smith
- Department of Pharmacology, University of Oxford, Mansfield Road, Oxford, OX1 3QT, UK
| | - Oscar Chen
- Department of Pharmacology, University of Oxford, Mansfield Road, Oxford, OX1 3QT, UK
| | - Rupert Purchase
- Department of Chemistry, School of Life Sciences, University of Sussex, Falmer, Brighton, BN1 9QJ, UK
| | - Karin Tuschl
- MRC Centre for Developmental Neurobiology IoPPN, King's College London, London, SE1 1UL, UK
- Department of Cell and Developmental Biology, University College London, London, WC1E 6BT, UK
| | - John Spencer
- Department of Chemistry, School of Life Sciences, University of Sussex, Falmer, Brighton, BN1 9QJ, UK
| | - Frances Platt
- Department of Pharmacology, University of Oxford, Mansfield Road, Oxford, OX1 3QT, UK
| | - Philip J Blower
- School of Biomedical Engineering and Imaging Sciences, King's College London, St Thomas' Hospital, London, SE1 7EH, UK.
| |
Collapse
|
83
|
Fog CK, Kirkegaard T. Animal models for Niemann-Pick type C: implications for drug discovery & development. Expert Opin Drug Discov 2019; 14:499-509. [PMID: 30887840 DOI: 10.1080/17460441.2019.1588882] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
INTRODUCTION Niemann-Pick type C (NPC) is a neurovisceral, progressively detrimental lysosomal storage disease with very limited therapeutic options and no approved treatment available in the US. Despite its rarity, NPC has seen increased drug developmental efforts over the past decade, culminating in the completion of two potential registration trials in 2018. Areas covered: This review highlights the many available animal models that have been developed in the field and briefly covers classical and new cell technologies. This review provides a high-level evaluation and prioritization of the various models with regard to efficient and clinically translatable drug development, and briefly discusses the relevant developments and opportunities pertaining to this. Expert opinion: With a number of in vitro and in vivo models available, and with having several drugs, all with various mechanisms of action, either approved or in late stage development, the NPC field is in an exciting time. One of the challenges for researchers and developers will be the ability to make use of the lessons learnt from existing late-stage programs as well as the incorporation not only of the opportunities but also the limitations of the many models into successful drug discovery and translational development programs.
Collapse
|
84
|
Diagnostic performance evaluation of sulfate-conjugated cholesterol metabolites as urinary biomarkers of Niemann-Pick disease type C. Clin Chim Acta 2019; 494:58-63. [PMID: 30876856 DOI: 10.1016/j.cca.2019.03.1610] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2019] [Revised: 03/06/2019] [Accepted: 03/10/2019] [Indexed: 12/15/2022]
Abstract
BACKGROUND Niemann-Pick disease type C (NPC) is an autosomal recessive inherited disorder with progressive neuronal degeneration. Because conventional diagnostic methods are complicated and invasive, biomarker tests have drawn attention. We aimed to evaluate three urinary conjugated cholesterol metabolites as diagnostic biomarkers for NPC. METHODS Urine samples from 23 patients with NPC, 28 healthy controls, and 7 patients with inherited metabolic disorders were analyzed. 3β-Sulfooxy-7β-N-acetylglucosaminyl-5-cholen-24-oic acid and its glycine and taurine conjugates in urine were quantified by liquid chromatography-tandem mass spectrometry. The diagnostic performance of the three metabolites and their total concentration was evaluated. RESULT Creatinine-corrected concentrations of three metabolites and their total concentration were all significantly higher in NPC patients (0.0098 < P < .0448). The area under the receiver operating curve for all metabolites exceeded 0.95, the clinical specificity was 92-100%, and the clinical sensitivity was ~95%. In the urine of patients with other inherited metabolic diseases, the concentrations of the metabolites were lower than those in the urine of patients with NPC. CONCLUSION These conjugated cholesterol metabolites in urine can serve as useful diagnostic markers for noninvasive screening of NPC.
