1
|
Zhang YD, Ma C, Zheng KW, Han SQ, Ha W, Shi YP. Direct and Rapid Visualization of the Spatial Distribution of Cholesterol in Alzheimer's and Cancer Tissue via MALDI Mass Spectrometry Imaging. JOURNAL OF THE AMERICAN SOCIETY FOR MASS SPECTROMETRY 2024; 35:1756-1767. [PMID: 39001840 DOI: 10.1021/jasms.4c00130] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/15/2024]
Abstract
Cholesterol is a vital component of the central nervous system and tissues, and understanding its spatial distribution is crucial for biology, pathophysiology, and diagnostics. However, direct imaging of cholesterol using matrix-assisted laser desorption/ionization mass spectrometry imaging (MALDI-MSI) remains challenging and time-consuming due to the difficulty in ionizing the sterol molecule. To tackle this issue, a MALDI-MSI method is established for direct and rapid analysis of the spatial distribution of cholesterol in Alzheimer's disease (AD), different cancer tissues and organs via MALDI-MSI. This excellent imaging performance depends on the study and systemic optimization of various conditions that affect the imaging of MALDI-MSI. In this case, we report the distribution and levels of cholesterol across specific structures of the AD mouse brain and different tumor tissue and organs. According to the results, the content of cholesterol in the AD mouse cerebellum, especially in the arborvitae, was significantly higher than that in the wild type (WT) model. Furthermore, we successfully visualize the distribution of cholesterol in other organs, such as the heart, liver, spleen, kidney, pancreas, as well as tumor tissues parenchyma and interstitium using MALDI-MSI. Notably, the attribution of cholesterol MS/MS hydrocarbon fragments was systematically investigated. Our presented optimization strategy and established MALDI-MSI method can be easily generalized for different animal tissues or live samples, thereby facilitating the potential for applications of MALDI-MSI in clinical, medical and biological research.
Collapse
Affiliation(s)
- Yi-Da Zhang
- CAS Key Laboratory of Chemistry of Northwestern Plant Resources, Key Laboratory for Natural Medicine of Gansu Province, Lanzhou Institute of Chemical Physics, Chinese Academy of Sciences (CAS), Lanzhou 730000, P. R. China
| | - Chen Ma
- Gansu Province Key Laboratory of Evidence Science Techniques Research and Application, Gansu University of Political Science and Law, Lanzhou 730070, P. R. China
| | - Kai-Wen Zheng
- CAS Key Laboratory of Chemistry of Northwestern Plant Resources, Key Laboratory for Natural Medicine of Gansu Province, Lanzhou Institute of Chemical Physics, Chinese Academy of Sciences (CAS), Lanzhou 730000, P. R. China
- University of Chinese Academy of Sciences, Beijing 100049, P. R. China
| | - Si-Qi Han
- CAS Key Laboratory of Chemistry of Northwestern Plant Resources, Key Laboratory for Natural Medicine of Gansu Province, Lanzhou Institute of Chemical Physics, Chinese Academy of Sciences (CAS), Lanzhou 730000, P. R. China
- University of Chinese Academy of Sciences, Beijing 100049, P. R. China
| | - Wei Ha
- CAS Key Laboratory of Chemistry of Northwestern Plant Resources, Key Laboratory for Natural Medicine of Gansu Province, Lanzhou Institute of Chemical Physics, Chinese Academy of Sciences (CAS), Lanzhou 730000, P. R. China
| | - Yan-Ping Shi
- CAS Key Laboratory of Chemistry of Northwestern Plant Resources, Key Laboratory for Natural Medicine of Gansu Province, Lanzhou Institute of Chemical Physics, Chinese Academy of Sciences (CAS), Lanzhou 730000, P. R. China
| |
Collapse
|
2
|
Pristner M, Wasinger D, Seki D, Klebermaß-Schrehof K, Berger A, Berry D, Wisgrill L, Warth B. Neuroactive metabolites and bile acids are altered in extremely premature infants with brain injury. Cell Rep Med 2024; 5:101480. [PMID: 38518769 PMCID: PMC11031385 DOI: 10.1016/j.xcrm.2024.101480] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Revised: 10/02/2023] [Accepted: 02/27/2024] [Indexed: 03/24/2024]
Abstract
The gut microbiome is associated with pathological neurophysiological evolvement in extremely premature infants suffering from brain injury. The exact underlying mechanism and its associated metabolic signatures in infants are not fully understood. To decipher metabolite profiles linked to neonatal brain injury, we investigate the fecal and plasma metabolome of samples obtained from a cohort of 51 extremely premature infants at several time points, using liquid chromatography (LC)-high-resolution mass spectrometry (MS)-based untargeted metabolomics and LC-MS/MS-based targeted analysis for investigating bile acids and amidated bile acid conjugates. The data are integrated with 16S rRNA gene amplicon gut microbiome profiles as well as patient cytokine, growth factor, and T cell profiles. We find an early onset of differentiation in neuroactive metabolites between infants with and without brain injury. We detect several bacterially derived bile acid amino acid conjugates in plasma and feces. These results provide insights into the early-life metabolome of extremely premature infants.
Collapse
Affiliation(s)
- Manuel Pristner
- Department of Food Chemistry and Toxicology, University of Vienna, 1090 Vienna, Austria
| | - Daniel Wasinger
- Department of Food Chemistry and Toxicology, University of Vienna, 1090 Vienna, Austria
| | - David Seki
- Center for Microbiology and Environmental Systems Science, Department of Microbiology and Ecosystem Science, Division of Microbial Ecology, University of Vienna, 1090 Vienna, Austria; Joint Microbiome Facility of the Medical University of Vienna and the University of Vienna, 1090 Vienna, Austria
| | - Katrin Klebermaß-Schrehof
- Department of Pediatrics and Adolescent Medicine, Division of Neonatology, Pediatric Intensive Care and Neuropediatrics, Comprehensive Center for Pediatrics, Medical University of Vienna, 1090 Vienna, Austria
| | - Angelika Berger
- Department of Pediatrics and Adolescent Medicine, Division of Neonatology, Pediatric Intensive Care and Neuropediatrics, Comprehensive Center for Pediatrics, Medical University of Vienna, 1090 Vienna, Austria
| | - David Berry
- Center for Microbiology and Environmental Systems Science, Department of Microbiology and Ecosystem Science, Division of Microbial Ecology, University of Vienna, 1090 Vienna, Austria; Joint Microbiome Facility of the Medical University of Vienna and the University of Vienna, 1090 Vienna, Austria
| | - Lukas Wisgrill
- Department of Pediatrics and Adolescent Medicine, Division of Neonatology, Pediatric Intensive Care and Neuropediatrics, Comprehensive Center for Pediatrics, Medical University of Vienna, 1090 Vienna, Austria
| | - Benedikt Warth
- Department of Food Chemistry and Toxicology, University of Vienna, 1090 Vienna, Austria.
| |
Collapse
|
3
|
Neyroud AS, Rolland AD, Lecuyer G, Evrard B, Alary N, Dejucq-Rainsford N, Bujan L, Ravel C, Chalmel F. Sperm DNA methylation dynamics after chemotherapy: a longitudinal study of a patient with testicular germ cell tumor treatment. Andrology 2024; 12:396-409. [PMID: 37354024 DOI: 10.1111/andr.13485] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Revised: 05/26/2023] [Accepted: 06/19/2023] [Indexed: 06/25/2023]
Abstract
BACKGROUND An important issue for young men affected by testicular germ cell tumor (TGCT) is how TGCT and its treatment will affect, transiently or permanently, their future reproductive health. Previous studies have reported that xenobiotics can induce changes on human sperm epigenome and have the potential to promote epigenetic alterations in the offspring. OBJECTIVES Here, we report the first longitudinal DNA methylation profiling of frozen sperm from a TGCT patient before and up to 2 years after a bleomycin, etoposide, and cisplatin (BEP) chemotherapy. MATERIALS AND METHODS A TGCT was diagnosed in a 30-year-old patient. A cryopreservation of spermatozoa was proposed before adjuvant BEP treatment. Semen samples were collected before and after chemotherapy at 6, 9, 12, and 24 months. The DNA methylation status was determined by RRBS to detect DNA differentially methylated regions (DMRs). RESULTS The analysis revealed that among the 74 DMRs showing modified methylation status 6 months after therapy, 17 remained altered 24 months after treatment. We next associated DMRs with differentially methylated genes (DMGs), which were subsequently intersected with loci known to be important or expressed during early development. DISCUSSION AND CONCLUSION The consequences of the cancer treatment on the sperm epigenome during the recovery periods are topical issues of increasing significance as epigenetic modifications to the paternal genome may have deleterious effects on the offspring. The altered methylated status of these DMGs important for early development might modify their expression pattern and thus affect their function during key stages of embryogenesis, potentially leading to developmental disorders or miscarriages.
Collapse
Affiliation(s)
- Anne-Sophie Neyroud
- Univ Rennes, Inserm, EHESP, Irset (Institut de recherche en santé, environnement et travail) - UMR_S 1085, Rennes, France
- CHU de Rennes, Département de Gynécologie Obstétrique Reproduction-CECOS, Rennes, France
| | - Antoine Dominique Rolland
- Univ Rennes, Inserm, EHESP, Irset (Institut de recherche en santé, environnement et travail) - UMR_S 1085, Rennes, France
| | - Gwendoline Lecuyer
- Univ Rennes, Inserm, EHESP, Irset (Institut de recherche en santé, environnement et travail) - UMR_S 1085, Rennes, France
| | - Bertrand Evrard
- Univ Rennes, Inserm, EHESP, Irset (Institut de recherche en santé, environnement et travail) - UMR_S 1085, Rennes, France
| | - Nathan Alary
- Univ Rennes, Inserm, EHESP, Irset (Institut de recherche en santé, environnement et travail) - UMR_S 1085, Rennes, France
| | - Nathalie Dejucq-Rainsford
- Univ Rennes, Inserm, EHESP, Irset (Institut de recherche en santé, environnement et travail) - UMR_S 1085, Rennes, France
| | - Louis Bujan
- Développement Embryonnaire, Fertilité, Environnement (DEFE), UMR Inserm 1203 Université Toulouse 3 et Montpellier, Toulouse, France
- CECOS, Groupe d'activité de médecine de la reproduction, Hôpital Paule de Viguier, CHU Toulouse, Toulouse, France
| | - Célia Ravel
- Univ Rennes, Inserm, EHESP, Irset (Institut de recherche en santé, environnement et travail) - UMR_S 1085, Rennes, France
- CHU de Rennes, Département de Gynécologie Obstétrique Reproduction-CECOS, Rennes, France
| | - Frédéric Chalmel
- Univ Rennes, Inserm, EHESP, Irset (Institut de recherche en santé, environnement et travail) - UMR_S 1085, Rennes, France
| |
Collapse
|
4
|
Liu B, Meng Q, Gao X, Sun H, Xu Z, Wang Y, Zhou H. Lipid and glucose metabolism in senescence. Front Nutr 2023; 10:1157352. [PMID: 37680899 PMCID: PMC10481967 DOI: 10.3389/fnut.2023.1157352] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2023] [Accepted: 08/09/2023] [Indexed: 09/09/2023] Open
Abstract
Senescence is an inevitable biological process. Disturbances in glucose and lipid metabolism are essential features of cellular senescence. Given the important roles of these types of metabolism, we review the evidence for how key metabolic enzymes influence senescence and how senescence-related secretory phenotypes, autophagy, apoptosis, insulin signaling pathways, and environmental factors modulate glucose and lipid homeostasis. We also discuss the metabolic alterations in abnormal senescence diseases and anti-cancer therapies that target senescence through metabolic interventions. Our work offers insights for developing pharmacological strategies to combat senescence and cancer.