Collapse
|
85
|
Jiang X, Sidhu R, Orsini JJ, Farhat NY, Porter FD, Berry-Kravis E, Schaffer JE, Ory DS. Diagnosis of niemann-pick C1 by measurement of bile acid biomarkers in archived newborn dried blood spots. Mol Genet Metab 2019; 126:183-187. [PMID: 30172462 PMCID: PMC6365165 DOI: 10.1016/j.ymgme.2018.08.007] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/01/2018] [Revised: 08/18/2018] [Accepted: 08/18/2018] [Indexed: 01/29/2023]
Abstract
BACKGROUND Niemann-Pick disease type C1 (NPC1) is a rare, neurodegenerative cholesterol storage disorder. Diagnostic delay of >5 years is common due to the rarity of the disease and non-specific early symptoms. To improve diagnosis and facilitate early intervention, we previously developed a newborn screening assay based on newly identified plasma bile acid biomarkers. Because the newborn screen had been validated using dried blood spots (DBS) from already diagnosed NPC1 patients, an unanswered question was whether the screen would be able to detect individuals with NPC1 at birth. METHODS To address this critical question, we obtained the newborn DBS for already diagnosed NPC1 subjects (n = 15) and carriers (n = 3) residing in California, New York, and Michigan states that archive residual DBS in biorepositories. For each of the DBS, we obtained two neighbor controls - DBS from patients born on the same day and in the same hospital as the NPC1 patients and carriers. 3β,5α,6β-trihydroxycholanic acid (bile acid A) and trihydroxycholanic acid glycine conjugate (bile acid B) were measured in the DBS using a liquid chromatography-tandem mass spectrometry (LC-MS/MS) assay. RESULTS Bile acid B, the more specific biomarker for which the fully validated DBS assay was developed, was detected in 8/15 NPC1 patients, and elevated above the cut-off in 2/15 patients (the two samples with the shortest storage time). Bile acid B was detected in 2/2, 6/10, and 0/7 NPC1 samples that have been stored for <10.5 years, 13-20 years, and > 20 years, respectively, indicating that the glycine conjugate is detectable in DBS but may have reduced long-term stability compared with bile acid A, the precursor trihydroxycholanic acid, which was elevated in 15/15 NPC1 subjects, but not in carriers and controls. CONCLUSIONS These results demonstrate that newborn screening for NPC1 disease is feasible using bile acid biomarkers.
Collapse
Affiliation(s)
- Xuntian Jiang
- Diabetic Cardiovascular Disease Center, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Rohini Sidhu
- Diabetic Cardiovascular Disease Center, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Joseph J Orsini
- New York State Dept. of Health, Wadsworth Center, Albany, NY 12201, 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
| | - Forbes D Porter
- Section on Molecular Dysmorphology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, NIH, DHHS, Bethesda, MD 20892, USA
| | | | - Jean E Schaffer
- Diabetic Cardiovascular Disease Center, 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.
| |
Collapse
|
86
|
Oxysterols as a biomarker in diseases. Clin Chim Acta 2019; 491:103-113. [PMID: 30685361 DOI: 10.1016/j.cca.2019.01.022] [Citation(s) in RCA: 51] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2018] [Revised: 01/20/2019] [Accepted: 01/23/2019] [Indexed: 12/24/2022]
Abstract
Cholesterol is one of the most important chemical substances as a structural element in human cells, and it is very susceptible to oxidation reactions that form oxysterol. Oxysterols exhibit almost the exact structure as cholesterol and a cholesterol precursor (7-dehydrocholesterol) with an additional hydroxyl, epoxy or ketone moiety. The oxidation reaction is performed via an enzymatic or non-enzymatic mechanism. The wide array of enzymatic oxysterols encountered in the human body varies in origin and function. Oxysterols establish a concentration equilibrium in human body fluids. Disease may alter the equilibrium, and oxysterols may be used as a diagnostic tool. The current review presents the possibility of using non-enzymatic oxysterols and disturbances in enzymatic oxysterol equilibrium in the human body as a potential biomarker for diagnosing and/or monitoring of the progression of various diseases.
Collapse
|
87
|
Zielinski ZAM, Pratt DA. H-Atom Abstraction vs Addition: Accounting for the Diverse Product Distribution in the Autoxidation of Cholesterol and Its Esters. J Am Chem Soc 2019; 141:3037-3051. [DOI: 10.1021/jacs.8b11524] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Affiliation(s)
- Zosia A. M. Zielinski
- Department of Chemistry and Biomolecular Sciences, University of Ottawa, Ottawa, Ontario K1N 6N5, Canada
| | - Derek A. Pratt
- Department of Chemistry and Biomolecular Sciences, University of Ottawa, Ottawa, Ontario K1N 6N5, Canada
| |
Collapse
|
88
|
Houben T, Magro Dos Reis I, Oligschlaeger Y, Steinbusch H, Gijbels MJJ, Hendrikx T, Binder CJ, Cassiman D, Westerterp M, Prickaerts J, Shiri-Sverdlov R. Pneumococcal Immunization Reduces Neurological and Hepatic Symptoms in a Mouse Model for Niemann-Pick Type C1 Disease. Front Immunol 2019; 9:3089. [PMID: 30666257 PMCID: PMC6330339 DOI: 10.3389/fimmu.2018.03089] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2018] [Accepted: 12/13/2018] [Indexed: 11/13/2022] Open
Abstract
Niemann-Pick type C1 (NPC1) disease is caused by a deleterious mutation in the Npc1 gene, causing lysosomal accumulation of unesterified cholesterol and sphingolipids. Consequently, NPC1 disease patients suffer from severe neurovisceral symptoms which, in the absence of effective treatments, result in premature death. NPC1 disease patients display increased plasma levels of cholesterol oxidation products such as those enriched in oxidized low-density lipoprotein (oxLDL), a pro-inflammatory mediator. While it has been shown that inflammation precedes and exacerbates symptom severity in NPC1 disease, it is unclear whether oxLDL contributes to NPC1 disease progression. In this study, we investigated the effects of increasing anti-oxLDL IgM autoantibodies on systemic and neurological symptoms in an NPC1 disease mouse model. For this purpose, Npc1nih mice were immunized with heat-inactivated S. pneumoniae, an immunogen which elicits an IgM autoantibody-mediated immune response against oxLDL. Npc1nih mice injected with heat-inactivated pneumococci displayed an improved hepatic phenotype, including liver lipid accumulation and inflammation. In addition, regression of motor skills was delayed in immunized Npc1nih. In line with these results, brain analyses showed an improved cerebellar phenotype and neuroinflammation in comparison with control-treated subjects. This study highlights the potential of the pneumococcal immunization as a novel therapeutical approach in NPC1 disease. Future research should investigate whether implementation of this therapy can improve life span and quality of life of NPC1 disease patients.
Collapse
Affiliation(s)
- Tom Houben
- Department of Molecular Genetics, School of Nutrition and Translational Research in Metabolism, Maastricht University, Maastricht, Netherlands
| | - Inês Magro Dos Reis
- Department of Molecular Genetics, School of Nutrition and Translational Research in Metabolism, Maastricht University, Maastricht, Netherlands
| | - Yvonne Oligschlaeger
- Department of Molecular Genetics, School of Nutrition and Translational Research in Metabolism, Maastricht University, Maastricht, Netherlands
| | - Hellen Steinbusch
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience, Maastricht University, Maastricht, Netherlands
| | - Marion J J Gijbels
- Department of Molecular Genetics, School of Nutrition and Translational Research in Metabolism, Maastricht University, Maastricht, Netherlands
| | - Tim Hendrikx
- Department of Laboratory Medicine, Medical University of Vienna, Vienna, Austria.,Center for Molecular Medicine, Austrian Academy of Sciences, Vienna, Austria
| | - Christoph J Binder
- Department of Laboratory Medicine, Medical University of Vienna, Vienna, Austria.,Center for Molecular Medicine, Austrian Academy of Sciences, Vienna, Austria
| | - David Cassiman
- Liver Research Unit, University of Leuven, Leuven, Belgium.,Department of Gastroenterology-Hepatology and Metabolic Center, University Hospitals Leuven, Leuven, Belgium