Collapse
Affiliation(s)
- Bin Liu
- Department of Urology II, The First Hospital of Jilin University, Changchun, Jilin, China
| | - Qingfei Meng
- Key Laboratory of Pathobiology, Ministry of Education, Jilin University, Changchun, Jilin, China
| | - Xin Gao
- Department of Urology II, The First Hospital of Jilin University, Changchun, Jilin, China
| | - Huihui Sun
- Key Laboratory of Pathobiology, Ministry of Education, Jilin University, Changchun, Jilin, China
| | - Zhixiang Xu
- Key Laboratory of Pathobiology, Ministry of Education, Jilin University, Changchun, Jilin, China
| | - Yishu Wang
- Key Laboratory of Pathobiology, Ministry of Education, Jilin University, Changchun, Jilin, China
| | - Honglan Zhou
- Department of Urology II, The First Hospital of Jilin University, Changchun, Jilin, China
| |
Collapse
|
5
|
Tkemaladze T, Bratland E, Bregvadze K, Shatirishvili T, Tatishvili N, Abzianidze E, Houge G, Douzgou S. MSMO1 deficiency: a potentially partially treatable, ultrarare neurodevelopmental disorder with psoriasiform dermatitis, alopecia and polydactyly. Clin Dysmorphol 2023; 32:97-105. [PMID: 37195326 DOI: 10.1097/mcd.0000000000000461] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/18/2023]
Abstract
MSMO1 deficiency (OMIM #616834) is an ultrarare autosomal recessive disorder of distal cholesterol metabolism with only five cases reported to date. The disorder is caused by missense variants in the MSMO1 gene encoding methylsterol monooxygenase 1, leading to the accumulation of methylsterols. Clinically, MSMO1 deficiency is characterized by growth and developmental delay, often in association with congenital cataracts, microcephaly, psoriasiform dermatitis and immune dysfunction. Treatment with oral and topical cholesterol supplements and statins was reported to improve the biochemical, immunological, and cutaneous findings, supporting a potential treatment following the precision diagnosis of MSMO1 deficiency. We describe two siblings from a consanguineous family presenting with novel clinical features of polydactyly, alopecia and spasticity. Whole-exome sequencing revealed a novel, homozygous c.548A > C, p.(Glu183Ala) variant. Based on previously published treatment algorithms, we initiated a modified dosage regime with systemic cholesterol supplementation, statins and bile acid along with topical application of a cholesterol/statin formulation. This resulted in a marked improvement of psoriasiform dermatitis and some hair growth.
Collapse
Affiliation(s)
- Tinatin Tkemaladze
- Department of Molecular and Medical Genetics, Tbilisi State Medical University
- Department of Child Neurology, M. Iashvili Children's Central Hospital, Tbilisi, Georgia
| | - Eirik Bratland
- Department of Medical Genetics, Haukeland University Hospital
- Department of Clinical Science, University of Bergen, Bergen, Norway
| | - Kakha Bregvadze
- Department of Molecular and Medical Genetics, Tbilisi State Medical University
| | - Teona Shatirishvili
- Department of Child Neurology, M. Iashvili Children's Central Hospital, Tbilisi, Georgia
| | - Nino Tatishvili
- Department of Child Neurology, M. Iashvili Children's Central Hospital, Tbilisi, Georgia
| | - Elene Abzianidze
- Department of Molecular and Medical Genetics, Tbilisi State Medical University
| | - Gunnar Houge
- Department of Medical Genetics, Haukeland University Hospital
- Division of Evolution and Genomic Sciences, School of Biological Sciences, University of Manchester, Manchester, UK
| | - Sofia Douzgou
- Department of Medical Genetics, Haukeland University Hospital
- Division of Evolution and Genomic Sciences, School of Biological Sciences, University of Manchester, Manchester, UK
| |
Collapse
|
6
|
Hill C, Noureldein M, Karkhanis P, Kinning E, Vijay S, Gowda H. First case of desmosterolosis diagnosed by prenatal whole exome sequencing. Am J Med Genet A 2023; 191:859-863. [PMID: 36538928 DOI: 10.1002/ajmg.a.63083] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Revised: 11/15/2022] [Accepted: 12/01/2022] [Indexed: 12/24/2022]
Abstract
Desmosterolosis is a rare autosomal recessive disorder of cholesterol biosynthesis resulting in multiple congenital abnormalities and syndromic intellectual disability. It is caused by defects in DHCR24, the gene encoding 3-β-hydroxysterol-24-reductase (24-dehydrocholesterol reductase), which acts in conversion of cholesterol precursor desmosterol, hence resulting in elevated plasma desmosterol levels. To date, desmosterolosis has been reported in 10 patients. Here we report an eleventh patient with desmosterolosis, and the first one to be diagnosed antenatally. Diagnosis was made on whole exome sequencing after amniocentesis due to complex antenatal abnormalities including cerebellar hypoplasia, microgyria, aortic stenosis, and renal tract abnormalities. Sterol quantitation was subsequently done postnatally, which supported the diagnosis. Although the nonspecific features make desmosterolosis difficult to suspect, we demonstrate that disorders of cholesterol synthesis can be considered as a differential diagnosis antenatally.
Collapse
Affiliation(s)
- Chloe Hill
- Neonatal Unit, Heartlands Hospital, University Hospitals Birmingham, Birmingham, UK
| | - Mona Noureldein
- Neonatal Unit, Heartlands Hospital, University Hospitals Birmingham, Birmingham, UK
| | - Pallavi Karkhanis
- Fetal Medicine, Heartlands Hospital, University Hospitals Birmingham, Birmingham, UK
| | - Esther Kinning
- Clinical Genetics, Birmingham Women's and Children's, Birmingham, UK
| | - Suresh Vijay
- Paediatric Inherited Metabolic Disease, Birmingham Women's and Children's, Birmingham, UK
| | - Harsha Gowda
- Neonatal Unit, Heartlands Hospital, University Hospitals Birmingham, Birmingham, UK
| |
Collapse
|
7
|
Xu SF, Pang ZQ, Fan YG, Zhang YH, Meng YH, Bai CY, Jia MY, Chen YH, Wang ZY, Guo C. Astrocyte-specific loss of lactoferrin influences neuronal structure and function by interfering with cholesterol synthesis. Glia 2022; 70:2392-2408. [PMID: 35946355 DOI: 10.1002/glia.24259] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2021] [Revised: 07/26/2022] [Accepted: 07/29/2022] [Indexed: 11/07/2022]
Abstract
Growing evidence indicates that circulating lactoferrin (Lf) is implicated in peripheral cholesterol metabolism disorders. It has emerged that the distribution of Lf changes in astrocytes of aging brains and those exhibiting neurodegeneration; however, its physiological and/or pathological role remains unknown. Here, we demonstrate that astrocyte-specific knockout of Lf (designated cKO) led to decreased body weight and cognitive abnormalities during early life in mice. Accordingly, there was a reduction in neuronal outgrowth and synaptic structure in cKO mice. Importantly, Lf deficiency in the primary astrocytes led to decreased sterol regulatory element binding protein 2 (Srebp2) activation and cholesterol production, and cholesterol content in cKO mice and/or in astrocytes was restored by exogenous Lf or a Srebp2 agonist. Moreover, neuronal dendritic complexity and total dendritic length were decreased after culture with the culture medium of the primary astrocytes derived from cKO mice and that this decrease was reversed after cholesterol supplementation. Alternatively, these alterations were associated with an activation of AMP-activated protein kinase (AMPK) and inhibition of SREBP2 nuclear translocation. These data suggest that astrocytic Lf might directly or indirectly control in situ cholesterol synthesis, which may be implicated in neurodevelopment and several neurological diseases.
Collapse
Affiliation(s)
- Shuang-Feng Xu
- College of Life and Health Sciences, Institute of Neuroscience, Northeastern University, Shenyang, China
| | - Zhong-Qiu Pang
- College of Life and Health Sciences, Institute of Neuroscience, Northeastern University, Shenyang, China
| | - Yong-Gang Fan
- College of Life and Health Sciences, Institute of Neuroscience, Northeastern University, Shenyang, China
- Health Sciences Institute, Key Laboratory of Major Chronic Diseases of Nervous System, China Medical University, Shenyang, China
| | - Yan-Hui Zhang
- College of Life and Health Sciences, Institute of Neuroscience, Northeastern University, Shenyang, China
- Health Sciences Institute, Key Laboratory of Major Chronic Diseases of Nervous System, China Medical University, Shenyang, China
| | - Yu-Han Meng
- College of Life and Health Sciences, Institute of Neuroscience, Northeastern University, Shenyang, China
| | - Chen-Yang Bai
- College of Life and Health Sciences, Institute of Neuroscience, Northeastern University, Shenyang, China
| | - Meng-Yu Jia
- College of Life and Health Sciences, Institute of Neuroscience, Northeastern University, Shenyang, China
| | - Yan-Hong Chen
- College of Life and Health Sciences, Institute of Neuroscience, Northeastern University, Shenyang, China
| | - Zhan-You Wang
- Health Sciences Institute, Key Laboratory of Major Chronic Diseases of Nervous System, China Medical University, Shenyang, China
| | - Chuang Guo
- College of Life and Health Sciences, Institute of Neuroscience, Northeastern University, Shenyang, China
| |
Collapse
|
8
|
Lu F, Ferriero DM, Jiang X. Cholesterol in Brain Development and Perinatal Brain Injury: More than a Building Block. Curr Neuropharmacol 2022; 20:1400-1412. [PMID: 34766894 PMCID: PMC9881076 DOI: 10.2174/1570159x19666211111122311] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Revised: 07/21/2021] [Accepted: 10/06/2021] [Indexed: 11/22/2022] Open
Abstract
The central nervous system (CNS) is enriched with important classes of lipids, in which cholesterol is known to make up a major portion of myelin sheaths, besides being a structural and functional unit of CNS cell membranes. Unlike in the adult brain, where the cholesterol pool is relatively stable, cholesterol is synthesized and accumulated at the highest rate in the developing brain to meet the needs of rapid brain growth at this stage, which is also a critical period for neuroplasticity. In addition to its biophysical role in membrane organization, cholesterol is crucial for brain development due to its involvement in brain patterning, myelination, neuronal differentiation, and synaptogenesis. Thus any injuries to the immature brain that affect cholesterol homeostasis may have long-term adverse neurological consequences. In this review, we describe the unique features of brain cholesterol biosynthesis and metabolism, cholesterol trafficking between different cell types, and highlight cholesterol-dependent biological processes during brain maturation. We also discuss the association of impaired cholesterol homeostasis with several forms of perinatal brain disorders in term and preterm newborns, including hypoxic-ischemic encephalopathy. Strategies targeting the cholesterol pathways may open new avenues for the diagnosis and treatment of developmental brain injury.