| | - Marit Westerterp
- Section Molecular Genetics, Department of Pediatrics, University of Groningen, University Medical Center Groningen, Groningen, Netherlands
| | - Jos Prickaerts
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience, Maastricht University, Maastricht, Netherlands
| | - Ronit Shiri-Sverdlov
- Department of Molecular Genetics, School of Nutrition and Translational Research in Metabolism, Maastricht University, Maastricht, Netherlands
| |
Collapse
|
89
|
Chen X, Lee J, Wu H, Tsang AW, Furdui CM. Mass Spectrometry in Advancement of Redox Precision Medicine. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1140:327-358. [PMID: 31347057 PMCID: PMC9236553 DOI: 10.1007/978-3-030-15950-4_19] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Redox (portmanteau of reduction-oxidation) reactions involve the transfer of electrons between chemical species in biological processes fundamental to life. It is of outmost importance that cells maintain a healthy redox state by balancing the action of oxidants and antioxidants; failure to do so leads to a multitude of diseases including cancer, diabetes, fibrosis, autoimmune diseases, and cardiovascular and neurodegenerative diseases. From the perspective of precision medicine, it is therefore beneficial to interrogate the redox phenotype of the individual-similar to the use of genomic sequencing-in order to design tailored strategies for disease prevention and treatment. This chapter provides an overview of redox metabolism and focuses on how mass spectrometry (MS) can be applied to advance our knowledge in redox biology and precision medicine.
Collapse
Affiliation(s)
- Xiaofei Chen
- Department of Internal Medicine, Section on Molecular Medicine, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - Jingyun Lee
- Department of Internal Medicine, Section on Molecular Medicine, Wake Forest School of Medicine, Winston-Salem, NC, USA
- Wake Forest Baptist Comprehensive Cancer Center, Winston-Salem, NC, USA
| | - Hanzhi Wu
- Department of Internal Medicine, Section on Molecular Medicine, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - Allen W Tsang
- Department of Internal Medicine, Section on Molecular Medicine, Wake Forest School of Medicine, Winston-Salem, NC, USA
- Wake Forest Baptist Comprehensive Cancer Center, Winston-Salem, NC, USA
- Center for Redox Biology and Medicine, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - Cristina M Furdui
- Department of Internal Medicine, Section on Molecular Medicine, Wake Forest School of Medicine, Winston-Salem, NC, USA.
- Wake Forest Baptist Comprehensive Cancer Center, Winston-Salem, NC, USA.
- Center for Redox Biology and Medicine, Wake Forest School of Medicine, Winston-Salem, NC, USA.
| |
Collapse
|
90
|
van der Lienden MJC, Gaspar P, Boot R, Aerts JMFG, van Eijk M. Glycoprotein Non-Metastatic Protein B: An Emerging Biomarker for Lysosomal Dysfunction in Macrophages. Int J Mol Sci 2018; 20:E66. [PMID: 30586924 PMCID: PMC6337583 DOI: 10.3390/ijms20010066] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2018] [Revised: 12/20/2018] [Accepted: 12/21/2018] [Indexed: 12/18/2022] Open
Abstract
Several diseases are caused by inherited defects in lysosomes, the so-called lysosomal storage disorders (LSDs). In some of these LSDs, tissue macrophages transform into prominent storage cells, as is the case in Gaucher disease. Here, macrophages become the characteristic Gaucher cells filled with lysosomes laden with glucosylceramide, because of their impaired enzymatic degradation. Biomarkers of Gaucher cells were actively searched, particularly after the development of costly therapies based on enzyme supplementation and substrate reduction. Proteins selectively expressed by storage macrophages and secreted into the circulation were identified, among which glycoprotein non-metastatic protein B (GPNMB). This review focusses on the emerging potential of GPNMB as a biomarker of stressed macrophages in LSDs as well as in acquired pathologies accompanied by an excessive lysosomal substrate load in macrophages.