Collapse
Affiliation(s)
- Fuxin Lu
- Departments of Neurology, University of California San Francisco, San Francisco, CA, USA;
| | - Donna M. Ferriero
- Departments of Neurology, University of California San Francisco, San Francisco, CA, USA; ,Departments of Pediatrics, University of California San Francisco, San Francisco, CA, USA
| | - Xiangning Jiang
- Departments of Neurology, University of California San Francisco, San Francisco, CA, USA; ,Address correspondence to this author at the Department of Neurology, University of California, San Francisco, 675 Nelson Rising Lane Room 494, San Francisco, CA 94158, USA; Tel/Fax: 415-502-7285; E-mail:
| |
Collapse
|
9
|
Guidara W, Messedi M, Naifar M, Maalej M, Khrouf W, Grayaa S, Maalej M, Bonnefont-Rousselot D, Lamari F, Ayadi F. Plasma oxysterols in drug-free patients with schizophrenia. J Steroid Biochem Mol Biol 2022; 221:106123. [PMID: 35550868 DOI: 10.1016/j.jsbmb.2022.106123] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Revised: 04/29/2022] [Accepted: 05/05/2022] [Indexed: 11/15/2022]
Abstract
Evidence from clinical, genetic, and medical studies has shown the neuronal developmental disorder aspect of schizophrenia (SZ). Whereas oxysterols are vital factors in neurodevelopment, it is still unknown whether they are involved in the pathophysiology of SZ. The current study aims to explore the profile of oxysterols in plasma, ratio to total cholesterol (Tchol) and the association with clinical factors in patients with SZ. Forty men diagnosed with SZ and forty healthy controls matched for age and sex were included in the study. The ratios of cholestane-3β,5α,6β-triol, 27-hydroxycholesterol (27-OHC) and Cholestanol to Tchol increased in the schizophrenic group compared to controls. However, levels of 24S-hydroxycholesterol (24-OHC) were not significantly different between patients and controls. For the SZ patients, the plasma 24-OHC levels were positively correlated with the positive and negative syndrome total scores (PANSS) but negatively correlated with the Montreal Cognitive Assessment scores (MOCA). Moreover, the ratio Cholestanol to Tchol was negatively correlated with MOCA scores and positively correlated with PANSS general. The binary logistic regression analysis revealed that the ratio Cholestane-3β,5α,6β-triol/TChol could be considered as an independent risk factor for SZ. On the other hand, the receiver's operating characteristics analysis corresponding to potential biomarkers on SZ showed Areas Under the Curve (AUCs) of 82.1%; 69.7% and 77.6% for the ratio of Cholestane-3β,5α,6β-triol/TChol, 27-OHC/TChol and Cholestanol/TChol respectively. The relevance of Cholestane-3β,5α,6β-triol, 27-OHC and Cholestanol assays as biomarkers of this disease deserves further investigation.
Collapse
Affiliation(s)
- Wassim Guidara
- Research Laboratory "Molecular Basis of Human Diseases", LR19ES13, Sfax Medicine School, University of Sfax, Tunisia.
| | - Meriam Messedi
- Research Laboratory "Molecular Basis of Human Diseases", LR19ES13, Sfax Medicine School, University of Sfax, Tunisia
| | - Manel Naifar
- Research Laboratory "Molecular Basis of Human Diseases", LR19ES13, Sfax Medicine School, University of Sfax, Tunisia; Biochemistry Laboratory, Habib Bourguiba Hospital, Sfax, Tunisia
| | - Manel Maalej
- Psychiatry C-department, Hédi Chaker Hospital, Sfax, Tunisia
| | - Walid Khrouf
- Service de Biochimie Métabolique, AP-HP.Sorbonne Université, Hôpitaux Universitaires Pitié-Salpêtrière-Charles Foix, DMU BioGeM, F-75013 Paris, France
| | - Sahar Grayaa
- Research Laboratory "Molecular Basis of Human Diseases", LR19ES13, Sfax Medicine School, University of Sfax, Tunisia
| | - Mohamed Maalej
- Psychiatry C-department, Hédi Chaker Hospital, Sfax, Tunisia
| | - Dominique Bonnefont-Rousselot
- Service de Biochimie Métabolique, AP-HP.Sorbonne Université, Hôpitaux Universitaires Pitié-Salpêtrière-Charles Foix, DMU BioGeM, F-75013 Paris, France; Université de Paris, CNRS, Inserm, UTCBS, F-75006 Paris, France
| | - Foudil Lamari
- Service de Biochimie Métabolique, AP-HP.Sorbonne Université, Hôpitaux Universitaires Pitié-Salpêtrière-Charles Foix, DMU BioGeM, F-75013 Paris, France
| | - Fatma Ayadi
- Research Laboratory "Molecular Basis of Human Diseases", LR19ES13, Sfax Medicine School, University of Sfax, Tunisia; Biochemistry Laboratory, Habib Bourguiba Hospital, Sfax, Tunisia
| |
Collapse
|
10
|
Elhani I, Hentgen V, Grateau G, Georgin-Lavialle S. Neurological manifestations in mevalonate kinase deficiency: A systematic review. Mol Genet Metab 2022; 136:85-93. [PMID: 35525811 DOI: 10.1016/j.ymgme.2022.04.006] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Revised: 04/26/2022] [Accepted: 04/26/2022] [Indexed: 11/22/2022]
Abstract
INTRODUCTION Mevalonate kinase deficiency (MKD) is a monogenic auto-inflammatory disease. Its manifestations range from partial MKD to mevalonic aciduria (MVA). All patients display a periodic fever, and MVA patients additionally exhibit severe neurological involvement. The objective of this work was to describe neurological manifestations of MKD. METHODS A systematic literature review was performed from January 1990 to January 2022. Forty-five patients from 18 case reports and five cohort studies were included in the analysis. RESULTS In cohort studies, the most-reported manifestations were headaches (41%) and fatigue (31%). Serious involvements including ataxia and developmental delay were described less than 1% of patients but 22-31% of case reports. They consistently appeared in the first years of life. Retinal dystrophy was frequently reported (31%) in case reports. Other manifestations, including uveitis, aseptic meningitis, and stroke remained rare. DISCUSSION Severe neurological manifestations are rare in MKD but are responsible for major functional disabilities. They are present at onset and never appear at follow-up of patients with mild MKD. Conversely, headaches and fatigue are frequent symptoms that should be investigated. Visual examinations should be performed on the appearance of visual symptoms. The efficacy of anti-IL-1β therapy on neurological manifestations should be further investigated.
Collapse
Affiliation(s)
- Inès Elhani
- Sorbonne University, Department of Internal Medicine, AP-HP, Hôpital Tenon, Centre de Référence des Maladies Auto-Inflammatoires et des Amyloses Inflammatoire (CEREMAIA), Paris, France; Department of Pediatrics, National Reference Center for Auto-inflammatory Diseases and Amyloidosis, CEREMAIA, Versailles Hospital, Versailles, France
| | - Véronique Hentgen
- Department of Pediatrics, National Reference Center for Auto-inflammatory Diseases and Amyloidosis, CEREMAIA, Versailles Hospital, Versailles, France
| | - Gilles Grateau
- Sorbonne University, Department of Internal Medicine, AP-HP, Hôpital Tenon, Centre de Référence des Maladies Auto-Inflammatoires et des Amyloses Inflammatoire (CEREMAIA), Paris, France
| | - Sophie Georgin-Lavialle
- Sorbonne University, Department of Internal Medicine, AP-HP, Hôpital Tenon, Centre de Référence des Maladies Auto-Inflammatoires et des Amyloses Inflammatoire (CEREMAIA), Paris, France.
| |
Collapse
|
11
|
Garg C, khan H, Kaur A, Singh TG, Sharma VK, Singh SK. Therapeutic Implications of Sonic Hedgehog Pathway in Metabolic Disorders: Novel Target for Effective Treatment. Pharmacol Res 2022; 179:106194. [DOI: 10.1016/j.phrs.2022.106194] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/06/2021] [Revised: 03/24/2022] [Accepted: 03/24/2022] [Indexed: 12/13/2022]
|
12
|
Bharadwaj T, Schrauwen I, Rehman S, Liaqat K, Acharya A, Giese APJ, Nouel-Saied LM, Nasir A, Everard JL, Pollock LM, Zhu S, Bamshad MJ, Nickerson DA, Ali RH, Ullah A, Wali A, Ali G, Santos-Cortez RLP, Ahmed ZM, McDermott BM, Ansar M, Riazuddin S, Ahmad W, Leal SM. ADAMTS1, MPDZ, MVD, and SEZ6: candidate genes for autosomal recessive nonsyndromic hearing impairment. Eur J Hum Genet 2022; 30:22-33. [PMID: 34135477 PMCID: PMC8738740 DOI: 10.1038/s41431-021-00913-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2021] [Revised: 05/12/2021] [Accepted: 05/21/2021] [Indexed: 11/08/2022] Open
Abstract
Hearing impairment (HI) is a common disorder of sensorineural function with a highly heterogeneous genetic background. Although substantial progress has been made in the understanding of the genetic etiology of hereditary HI, many genes implicated in HI remain undiscovered. Via exome and Sanger sequencing of DNA samples obtained from consanguineous Pakistani families that segregate profound prelingual sensorineural HI, we identified rare homozygous missense variants in four genes (ADAMTS1, MPDZ, MVD, and SEZ6) that are likely the underlying cause of HI. Linkage analysis provided statistical evidence that these variants are associated with autosomal recessive nonsyndromic HI. In silico analysis of the mutant proteins encoded by these genes predicted structural, conformational or interaction changes. RNAseq data analysis revealed expression of these genes in the sensory epithelium of the mouse inner ear during embryonic, postnatal, and adult stages. Immunohistochemistry of the mouse cochlear tissue, further confirmed the expression of ADAMTS1, SEZ6, and MPDZ in the neurosensory hair cells of the organ of Corti, while MVD expression was more prominent in the spiral ganglion cells. Overall, supported by in silico mutant protein analysis, animal models, linkage analysis, and spatiotemporal expression profiling in the mouse inner ear, we propose four new candidate genes for HI and expand our understanding of the etiology of HI.
Collapse
Affiliation(s)
- Thashi Bharadwaj
- Center for Statistical Genetics, Gertrude H. Sergievsky Center, and the Department of Neurology, Columbia University Medical Center, New York, NY, USA
| | - Isabelle Schrauwen
- Center for Statistical Genetics, Gertrude H. Sergievsky Center, and the Department of Neurology, Columbia University Medical Center, New York, NY, USA
| | - Sakina Rehman
- Department of Otorhinolaryngology - Head and Neck Surgery, University of Maryland, Baltimore, MD, USA
| | - Khurram Liaqat
- Center for Statistical Genetics, Gertrude H. Sergievsky Center, and the Department of Neurology, Columbia University Medical Center, New York, NY, USA
| | - Anushree Acharya
- Center for Statistical Genetics, Gertrude H. Sergievsky Center, and the Department of Neurology, Columbia University Medical Center, New York, NY, USA
| | - Arnaud P J Giese
- Department of Otorhinolaryngology - Head and Neck Surgery, University of Maryland, Baltimore, MD, USA
| | - Liz M Nouel-Saied
- Center for Statistical Genetics, Gertrude H. Sergievsky Center, and the Department of Neurology, Columbia University Medical Center, New York, NY, USA
| | - Abdul Nasir
- Synthetic Protein Engineering Lab (SPEL), Department of Molecular Science and Technology, Ajou University, Suwon, South Korea
| | - Jenna L Everard
- Center for Statistical Genetics, Gertrude H. Sergievsky Center, and the Department of Neurology, Columbia University Medical Center, New York, NY, USA
| | - Lana M Pollock
- Case Western Reserve University, Department of Otolaryngology, Head and Neck Surgery, Cleveland, OH, USA
| | - Shaoyuan Zhu
- Case Western Reserve University, Department of Otolaryngology, Head and Neck Surgery, Cleveland, OH, USA
| | - Michael J Bamshad
- Department of Genome Sciences, University of Washington, Seattle, WA, USA
- Department of Pediatrics, University of Washington, Seattle, WA, USA
| | | | - Raja Hussain Ali
- Department of Hematology/Oncology, Boston Children's Hospital, Boston, MA, USA
| | - Asmat Ullah
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Abdul Wali
- Department of Biotechnology and Informatics, Faculty of Life Sciences and Informatics, BUITEMS, Quetta, Pakistan
| | - Ghazanfar Ali
- Department of Biotechnology, University of Azad Jammu and Kashmir, Muzaffarabad, Pakistan
| | - Regie Lyn P Santos-Cortez
- Department of Otolaryngology - Head and Neck Surgery, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Zubair M Ahmed
- Department of Otorhinolaryngology - Head and Neck Surgery, University of Maryland, Baltimore, MD, USA
| | - Brian M McDermott
- Case Western Reserve University, Department of Otolaryngology, Head and Neck Surgery, Cleveland, OH, USA
| | - Muhammad Ansar
- Department of Biochemistry, Faculty of Biological Sciences, Quaid-i-Azam University, Islamabad, Pakistan
| | - Saima Riazuddin
- Department of Otorhinolaryngology - Head and Neck Surgery, University of Maryland, Baltimore, MD, USA
| | - Wasim Ahmad
- Department of Biochemistry, Faculty of Biological Sciences, Quaid-i-Azam University, Islamabad, Pakistan
| | - Suzanne M Leal
- Center for Statistical Genetics, Gertrude H. Sergievsky Center, and the Department of Neurology, Columbia University Medical Center, New York, NY, USA.
- Taub Institute for Alzheimer's Disease and the Aging Brain, Columbia University Medical Center, New York, NY, USA.
| |
Collapse
|
13
|
Westra M, Gutierrez Y, MacGillavry HD. Contribution of Membrane Lipids to Postsynaptic Protein Organization. Front Synaptic Neurosci 2021; 13:790773. [PMID: 34887741 PMCID: PMC8649999 DOI: 10.3389/fnsyn.2021.790773] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2021] [Accepted: 11/02/2021] [Indexed: 11/13/2022] Open
Abstract
The precise subsynaptic organization of proteins at the postsynaptic membrane controls synaptic transmission. In particular, postsynaptic receptor complexes are concentrated in distinct membrane nanodomains to optimize synaptic signaling. However, despite the clear functional relevance of subsynaptic receptor organization to synaptic transmission and plasticity, the mechanisms that underlie the nanoscale organization of the postsynaptic membrane remain elusive. Over the last decades, the field has predominantly focused on the role of protein-protein interactions in receptor trafficking and positioning in the synaptic membrane. In contrast, the contribution of lipids, the principal constituents of the membrane, to receptor positioning at the synapse remains poorly understood. Nevertheless, there is compelling evidence that the synaptic membrane is enriched in specific lipid species and that deregulation of lipid homeostasis in neurons severely affects synaptic functioning. In this review we focus on how lipids are organized at the synaptic membrane, with special emphasis on how current models of membrane organization could contribute to protein distribution at the synapse and synaptic transmission. Finally, we will present an outlook on how novel technical developments could be applied to study the dynamic interplay between lipids and proteins at the postsynaptic membrane.
Collapse
Affiliation(s)
- Manon Westra
- Cell Biology, Neurobiology and Biophysics, Department of Biology, Faculty of Science, Utrecht University, Utrecht, Netherlands
| | - Yolanda Gutierrez
- Cell Biology, Neurobiology and Biophysics, Department of Biology, Faculty of Science, Utrecht University, Utrecht, Netherlands
| | - Harold D MacGillavry
- Cell Biology, Neurobiology and Biophysics, Department of Biology, Faculty of Science, Utrecht University, Utrecht, Netherlands
| |
Collapse
|
14
|
Zhou X, Shin S, He C, Zhang Q, Rasband MN, Ren J, Dai C, Zorrilla-Veloz RI, Shingu T, Yuan L, Wang Y, Chen Y, Lan F, Hu J. Qki regulates myelinogenesis through Srebp2-dependent cholesterol biosynthesis. eLife 2021; 10:60467. [PMID: 33942715 PMCID: PMC8139834 DOI: 10.7554/elife.60467] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2020] [Accepted: 05/01/2021] [Indexed: 01/14/2023] Open
Abstract
Myelination depends on timely, precise control of oligodendrocyte differentiation and myelinogenesis. Cholesterol is the most abundant component of myelin and essential for myelin membrane assembly in the central nervous system. However, the underlying mechanisms of precise control of cholesterol biosynthesis in oligodendrocytes remain elusive. In the present study, we found that Qki depletion in neural stem cells or oligodendrocyte precursor cells in neonatal mice resulted in impaired cholesterol biosynthesis and defective myelinogenesis without compromising their differentiation into Aspa+Gstpi+ myelinating oligodendrocytes. Mechanistically, Qki-5 functions as a co-activator of Srebp2 to control transcription of the genes involved in cholesterol biosynthesis in oligodendrocytes. Consequently, Qki depletion led to substantially reduced concentration of cholesterol in mouse brain, impairing proper myelin assembly. Our study demonstrated that Qki-Srebp2-controlled cholesterol biosynthesis is indispensable for myelinogenesis and highlights a novel function of Qki as a transcriptional co-activator beyond its canonical function as an RNA-binding protein.
Collapse
Affiliation(s)
- Xin Zhou
- Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, United States.,Cancer Research Institute of Jilin University, The First Hospital of Jilin University, Changchun, Jilin, China
| | - Seula Shin
- Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, United States.,Cancer Biology Program, MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, Houston, United States
| | - Chenxi He
- Shanghai Key Laboratory of Medical Epigenetics, International Co-laboratory of Medical Epigenetics and Metabolism, Ministry of Science and Technology, Institutes of Biomedical Sciences, Fudan University, and Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Liver Cancer Institute, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Qiang Zhang
- Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, United States
| | - Matthew N Rasband
- Department of Neuroscience, Baylor College of Medicine, Houston, United States
| | - Jiangong Ren
- Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, United States
| | - Congxin Dai
- Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, United States.,Department of Neurosurgery, Beijing Tongren Hospital, Capital Medical University, Beijing, China
| | - Rocío I Zorrilla-Veloz
- Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, United States.,Cancer Biology Program, MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, Houston, United States
| | - Takashi Shingu
- Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, United States
| | - Liang Yuan
- Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, United States.,Graduate School of Biomedical Sciences, Tufts University, Boston, United States
| | - Yunfei Wang
- Clinical Science Division, H. Lee Moffitt Cancer Center & Research Institute, Tampa, United States
| | - Yiwen Chen
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, United States
| | - Fei Lan
- Shanghai Key Laboratory of Medical Epigenetics, International Co-laboratory of Medical Epigenetics and Metabolism, Ministry of Science and Technology, Institutes of Biomedical Sciences, Fudan University, and Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Liver Cancer Institute, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Jian Hu
- Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, United States.,Cancer Biology Program, MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, Houston, United States.,Neuroscience Program, MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, Houston, United States
| |
Collapse
|
15
|
Angelini R, Yutuc E, Wyatt MF, Newton J, Yusuf FA, Griffiths L, Cooze BJ, El Assad D, Frache G, Rao W, Allen LB, Korade Z, Nguyen TTA, Rathnayake RAC, Cologna SM, Howell OW, Clench MR, Wang Y, Griffiths WJ. Visualizing Cholesterol in the Brain by On-Tissue Derivatization and Quantitative Mass Spectrometry Imaging. Anal Chem 2021; 93:4932-4943. [PMID: 33687199 PMCID: PMC7992047 DOI: 10.1021/acs.analchem.0c05399] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
![]()
Despite being a critical
molecule in the brain, mass spectrometry
imaging (MSI) of cholesterol has been under-reported compared to
other lipids due to the difficulty in ionizing the sterol molecule.