Collapse
Affiliation(s)
| | - Paulo Gaspar
- Leiden Institute of Chemistry, Leiden University, 2333 CC Leiden, The Netherlands.
| | - Rolf Boot
- Leiden Institute of Chemistry, Leiden University, 2333 CC Leiden, The Netherlands.
| | - Johannes M F G Aerts
- Leiden Institute of Chemistry, Leiden University, 2333 CC Leiden, The Netherlands.
| | - Marco van Eijk
- Leiden Institute of Chemistry, Leiden University, 2333 CC Leiden, The Netherlands.
| |
Collapse
|
91
|
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: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2017] [Accepted: 02/14/2018] [Indexed: 01/19/2023]
|
92
|
Dias IH, Wilson SR, Roberg-Larsen H. Chromatography of oxysterols. Biochimie 2018; 153:3-12. [DOI: 10.1016/j.biochi.2018.05.004] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2018] [Accepted: 05/04/2018] [Indexed: 12/16/2022]
|
93
|
Niemann-Pick type C disease: The atypical sphingolipidosis. Adv Biol Regul 2018; 70:82-88. [PMID: 30205942 DOI: 10.1016/j.jbior.2018.08.001] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2018] [Accepted: 08/27/2018] [Indexed: 12/29/2022]
Abstract
Niemann-Pick type C (NPC) disease is a lysosomal storage disorder resulting from mutations in either the NPC1 (95%) or NPC2 (5%) genes. NPC typically presents in childhood with visceral lipid accumulation and complex progressive neurodegeneration characterized by cerebellar ataxia, dysphagia, and dementia, resulting in a shortened lifespan. While cholesterol is widely acknowledged as the principal storage lipid in NPC, multiple species of sphingolipids accumulate as well. This accumulation of sphingolipids led to the initial assumption that NPC disease was caused by a deficiency in a sphingolipid catabolism enzyme, similar to sphingomyelinase deficiencies with which it shares a family name. It took about half a century to determine that NPC was in fact caused by a cholesterol trafficking defect, and still as we approach a century after the initial identification of the disease, the mechanisms by which sphingolipids accumulate remain poorly understood. Here we focus on the defects of sphingolipid catabolism in the endolysosomal compartment and how they contribute to the biology and pathology observed in NPC disease. This review highlights the need for further work on understanding and possibly developing treatments to correct the accumulation of sphingolipids in addition to cholesterol in this currently untreatable disease.
Collapse
|
94
|
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: 33] [Impact Index Per Article: 5.5] [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
|
95
|
Pineda M, Walterfang M, Patterson MC. Miglustat in Niemann-Pick disease type C patients: a review. Orphanet J Rare Dis 2018; 13:140. [PMID: 30111334 PMCID: PMC6094874 DOI: 10.1186/s13023-018-0844-0] [Citation(s) in RCA: 106] [Impact Index Per Article: 17.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2018] [Accepted: 06/14/2018] [Indexed: 01/08/2023] Open
Abstract
OBJECTIVE Niemann-Pick disease type C (NP-C) is a rare, autosomal recessive, neurodegenerative disease associated with a wide variety of progressive neurological manifestations. Miglustat is indicated for the treatment of progressive neurological manifestations in both adults and children. Since approval in 2009 there has been a vast growth in clinical experience with miglustat. The effectiveness of miglustat has been assessed using a range of measures. METHODS Comprehensive review of published data from studies of cellular neuropathological markers and structural neurological indices in the brain, clinical impairment/disability, specific clinical neurological manifestations, and patient survival. RESULTS Cranial diffusion tensor imaging and magnetic resonance spectroscopy studies have shown reduced levels of choline (a neurodegeneration marker), and choline/N-acetyl aspartate ratio (indicating increased neuronal viability) in the brain during up to 5 years of miglustat therapy, as well as a slowing of reductions in fractional anisotropy (an axonal/myelin integrity marker). A 2-year immunoassay study showed significant reductions in CSF-calbindin during treatment, indicating reduced cerebellar Purkinje cell loss. Magnetic resonance imaging studies have demonstrated a protective effect of miglustat on cerebellar and subcortical structure that correlated with clinical symptom severity. Numerous cohort studies assessing core neurological manifestations (impaired ambulation, manipulation, speech, swallowing, other) using NP-C disability scales indicate neurological stabilization over 2-8 years, with a trend for greater benefits in patients with older (non-infantile) age at neurological onset. A randomized controlled trial and several cohort studies have reported improvements or stabilization of saccadic eye movements during 1-5 years of therapy. Swallowing was also shown to improve/remain stable during the randomized trial (up to 2 years), as well as in long-term observational cohorts (up to 6 years). A meta-analysis of dysphagia - a potent risk factor for aspiration pneumonia and premature death in NP-C - demonstrated a survival benefit with miglustat due to improved/stabilized swallowing function. CONCLUSIONS The effects of miglustat on neurological NP-C manifestations has been assessed using a range of approaches, with benefits ranging from cellular changes in the brain through to visible clinical improvements and improved survival.
Collapse
Affiliation(s)
- Mercè Pineda
- Fundacio Hospital Sant Joan de Déu, Barcelona, Spain. .,Hospital Sant Joan de Déu, Passeig de Sant Joan de Déu No. 2, Esplugues, 8950, Barcelona, Spain.
| | - Mark Walterfang
- Florey Institute of Neuroscience and Mental Health, Royal Melbourne Hospital, University of Melbourne, Melbourne, Australia
| | | |
Collapse
|
96
|
Lin JB, Sene A, Santeford A, Fujiwara H, Sidhu R, Ligon MM, Shankar VA, Ban N, Mysorekar IU, Ory DS, Apte RS. Oxysterol Signatures Distinguish Age-Related Macular Degeneration from Physiologic Aging. EBioMedicine 2018; 32:9-20. [PMID: 29903570 PMCID: PMC6021272 DOI: 10.1016/j.ebiom.2018.05.035] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2018] [Revised: 05/26/2018] [Accepted: 05/29/2018] [Indexed: 01/16/2023] Open
Abstract
Macrophage aging is pathogenic in numerous diseases, including age-related macular degeneration (AMD), a leading cause of blindness in older adults. Although prior studies have explored the functional consequences of macrophage aging, less is known about its cellular basis or what defines the transition from physiologic aging to disease. Here, we show that despite their frequent self-renewal, macrophages from old mice exhibited numerous signs of aging, such as impaired oxidative respiration. Transcriptomic profiling of aged murine macrophages revealed dysregulation of diverse cellular pathways, especially in cholesterol homeostasis, that manifested in altered oxysterol signatures. Although the levels of numerous oxysterols in human peripheral blood mononuclear cells and plasma exhibited age-associated changes, plasma 24-hydroxycholesterol levels were specifically associated with AMD. These novel findings demonstrate that oxysterol levels can discriminate disease from physiologic aging. Furthermore, modulation of cholesterol homeostasis may be a novel strategy for treating age-associated diseases in which macrophage aging is pathogenic.
Collapse
Affiliation(s)
- Jonathan B Lin
- Department of Ophthalmology & Visual Sciences, Washington University School of Medicine, St. Louis, MO, USA; Neuroscience Graduate Program, Division of Biology and Biomedical Sciences, Washington University School of Medicine, St. Louis, MO, USA
| | - Abdoulaye Sene
- Department of Ophthalmology & Visual Sciences, Washington University School of Medicine, St. Louis, MO, USA
| | - Andrea Santeford
- Department of Ophthalmology & Visual Sciences, Washington University School of Medicine, St. Louis, MO, USA
| | - Hideji Fujiwara
- Diabetic Cardiovascular Disease Center, Washington University School of Medicine, St. Louis, MO, USA; Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Rohini Sidhu
- Diabetic Cardiovascular Disease Center, Washington University School of Medicine, St. Louis, MO, USA; Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Marianne M Ligon
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, USA
| | - Vikram A Shankar
- Department of Ophthalmology & Visual Sciences, Washington University School of Medicine, St. Louis, MO, USA
| | - Norimitsu Ban
- Department of Ophthalmology & Visual Sciences, Washington University School of Medicine, St. Louis, MO, USA
| | - Indira U Mysorekar
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, USA; Center for Reproductive Health Sciences, Department of Obstetrics and Gynecology, Washington University School of Medicine, St. Louis, MO, USA
| | - Daniel S Ory
- Diabetic Cardiovascular Disease Center, Washington University School of Medicine, St. Louis, MO, USA; Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Rajendra S Apte
- Department of Ophthalmology & Visual Sciences, Washington University School of Medicine, St. Louis, MO, USA; Diabetic Cardiovascular Disease Center, Washington University School of Medicine, St. Louis, MO, USA; Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA; Department of Developmental Biology, Washington University School of Medicine, St. Louis, MO, USA.