In the present work, we have employed an on-tissue enzyme-assisted
derivatization strategy to improve detection of cholesterol in brain
tissue sections. We report distribution and levels of cholesterol
across specific structures of the mouse brain, in a model of Niemann-Pick
type C1 disease, and during brain development. MSI revealed that in
the adult mouse, cholesterol is the highest in the pons and medulla
and how its distribution changes during development. Cholesterol was
significantly reduced in the corpus callosum and other brain regions
in the Npc1 null mouse, confirming hypomyelination
at the molecular level. Our study demonstrates the potential of MSI
to the study of sterols in neuroscience.
Collapse
Affiliation(s)
- Roberto Angelini
- Medical School, Swansea University, Singleton Park, Swansea SA2 8PP, Wales, U.K
| | - Eylan Yutuc
- Medical School, Swansea University, Singleton Park, Swansea SA2 8PP, Wales, U.K
| | - Mark F Wyatt
- Medical School, Swansea University, Singleton Park, Swansea SA2 8PP, Wales, U.K
| | - Jillian Newton
- Centre for Mass Spectrometry Imaging, Biomolecular Research Centre, Sheffield Hallam University, Howard Street, Sheffield S1 1WB, U.K
| | - Fowzi A Yusuf
- Medical School, Swansea University, Singleton Park, Swansea SA2 8PP, Wales, U.K
| | - Lauren Griffiths
- Medical School, Swansea University, Singleton Park, Swansea SA2 8PP, Wales, U.K
| | - Benjamin J Cooze
- Medical School, Swansea University, Singleton Park, Swansea SA2 8PP, Wales, U.K
| | - Dana El Assad
- Materials Research and Technology, Luxembourg Institute of Science and Technology, Belvaux L-4422, Luxembourg
| | - Gilles Frache
- Materials Research and Technology, Luxembourg Institute of Science and Technology, Belvaux L-4422, Luxembourg
| | - Wei Rao
- European Application Laboratory, Waters Corporation, Stamford Avenue, Altrincham Road, Wilmslow SK9 4AX, U.K
| | - Luke B Allen
- Departments of Pediatrics and Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, Nebraska 68198, United States
| | - Zeljka Korade
- Departments of Pediatrics and Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, Nebraska 68198, United States
| | - Thu T A Nguyen
- Department of Chemistry and Laboratory of Integrated Neuroscience, University of Illinois at Chicago, Chicago, Illinois 60607, United States
| | - Rathnayake A C Rathnayake
- Department of Chemistry and Laboratory of Integrated Neuroscience, University of Illinois at Chicago, Chicago, Illinois 60607, United States
| | - Stephanie M Cologna
- Department of Chemistry and Laboratory of Integrated Neuroscience, University of Illinois at Chicago, Chicago, Illinois 60607, United States
| | - Owain W Howell
- Medical School, Swansea University, Singleton Park, Swansea SA2 8PP, Wales, U.K
| | - Malcolm R Clench
- Centre for Mass Spectrometry Imaging, Biomolecular Research Centre, Sheffield Hallam University, Howard Street, Sheffield S1 1WB, U.K
| | - Yuqin Wang
- Medical School, Swansea University, Singleton Park, Swansea SA2 8PP, Wales, U.K
| | - William J Griffiths
- Medical School, Swansea University, Singleton Park, Swansea SA2 8PP, Wales, U.K
| |
Collapse
|
16
|
A precise and rapid isotopomic analysis of small quantities of cholesterol at natural abundance by optimized 1H- 13C 2D NMR. Anal Bioanal Chem 2021; 413:1521-1532. [PMID: 33506339 DOI: 10.1007/s00216-020-03135-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2020] [Revised: 12/15/2020] [Accepted: 12/16/2020] [Indexed: 10/22/2022]
Abstract
Cholesterol, the principal zoosterol, is a key metabolite linked to several health complications. Studies have shown its potential as a metabolic biomarker for predicting various diseases and determining food origin. However, the existing INEPT (insensitive nuclei enhanced by polarization transfer) 13C position-specific isotope analysis method of cholesterol by NMR was not suitable for very precise analysis of small quantities due to its long acquisition time and therefore is restricted to products rich in cholesterol. In this work, a symmetric and adiabatic heteronuclear single quantum coherence (HSQC) 2D NMR sequence was developed for the high-precision (few permil) analysis of small quantities of cholesterol. Adiabatic pulses were incremented for improving precision and sensitivity. Moreover, several strategies such as the use of non-uniform sampling, linear prediction, and variable recycling time were optimized to reduce the acquisition time. The number of increments and spectral range were also adjusted. The method was developed on a system with a cryogenically cooled probe and was not tested on a room-temperature system. Our new approach allowed analyzing as low as 5 mg of cholesterol in 31 min with a long-term repeatability lower than 2‰ on the 24 non-quaternary carbon atoms of the molecule comparing to 16.2 h for the same quantity using the existing INEPT method. This result makes conceivable the isotope analysis of matrices low in cholesterol. Graphical abstract.
Collapse
|
17
|
Sun Z, Zhao L, Bo Q, Mao Z, He Y, Jiang T, Li Y, Wang C, Li R. Brain-Specific Oxysterols and Risk of Schizophrenia in Clinical High-Risk Subjects and Patients With Schizophrenia. Front Psychiatry 2021; 12:711734. [PMID: 34408685 PMCID: PMC8367079 DOI: 10.3389/fpsyt.2021.711734] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/19/2021] [Accepted: 07/05/2021] [Indexed: 01/19/2023] Open
Abstract
Accumulating evidence from clinical, genetic, and epidemiologic studies suggest that schizophrenia might be a neuronal development disorder. While oxysterols are important factors in neurodevelopment, it is unknown whether oxysterols might be involved in development of schizophrenia. The present study investigated the relationship between tissue-specifically originated oxysterols and risk of schizophrenia. A total of 216 individuals were recruited in this study, including 76 schizophrenia patients, 39 clinical high-risk (CHR) subjects, and 101 healthy controls (HC). We investigated the circulating levels of brain-specific oxysterol 24(S)-hydroxycholesterol (24OHC) and peripheral oxysterol 27-hydroxycholesterol (27OHC) in all participants and analyzed the potential links between the oxysterols and specific clinical symptoms in schizophrenic patients and CHR. Our data showed an elevation of 24OHC in both schizophrenia patients and CHR than that in HC, while a lower level of 27OHC in the schizophrenia group only. The ratio of 24OHC to 27OHC was only increased in the schizophrenic group compared with CHR and HC. For the schizophrenic patients, the circulating 24OHC levels are significantly associated with disease duration, positively correlated with the positive and negative syndrome total scores, while the 27OHC levels were inversely correlated with the positive symptom scores. Together, our data demonstrated the disruption of tissue-specifically originated cholesterol metabolism in schizophrenia and CHR, suggesting the circulating 24OHC or 24OHC/27OHC ratio might not only be a potential indicator for risk for schizophrenia but also be biomarkers for functional abnormalities in neuropathology of schizophrenia.
Collapse
Affiliation(s)
- Zuoli Sun
- The National Clinical Research Center for Mental Disorders and Beijing Key Laboratory of Mental Disorders, Beijing Anding Hospital, Capital Medical University, Beijing, China
| | - Lei Zhao
- The National Clinical Research Center for Mental Disorders and Beijing Key Laboratory of Mental Disorders, Beijing Anding Hospital, Capital Medical University, Beijing, China
| | - Qijing Bo
- The National Clinical Research Center for Mental Disorders and Beijing Key Laboratory of Mental Disorders, Beijing Anding Hospital, Capital Medical University, Beijing, China
| | - Zhen Mao
- The National Clinical Research Center for Mental Disorders and Beijing Key Laboratory of Mental Disorders, Beijing Anding Hospital, Capital Medical University, Beijing, China
| | - Yi He
- The National Clinical Research Center for Mental Disorders and Beijing Key Laboratory of Mental Disorders, Beijing Anding Hospital, Capital Medical University, Beijing, China
| | - Tao Jiang
- The National Clinical Research Center for Mental Disorders and Beijing Key Laboratory of Mental Disorders, Beijing Anding Hospital, Capital Medical University, Beijing, China
| | - Yuhong Li
- The National Clinical Research Center for Mental Disorders and Beijing Key Laboratory of Mental Disorders, Beijing Anding Hospital, Capital Medical University, Beijing, China.,Beijing Institute for Brain Disorders, Capital Medical University, Beijing, China
| | - Chuanyue Wang
- The National Clinical Research Center for Mental Disorders and Beijing Key Laboratory of Mental Disorders, Beijing Anding Hospital, Capital Medical University, Beijing, China.,Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing, China
| | - Rena Li
- The National Clinical Research Center for Mental Disorders and Beijing Key Laboratory of Mental Disorders, Beijing Anding Hospital, Capital Medical University, Beijing, China.,Beijing Institute for Brain Disorders, Capital Medical University, Beijing, China
| |
Collapse
|
18
|
Puthanveetil P, Kong X, Bräse S, Voros G, Peer WA. Transcriptome analysis of two structurally related flavonoids; Apigenin and Chrysin revealed hypocholesterolemic and ketogenic effects in mouse embryonic fibroblasts. Eur J Pharmacol 2020; 893:173804. [PMID: 33347826 DOI: 10.1016/j.ejphar.2020.173804] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Revised: 12/02/2020] [Accepted: 12/10/2020] [Indexed: 12/14/2022]
Abstract
There is no known single therapeutic drug for treating hypercholesterolemia that comes with negligible systemic side effects. In the current study, using next generation RNA sequencing approach in mouse embryonic fibroblasts we discovered that two structurally related flavonoid compounds. Apigenin and Chrysin exhibited moderate blocking ability of multiple transcripts that regulate rate limiting enzymes in the cholesterol biosynthesis pathway. The observed decrease in cholesterol biosynthesis pathway correlated well with an increase in transcripts involved in generation and trafficking of ketone bodies as evident by the upregulation of Bdh1 and Slc16a6 transcripts. The hypocholesterolemic potential of Apigenin and Chrysin at higher concentrations along with their ability to generate ketogenic substrate especially during embryonic stage is useful or detrimental for embryonic health is not clear and still debatable. Our study will serve as a steppingstone to further the investigation in whole animal studies and also in translating this knowledge to human studies.
Collapse
Affiliation(s)
- Prasanth Puthanveetil
- Department of Pharmacology, College of Graduate Studies, Midwestern University, Downers Grove, IL, USA.
| | - Xiaoli Kong
- Department of Mathematics and Statistics, Loyola University Chicago, Chicago, IL, USA.
| | - Stefan Bräse
- Institute of Biological and Chemical Systems (IBCS), Karlsruhe Institute of Technology (KIT), Hermann von Helmholtz Platz 1, 76344, Eggenstein Leopoldshafen, Germany.
| | - Gabor Voros
- Department of Cardiovascular Diseases, University Hospital Gasthuisberg, Catholic University Leuven, Herestraat 49, 3000, Leuven, Belgium.
| | - Wendy Ann Peer
- Department of Environmental Science and Technology, College of Agricultural and Natural Resources, University of Maryland, MD, USA.
| |
Collapse
|
19
|
Peterson SJ, Choudhary A, Kalsi AK, Zhao S, Alex R, Abraham NG. OX-HDL: A Starring Role in Cardiorenal Syndrome and the Effects of Heme Oxygenase-1 Intervention. Diagnostics (Basel) 2020; 10:E976. [PMID: 33233550 PMCID: PMC7699797 DOI: 10.3390/diagnostics10110976] [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: 11/03/2020] [Revised: 11/17/2020] [Accepted: 11/19/2020] [Indexed: 12/14/2022] Open
Abstract
In this review, we will evaluate how high-density lipoprotein (HDL) and the reverse cholesterol transport (RCT) pathway are critical for proper cardiovascular-renal physiology. We will begin by reviewing the basic concepts of HDL cholesterol synthesis and pathway regulation, followed by cardiorenal syndrome (CRS) pathophysiology. After explaining how the HDL and RCT pathways become dysfunctional through oxidative processes, we will elaborate on the potential role of HDL dysfunction in CRS. We will then present findings on how HDL function and the inducible antioxidant gene heme oxygenase-1 (HO-1) are interconnected and how induction of HO-1 is protective against HDL dysfunction and important for the proper functioning of the cardiovascular-renal system. This will substantiate the proposal of HO-1 as a novel therapeutic target to prevent HDL dysfunction and, consequently, cardiovascular disease, renal dysfunction, and the onset of CRS.
Collapse
Affiliation(s)
- Stephen J. Peterson
- Department of Medicine, Weill Cornell Medicine, New York, NY 10065, USA;
- Department of Medicine, New York Presbyterian Brooklyn Methodist Hospital, Brooklyn, NY 11215, USA; (A.C.); (A.K.K.); (S.Z.)
| | - Abu Choudhary
- Department of Medicine, New York Presbyterian Brooklyn Methodist Hospital, Brooklyn, NY 11215, USA; (A.C.); (A.K.K.); (S.Z.)
| | - Amardeep K. Kalsi
- Department of Medicine, New York Presbyterian Brooklyn Methodist Hospital, Brooklyn, NY 11215, USA; (A.C.); (A.K.K.); (S.Z.)
| | - Shuyang Zhao
- Department of Medicine, New York Presbyterian Brooklyn Methodist Hospital, Brooklyn, NY 11215, USA; (A.C.); (A.K.K.); (S.Z.)
| | - Ragin Alex
- Department of Medicine, New York Medical College, Valhalla, NY 10595, USA;
| | - Nader G. Abraham
- Department of Medicine, New York Medical College, Valhalla, NY 10595, USA;
- Department of Pharmacology, New York Medical College, Valhalla, NY 10595, USA
- Department of Medicine, Joan C. Edwards School of Medicine, Marshall University, Huntington, WV 25701, USA
| |
Collapse
|
20
|
Kalay Yildizhan I, Gökpınar İli E, Onoufriadis A, Kocyigit P, Kesidou E, Simpson MA, McGrath JA, Kutlay NY, Kundakci N. New Homozygous Missense MSMO1 Mutation in Two Siblings with SC4MOL Deficiency Presenting with Psoriasiform Dermatitis. Cytogenet Genome Res 2020; 160:523-530. [PMID: 33161406 DOI: 10.1159/000511126] [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] [Received: 07/02/2020] [Accepted: 08/10/2020] [Indexed: 11/19/2022] Open
Abstract
Sterol-C4-methyl oxidase (SC4MOL) deficiency was recently described as an autosomal recessive cholesterol biosynthesis disorder caused by mutations in the MSMO1 (sometimes also referred to as SC4MOL) gene. To date, 5 patients from 4 unrelated families with SC4MOL deficiency have been reported. Diagnosis can be challenging as the biochemical accumulation of methylsterols can affect global development and cause skin and ocular pathology. Herein, we describe 2 siblings from a consanguineous Turkish family with SC4MOL deficiency presenting with psoriasiform dermatitis, ocular abnormalities (nystagmus, optic hypoplasia, myopia, and strabismus), severe intellectual disability, and growth and motor delay. We undertook whole-exome sequencing and identified a new homozygous missense mutation c.81A>C; p.Asn27Thr in MSMO1. Segregation analysis in all available family members confirmed recessive inheritance of the mutation. The siblings were treated with a combination of oral and topical statin and cholesterol which resulted in clinical improvement. This study demonstrates how genomics-based diagnosis and therapy can be helpful in clinical practice.
Collapse
Affiliation(s)
| | - Ezgi Gökpınar İli
- Department of Medical Genetics, School of Medicine, Ankara University, Ankara, Turkey.,Genetic Diseases Center, Başakşehir Pine and Sakura City Hospital, Istanbul, Turkey
| | - Alexandros Onoufriadis
- St John's Institute of Dermatology, School of Basic and Medical Biosciences, King's College London, London, United Kingdom
| | - Pelin Kocyigit
- Department of Dermatology, School of Medicine, Ankara University, Ankara, Turkey
| | - Evangelia Kesidou
- St John's Institute of Dermatology, School of Basic and Medical Biosciences, King's College London, London, United Kingdom
| | - Michael A Simpson
- Department of Medical and Molecular Genetics, School of Basic and Medical Biosciences, King's College London, London, United Kingdom
| | - John A McGrath
- St John's Institute of Dermatology, School of Basic and Medical Biosciences, King's College London, London, United Kingdom
| | - Nüket Yürür Kutlay
- Department of Medical Genetics, School of Medicine, Ankara University, Ankara, Turkey
| | - Nihal Kundakci
- Department of Dermatology, School of Medicine, Ankara University, Ankara, Turkey
| |
Collapse
|
21
|
Ritz B, Yan Q, Uppal K, Liew Z, Cui X, Ling C, Inoue K, von Ehrenstein O, Walker DI, Jones DP. Untargeted Metabolomics Screen of Mid-pregnancy Maternal Serum and Autism in Offspring. Autism Res 2020; 13:1258-1269. [PMID: 32496662 DOI: 10.1002/aur.2311] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2019] [Revised: 03/24/2020] [Accepted: 04/15/2020] [Indexed: 12/15/2022]
Abstract
Discovering pathophysiologic networks in a blood-based approach may help to generate valuable tools for early treatment or preventive measures in autism. To date targeted or untargeted metabolomics approaches to identify metabolic features and pathways affecting fetal neurodevelopment have rarely been applied to pregnancy samples, that is, an early period potentially relevant for the development of autism spectrum disorders (ASD). We conducted a population-based study relying on autism diagnoses retrieved from California Department of Developmental Services record. After linking cases to and sampling controls from birth certificates, we retrieved stored maternal mid-pregnancy serum samples collected as part of the California Prenatal Screening Program from the California Biobank for children born 2004 to 2010 in the central valley of California. We retrieved serum for 52 mothers whose children developed autism and 62 population controls originally selected from all eligible children matched by birth year and child's sex. Also, we required that these mothers were relatively low or unexposed to air pollution and select pesticides during early pregnancy. We identified differences in metabolite levels in several metabolic pathways, including glycosphingolipid biosynthesis and metabolism, N-glycan and pyrimidine metabolism, bile acid pathways and, importantly, C21-steroid hormone biosynthesis and metabolism. Disturbances in these pathways have been shown to be relevant for neurodevelopment in rare genetic syndromes or implicated in previous studies of autism. This study provides new insight into maternal mid-pregnancy metabolic features possibly related to the development of autism and an incentive to explore whether these pathways and metabolites are useful for early diagnosis, treatment, or prevention. LAY SUMMARY: This study found that in mid-pregnancy the blood of mothers who give birth to a child that develops autism has some characteristic features that are different from those of blood samples taken from control mothers. These features are related to biologic mechanisms that can affect fetal brain development. In the future, these insights may help identify biomarkers for early autism diagnosis and treatment or preventive measures. Autism Res 2020, 13: 1258-1269. © 2020 International Society for Autism Research, Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Beate Ritz
- Department of Epidemiology, UCLA Fielding School of Public Health, Los Angeles, California, USA.,Department of Neurology, UCLA School of Medicine, Los Angeles, California, USA
| | - Qi Yan
- Department of Epidemiology, UCLA Fielding School of Public Health, Los Angeles, California, USA
| | - Karan Uppal
- Clinical Biomarkers Laboratory, Division of Pulmonary, Allergy, and Critical Care Medicine, School of Medicine, Emory University, Atlanta, Georgia, USA
| | - Zeyan Liew
- Department of Environmental Health Sciences, Yale School of Public Health, New Haven, Connecticut, USA.,Yale Center for Perinatal, Pediatric, and Environmental Epidemiology, Yale School of Public Health, New Haven, Connecticut, USA
| | - Xin Cui
- Perinatal Epidemiology and Health Outcomes Research Unit, Division of Neonatology, Department of Pediatrics, Stanford University School of Medicine and Lucile Packard Children's Hospital, Palo Alto, California, USA.,California Perinatal Quality Care Collaborative, Palo Alto, California, USA
| | - Chenxiao Ling
- Department of Epidemiology, UCLA Fielding School of Public Health, Los Angeles, California, USA
| | - Kosuke Inoue
- Department of Epidemiology, UCLA Fielding School of Public Health, Los Angeles, California, USA
| | - Ondine von Ehrenstein
- Department of Epidemiology, UCLA Fielding School of Public Health, Los Angeles, California, USA
| | - Douglas I Walker
- Department of Environmental Medicine and Public Health, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Dean P Jones
- Clinical Biomarkers Laboratory, Division of Pulmonary, Allergy, and Critical Care Medicine, School of Medicine, Emory University, Atlanta, Georgia, USA
| |
Collapse
|
22
|
Localization of sterols and oxysterols in mouse brain reveals distinct spatial cholesterol metabolism. Proc Natl Acad Sci U S A 2020; 117:5749-5760. [PMID: 32132201 PMCID: PMC7084107 DOI: 10.1073/pnas.1917421117] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
The brain is a remarkably complex organ and cholesterol homeostasis underpins brain function. It is known that cholesterol is not evenly distributed across different brain regions; however, the precise map of cholesterol metabolism in the brain remains unclear. If cholesterol metabolism is to be correlated with brain function it is essential to generate such a map. Here we describe an advanced mass spectrometry platform to reveal spatial cholesterol metabolism in situ at 400-µm spot diameter on 10-µm tissue slices from mouse brain. We mapped, not only cholesterol, but also other biologically active sterols arising from cholesterol turnover in both wild type and mice lacking cholesterol 24S-hydroxylase (CYP46A1), the major cholesterol metabolizing enzyme. Dysregulated cholesterol metabolism is implicated in a number of neurological disorders. Many sterols, including cholesterol and its precursors and metabolites, are biologically active and important for proper brain function. However, spatial cholesterol metabolism in brain and the resulting sterol distributions are poorly defined. To better understand cholesterol metabolism in situ across the complex functional regions of brain, we have developed on-tissue enzyme-assisted derivatization in combination with microliquid extraction for surface analysis and liquid chromatography-mass spectrometry to locate sterols in tissue slices (10 µm) of mouse brain. The method provides sterolomic analysis at 400-µm spot diameter with a limit of quantification of 0.01 ng/mm2. It overcomes the limitations of previous mass spectrometry imaging techniques in analysis of low-abundance and difficult-to-ionize sterol molecules, allowing isomer differentiation and structure identification. Here we demonstrate the spatial distribution and quantification of multiple sterols involved in cholesterol metabolic pathways in wild-type and cholesterol 24S-hydroxylase knockout mouse brain. The technology described provides a powerful tool for future studies of spatial cholesterol metabolism in healthy and diseased tissues.