| |
Collapse
|
97
|
Lütjohann D, Lopez AM, Chuang JC, Kerksiek A, Turley SD. Identification of Correlative Shifts in Indices of Brain Cholesterol Metabolism in the C57BL6/Mecp2 tm1.1Bird Mouse, a Model for Rett Syndrome. Lipids 2018; 53:363-373. [PMID: 29770459 DOI: 10.1002/lipd.12041] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2017] [Revised: 02/26/2018] [Accepted: 04/03/2018] [Indexed: 01/14/2023]
Abstract
Rett syndrome (RS) is a pervasive neurodevelopmental disorder resulting from loss-of-function mutations in the X-linked gene methyl-Cpg-binding protein 2 (MECP2). Using a well-defined model for RS, the C57BL6/Mecp2tm1.1Bird mouse, we have previously found a moderate but persistently lower rate of cholesterol synthesis, measured in vivo, in the brains of Mecp2-/y mice, starting from about the third week after birth. There was no genotypic difference in the total cholesterol concentration throughout the brain at any age. This raised the question of whether the lower rate of cholesterol synthesis in the mutants was balanced by a fall in the rate at which cholesterol was converted via cholesterol 24-hydroxylase (Cyp46A1) to 24S-hydroxycholesterol (24S-OHC), the principal route through which cholesterol is ordinarily removed from the brain. Here, we show that while there were no genotypic differences in the concentrations in plasma and liver of three cholesterol precursors (lanosterol, lathosterol, and desmosterol), two plant sterols (sitosterol and campesterol), and two oxysterols (27-hydroxycholesterol [27-OHC] and 24S-OHC), the brains of the Mecp2 -/y mice had significantly lower concentrations of all three cholesterol precursors, campesterol, and both oxysterols, with the level of 24S-OHC being ~20% less than in their Mecp2 +/y controls. Together, these data suggest that coordinated regulation of cholesterol synthesis and catabolism in the central nervous system is maintained in this model for RS. Furthermore, we speculate that the adaptive changes in these two pathways conceivably resulted from a shift in the permeability of the blood-brain barrier as implied by the significantly lower campesterol and 27-OHC concentrations in the brains of the Mecp2-/y mice.
Collapse
Affiliation(s)
- Dieter Lütjohann
- Institute for Clinical Chemistry and Clinical Pharmacology, University Clinics of Bonn, Sigmund-Freud-Str. 25, 53127 Bonn, Germany
| | - Adam M Lopez
- Department of Internal Medicine, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd., Dallas, TX 75390-9151, USA
| | - Jen-Chieh Chuang
- Children's Medical Center Research Institute, University of Texas Southwestern Medical Center, 6000 Harry Hines Blvd., Dallas, TX 75390-9060, USA
| | - Anja Kerksiek
- Institute for Clinical Chemistry and Clinical Pharmacology, University Clinics of Bonn, Sigmund-Freud-Str. 25, 53127 Bonn, Germany
| | - Stephen D Turley
- Department of Internal Medicine, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd., Dallas, TX 75390-9151, USA
| |
Collapse
|
98
|
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: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 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
|
99
|
Geberhiwot T, Moro A, Dardis A, Ramaswami U, Sirrs S, Marfa MP, Vanier MT, Walterfang M, Bolton S, Dawson C, Héron B, Stampfer M, Imrie J, Hendriksz C, Gissen P, Crushell E, Coll MJ, Nadjar Y, Klünemann H, Mengel E, Hrebicek M, Jones SA, Ory D, Bembi B, Patterson M. Consensus clinical management guidelines for Niemann-Pick disease type C. Orphanet J Rare Dis 2018; 13:50. [PMID: 29625568 PMCID: PMC5889539 DOI: 10.1186/s13023-018-0785-7] [Citation(s) in RCA: 174] [Impact Index Per Article: 29.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2017] [Accepted: 03/13/2018] [Indexed: 01/30/2023] Open
Abstract
Niemann-Pick Type C (NPC) is a progressive and life limiting autosomal recessive disorder caused by mutations in either the NPC1 or NPC2 gene. Mutations in these genes are associated with abnormal endosomal-lysosomal trafficking, resulting in the accumulation of multiple tissue specific lipids in the lysosomes. The clinical spectrum of NPC disease ranges from a neonatal rapidly progressive fatal disorder to an adult-onset chronic neurodegenerative disease. The age of onset of the first (beyond 3 months of life) neurological symptom may predict the severity of the disease and determines life expectancy. NPC has an estimated incidence of ~ 1: 100,000 and the rarity of the disease translate into misdiagnosis, delayed diagnosis and barriers to good care. For these reasons, we have developed clinical guidelines that define standard of care for NPC patients, foster shared care arrangements between expert centres and family physicians, and empower patients. The information contained in these guidelines was obtained through a systematic review of the literature and the experiences of the authors in their care of patients with NPC. We adopted the Appraisal of Guidelines for Research & Evaluation (AGREE II) system as method of choice for the guideline development process. We made a series of conclusive statements and scored them according to level of evidence, strengths of recommendations and expert opinions. These guidelines can inform care providers, care funders, patients and their carers of best practice of care for patients with NPC. In addition, these guidelines have identified gaps in the knowledge that must be filled by future research. It is anticipated that the implementation of these guidelines will lead to a step change in the quality of care for patients with NPC irrespective of their geographical location.
Collapse
Affiliation(s)
- Tarekegn Geberhiwot
- Institute of Metabolism and System Research, University of Birmingham, Birmingham, UK.
| | | | | | | | | | | | - Marie T Vanier
- INSERM U820, Université de Lyon, Faculté de Médecine Lyon-Est, Lyon, 69372, France
| | | | - Shaun Bolton
- University Hospitals Birmingham NHS Foundation Trust, Birmingham, UK
| | - Charlotte Dawson
- University Hospitals Birmingham NHS Foundation Trust, Birmingham, UK
| | - Bénédicte Héron
- Department of Pediatric Neurology, Reference Center of Lysosomal Diseases, Trousseau Hospital, APHP, GRC ConCer-LD, Sorbonne Universities, UPMC University 06, Paris, France
| | - Miriam Stampfer
- Universitatsklinikum Tubingen Institut fur Medizinische Genetik undangewandte Genomik, Tubingen, Germany
| | | | | | - Paul Gissen
- MRC Laboratory for Molecular Cell Biology, London, UK
| | - Ellen Crushell
- Children's University Hospital, Dublin, Republic of Ireland
| | | | - Yann Nadjar
- Hopital Universitaire Pitie Salpetriere, Paris, France
| | - Hans Klünemann
- Universitatsklinikum Regensburg Klinik und Poliklinik fur Chirurgie, Regensburg, Germany
| | | | | | - Simon A Jones
- Central Manchester University Hospitals NHS Foundation Trust, Manchester, UK
| | - Daniel Ory
- University of Washington School of Medicine, Seattle, USA
| | | | - Marc Patterson
- Mayo 1290 Clinic Department of Pediatric and Adolescent Medicine, Minnesota, USA
| | | |
Collapse
|
100
|
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: 3.3] [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
|