Collapse
|
23
|
Kumar GA, Chattopadhyay A. Statin-Induced Chronic Cholesterol Depletion Switches GPCR Endocytosis and Trafficking: Insights from the Serotonin 1A Receptor. ACS Chem Neurosci 2020; 11:453-465. [PMID: 31880914 DOI: 10.1021/acschemneuro.9b00659] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
Endocytosis is a key regulatory mechanism adopted by G protein-coupled receptors (GPCRs) to modulate downstream signaling responses within a stringent spatiotemporal regime. Although the role of membrane lipids has been extensively studied in the context of the function, organization, and dynamics of GPCRs, their role in receptor endocytosis remains largely unexplored. Cholesterol, the predominant sterol in higher eukaryotes, plays a crucial role in maintaining the structure and organization of cell membranes and is involved in essential cellular processes in health and disease. The serotonin1A receptor is a representative GPCR involved in neuronal development and in neuropsychiatric disorders such as anxiety and depression. We recently combined quantitative flow cytometric and confocal microscopic approaches to demonstrate that the serotonin1A receptor undergoes clathrin-mediated endocytosis upon agonist stimulation and subsequently traffics along the endosomal recycling pathway. In this work, we show that statin-induced chronic cholesterol depletion switches the endocytic pathway of the serotonin1A receptor from clathrin- to caveolin-mediated endocytosis. Interestingly, under these conditions, a significant proportion of endocytosed receptors is rerouted toward lysosomal degradation. To the best of our knowledge, these results constitute one of the first comprehensive reports on the role of membrane cholesterol in GPCR endocytosis and trafficking. These results are significant in our overall understanding of the modulatory effects of membrane lipids on GPCR endocytosis and trafficking and could provide novel insight in developing therapeutic interventions against neuropsychiatric disorders such as depression.
Collapse
Affiliation(s)
- G. Aditya Kumar
- CSIR-Centre for Cellular and Molecular Biology, Uppal Road, Hyderabad 500 007, India
| | | |
Collapse
|
24
|
Zeng B, Zhao G, Liu HL. The Differential Effect of Treadmill Exercise Intensity on Hippocampal Soluble Aβ and Lipid Metabolism in APP/PS1 Mice. Neuroscience 2020; 430:73-81. [PMID: 31954827 DOI: 10.1016/j.neuroscience.2020.01.005] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2019] [Revised: 01/02/2020] [Accepted: 01/06/2020] [Indexed: 12/22/2022]
Abstract
Alzheimer's disease (AD) is characterized clinically by progressive impairments in learning and memory. Accumulating evidence suggests that regular exercise plays a neuroprotective role in aging-associated memory loss. Our previous study has confirmed that long-term treadmill exercise initiated either before or during the onset of β-amyloid (Aβ) pathology, was beneficial for reducing the levels of soluble Aβ and further improved cognition. In this study, in APP/PS1 mice, we assessed changes in soluble Aβ, and various blood biochemistry and molecular biological indices to assess whether exercise modulated lipid metabolism and thereby decelerated AD progression. Our results show that long-term treadmill exercise reduced the total cholesterol, triglyceride, and low-density lipoprotein cholesterol levels, and increased the level of high-density lipoprotein cholesterol. Exercise also decreased the levels of soluble Aβ1-40 and Aβ1-42, down-regulated retinoid X receptor expression, and up-regulated liver X receptor, Apolipoprotein E, Low density lipoprotein receptor, Low density lipoprotein receptor-related protein 1, and ATP-binding cassette transporter A1 expression. This indicates that long-term treadmill exercise alters the lipoprotein content, increases lipid metabolism and cholesterol transportation, reduces the soluble Aβ, and therein plays an important neuroprotective role and delays AD progression. We further show that medium exercise intensity (60%-70% of maximal oxygen uptake) was more efficacious in increasing lipid metabolism and reducing blood lipid levels and soluble Aβ levels, than low-intensity exercise (45-55% of maximal oxygen uptake). This research has broad prospects and implications, and offers a theoretical basis for the prevention of AD.
Collapse
Affiliation(s)
- B Zeng
- Department of Sports Medicine, China Medical University, Shenyang 110122, PR China
| | - G Zhao
- Department of Sports Medicine, China Medical University, Shenyang 110122, PR China
| | - H L Liu
- Department of Sports Medicine, China Medical University, Shenyang 110122, PR China.
| |
Collapse
|
25
|
Pecks U, Bornemann V, Klein A, Segger L, Maass N, Alkatout I, Eckmann-Scholz C, Elessawy M, Lütjohann D. Estimating fetal cholesterol synthesis rates by cord blood analysis in intrauterine growth restriction and normally grown fetuses. Lipids Health Dis 2019; 18:185. [PMID: 31653257 PMCID: PMC6815065 DOI: 10.1186/s12944-019-1117-1] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2019] [Accepted: 09/12/2019] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Cholesterol is an essential component in human development. In fetuses affected by intrauterine growth restriction (IUGR), fetal blood cholesterol levels are low. Whether this is the result of a reduced materno-fetal cholesterol transport, or due to low fetal de novo synthesis rates, remains a matter of debate. By analyzing cholesterol interbolites and plant sterols we aimed at deeper insights into transplacental cholesterol transport and fetal cholesterol handling in IUGR with potential targets for future therapy. We hypothesized that placental insufficiency results in a diminished cholesterol supply to the fetus. METHODS Venous umbilical cord sera were sampled post-partum from fetuses delivered between 24 weeks of gestation and at full term. IUGR fetuses were matched to 49 adequate-for-age delivered preterm and term neonates (CTRL) according to gestational age at delivery. Cholesterol was measured by gas chromatography-flame ionization detection using 5a-cholestane as internal standard. Cholesterol precursors and synthesis markers, such as lanosterol, lathosterol, and desmosterol, the absorption markers, 5α-cholestanol and plant sterols, such as campesterol and sitosterol, as well as enzymatically oxidized cholesterol metabolites (oxysterols), such as 24S- or 27-hydroxycholesterol, were analyzed by gas chromatography-mass spectrometry, using epicoprostanol as internal standard for the non-cholesterol sterols and deuterium labeled oxysterols for 24S- and 27-hydroxycholesterol. RESULTS Mean cholesterol levels were 25% lower in IUGR compared with CTRL (p < 0.0001). Lanosterol and lathosterol to cholesterol ratios were similar in IUGR and CTRL. In relation to cholesterol mean, desmosterol, 24S-hydroxycholesterol, and 27-hydroxycholesterol levels were higher by 30.0, 39.1 and 60.7%, respectively, in IUGR compared to CTRL (p < 0.0001). Equally, 5α-cholestanol, campesterol, and β-sitosterol to cholesterol ratios were higher in IUGR than in CTRL (17.2%, p < 0.004; 33.5%, p < 0.002; 29.3%, p < 0.021). CONCLUSIONS Cholesterol deficiency in IUGR is the result of diminished fetal de novo synthesis rates rather than diminished maternal supply. However, increased oxysterol- and phytosterol to cholesterol ratios suggest a lower sterol elimination rate. This is likely caused by a restricted hepatobiliary function. Understanding the fetal cholesterol metabolism is important, not only for neonatal nutrition, but also for the development of strategies to reduce the known risk of future cardiovascular diseases in the IUGR fetus.
Collapse
Affiliation(s)
- Ulrich Pecks
- Department of Obstetrics and Gynecology, University Hospital of Schleswig-Holstein Campus Kiel, Arnold-Heller-Straße 3, 24105, Kiel, Germany.
| | - Verena Bornemann
- Department of Obstetrics and Gynecology, University Hospital of Schleswig-Holstein Campus Kiel, Arnold-Heller-Straße 3, 24105, Kiel, Germany
| | - Anika Klein
- Department of Obstetrics and Gynecology, University Hospital of Schleswig-Holstein Campus Kiel, Arnold-Heller-Straße 3, 24105, Kiel, Germany
| | - Laura Segger
- Department of Obstetrics and Gynecology, University Hospital of Schleswig-Holstein Campus Kiel, Arnold-Heller-Straße 3, 24105, Kiel, Germany
| | - Nicolai Maass
- Department of Obstetrics and Gynecology, University Hospital of Schleswig-Holstein Campus Kiel, Arnold-Heller-Straße 3, 24105, Kiel, Germany
| | - Ibrahim Alkatout
- Department of Obstetrics and Gynecology, University Hospital of Schleswig-Holstein Campus Kiel, Arnold-Heller-Straße 3, 24105, Kiel, Germany
| | - Christel Eckmann-Scholz
- Department of Obstetrics and Gynecology, University Hospital of Schleswig-Holstein Campus Kiel, Arnold-Heller-Straße 3, 24105, Kiel, Germany
| | - Mohamed Elessawy
- Department of Obstetrics and Gynecology, University Hospital of Schleswig-Holstein Campus Kiel, Arnold-Heller-Straße 3, 24105, Kiel, Germany
| | - Dieter Lütjohann
- Institute for Clinical Chemistry and Clinical Pharmacology, University Clinics of Bonn, Bonn, Germany
| |
Collapse
|
26
|
BoAli AY, Alfadhel M, Tabarki B. Neurometabolic disorders and congenital malformations of the central nervous system. ACTA ACUST UNITED AC 2019; 23:97-103. [PMID: 29664449 PMCID: PMC8015440 DOI: 10.17712/nsj.2018.2.20170481] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Both malformations of the central nervous system and neurometabolic disorders are common, mainly in highly consanguineous populations. Both metabolic pathways and developmental pathways are closely related and interact with each other. Neurometabolic disorders can lead to disturbances in brain development through multiple mechanisms that include deficits in energy metabolism, critical nutrient deficiency, accumulation of neurotoxic substrates, abnormality in cell membrane constituents, and interference in cell-to-cell signaling pathways. The anomalies observed include absent or hypoplastic corpus callosum, midline brain defects, and malformations of the cortex, the cerebellum and the brain stem. Early diagnosis of an underlying inherited neurometabolic disorders is critical for the institution of treatment, which may positively influence prognosis, and allow for proper genetic counseling. In this review, we discuss those disorders in which the structural brain malformation is a dominant feature, and propose a practical approach that will permit a physician to investigate, and treat these disorders.
Collapse
Affiliation(s)
- Ahmed Y BoAli
- Divisions of Pediatric Neurology, Department of Pediatrics, Prince Sultan Military Medical City,Riyadh, Kingdom of Saudi Arabia
| | | | | |
Collapse
|
27
|
Pietrasanta C, Minoia F, Torreggiani S, Ronchi A, Gattorno M, Volpi S, Ceccherini I, Mosca F, Filocamo G, Pugni L. When neonatal inflammation does not mean infection: an early-onset mevalonate kinase deficiency with interstitial lung disease. Clin Immunol 2019; 205:25-28. [DOI: 10.1016/j.clim.2019.05.002] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2019] [Revised: 04/30/2019] [Accepted: 05/05/2019] [Indexed: 12/30/2022]
|
28
|
A gas chromatography–mass spectrometry-based whole-cell screening assay for target identification in distal cholesterol biosynthesis. Nat Protoc 2019; 14:2546-2570. [DOI: 10.1038/s41596-019-0193-z] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2018] [Accepted: 05/16/2019] [Indexed: 12/14/2022]
|
29
|
Fernández-Cabada T, Ramos-Gómez M. A Novel Contrast Agent Based on Magnetic Nanoparticles for Cholesterol Detection as Alzheimer's Disease Biomarker. NANOSCALE RESEARCH LETTERS 2019; 14:36. [PMID: 30684043 PMCID: PMC6349267 DOI: 10.1186/s11671-019-2863-8] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/12/2018] [Accepted: 01/10/2019] [Indexed: 05/03/2023]
Abstract
BACKGROUND Considering the high incidence of Alzheimer's disease among the world population over the years, and the costs that the disease poses in sanitary and social terms to countries, it is necessary to develop non-invasive diagnostic tests that allow to detect early biomarkers of the disease. Within the early diagnosis methods, the development of contrast agents for magnetic resonance imaging becomes especially useful. Accumulating evidence suggests that cholesterol may play a role in the pathogenesis of Alzheimer's disease since abnormal deposits of cholesterol surrounding senile plaques have been described in animal transgenic models and patients with Alzheimer's disease. In vivo experiments have also shown that diet-induced hypercholesterolemia enhances intraneuronal accumulation of β-amyloid protein accompanied by microgliosis and accelerates β-amyloid deposition in brains. PRESENTATION OF THE HYPOTHESIS In the present study, we propose for the first time the synthesis of a new nanoconjugate composed of magnetic nanoparticles bound to an anti-cholesterol antibody, to detect the abnormal deposits of cholesterol observed in senile plaques in Alzheimer's disease by magnetic resonance imaging. The nanoplatform could also reveal the decrease of cholesterol observed in neuronal plasmatic membranes associated with this pathology. TESTING THE HYPOTHESIS Experimental design to test the hypothesis will be done first in vitro and then in ex vivo and in vivo studies in a second stage. IMPLICATIONS OF THE HYPOTHESIS The designed nanoplatform could therefore detect cholesterol deposits at the cerebral level. The detection of this biomarker in areas coinciding with senile plaque accumulations could provide early information on the onset and progression of Alzheimer's disease.
Collapse
Affiliation(s)
- Tamara Fernández-Cabada
- Centre for Biomedical Technology (CTB), Universidad Politécnica de Madrid, Campus de Montegancedo, Pozuelo de Alarcón, 28223 Madrid, Spain
| | - Milagros Ramos-Gómez
- Centre for Biomedical Technology (CTB), Universidad Politécnica de Madrid, Campus de Montegancedo, Pozuelo de Alarcón, 28223 Madrid, Spain
- CIBER de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Madrid, Spain
| |
Collapse
|
30
|
A Critical Analysis of Molecular Mechanisms Underlying Membrane Cholesterol Sensitivity of GPCRs. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1115:21-52. [PMID: 30649754 DOI: 10.1007/978-3-030-04278-3_2] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
G protein-coupled receptors (GPCRs) are the largest and a diverse family of proteins involved in signal transduction across biological membranes. GPCRs mediate a wide range of physiological processes and have emerged as major targets for the development of novel drug candidates in all clinical areas. Since GPCRs are integral membrane proteins, regulation of their organization, dynamics, and function by membrane lipids, in particular membrane cholesterol, has emerged as an exciting area of research. Cholesterol sensitivity of GPCRs could be due to direct interaction of cholesterol with the receptor (specific effect). Alternately, GPCR function could be influenced by the effect of cholesterol on membrane physical properties (general effect). In this review, we critically analyze the specific and general mechanisms of the modulation of GPCR function by membrane cholesterol, taking examples from representative GPCRs. While evidence for both the proposed mechanisms exists, there appears to be no clear-cut distinction between these two mechanisms, and a combination of these mechanisms cannot be ruled out in many cases. We conclude that classifying the mechanism underlying cholesterol sensitivity of GPCR function merely into these two mutually exclusive classes could be somewhat arbitrary. A more holistic approach could be suitable for analyzing GPCR-cholesterol interaction.
Collapse
|
31
|
Agana M, Frueh J, Kamboj M, Patel DR, Kanungo S. Common metabolic disorder (inborn errors of metabolism) concerns in primary care practice. ANNALS OF TRANSLATIONAL MEDICINE 2018; 6:469. [PMID: 30740400 DOI: 10.21037/atm.2018.12.34] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Inborn errors of metabolism (IEMs) are rare genetic or inherited disorders resulting from an enzyme defect in biochemical and metabolic pathways affecting proteins, fats, carbohydrates metabolism or impaired organelle function presenting as complicated medical conditions involving several human organ systems. They involve great complexity of the underlying pathophysiology, biochemical workup, and molecular analysis, and have complicated therapeutic options for management. Age of presentation can vary from infancy to adolescence with the more severe forms appearing in early childhood accompanied by significant morbidity and mortality. The understanding of these complex disorders requires special in-depth training, American Board of Medical Genetics and Genomics (ABMGG) certification and experience. Most primary care physicians (PCPs) are reluctant to deal with IEM due to unfamiliarity and rarity of such conditions compounded by prompt progression to crisis situations along with paucity of time involved in dealing with such complex disorders. While there are biochemical geneticists aka metabolic specialists' expertise available, mostly in larger academic medical centers, with expertise to deal with these rare complex issues, their initial clinical presentation in most newborns, children, adolescents or adults including asymptomatic positive newborn screen (NBS), occur in the out-patient PCP settings. Therefore, it is important that PCPs' comfort to recognize early signs and symptoms is important to initiate appropriate diagnostic and therapeutic interventions, and be able to make appropriate referrals. The following article reviews common IEM clinical presentations for a robust diagnostic differential and discuss evaluation and management approaches of patients with known or suspected IEM.
Collapse
Affiliation(s)
- Marisha Agana
- Department of Pediatric and Adolescent Medicine, Western Michigan University Homer Stryker MD School of Medicine, Kalamazoo, MI, USA
| | - Julia Frueh
- Department of Pediatric and Adolescent Medicine, Western Michigan University Homer Stryker MD School of Medicine, Kalamazoo, MI, USA
| | - Manmohan Kamboj
- Department of Pediatrics, Nationwide Children's Hospital, Columbus, Ohio, USA
| | - Dilip R Patel
- Department of Pediatric and Adolescent Medicine, Western Michigan University Homer Stryker MD School of Medicine, Kalamazoo, MI, USA
| | - Shibani Kanungo
- Department of Pediatric and Adolescent Medicine, Western Michigan University Homer Stryker MD School of Medicine, Kalamazoo, MI, USA
| |
Collapse
|
32
|
Konecna A, Toth Hervay N, Bencova A, Morvova M, Sikurova L, Jancikova I, Gaskova D, Gbelska Y. Erg6 gene is essential for stress adaptation in Kluyveromyces lactis. FEMS Microbiol Lett 2018; 365:5162844. [PMID: 30398655 DOI: 10.1093/femsle/fny265] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2018] [Accepted: 11/05/2018] [Indexed: 01/20/2023] Open
Abstract
We investigated the effect of Kluyveromyces lactis ERG6 gene deletion on plasma membrane function and showed increased susceptibility of mutant cells to salt stress, cationic drugs and weak organic acids. Contrary to Saccharomyces cerevisiae, Klerg6 mutant cells exhibited increased tolerance to tunicamycin. The content of cell wall polysacharides did not significantly vary between wild-type and mutant cells. Although the expression of the NAD+-dependent glycerol 3-phosphate dehydrogenase (KlGPD1) in the Klerg6 mutant cells was only half of that in the parental strain, it was induced in the presence of calcofluor white. Also, cells exposed to this drug accumulated glycerol. The absence of KlErg6p led to plasma membrane hyperpolarization but had no statistically significant influence on the plasma membrane fluidity. We propose that the phenotype of Klerg6 mutant cells to a large extent was a result of the reduced activity of specific plasma membrane proteins that require proper lipid composition for full activity.
Collapse
Affiliation(s)
- Alexandra Konecna
- Faculty of Sciences, Department of Microbiology and Virology, Comenius University in Bratislava, Ilkovicova 6, 842 15 Bratislava, Slovakia
| | - Nora Toth Hervay
- Faculty of Sciences, Department of Microbiology and Virology, Comenius University in Bratislava, Ilkovicova 6, 842 15 Bratislava, Slovakia
| | - Alexandra Bencova
- Faculty of Sciences, Department of Microbiology and Virology, Comenius University in Bratislava, Ilkovicova 6, 842 15 Bratislava, Slovakia
| | - Marcela Morvova
- Faculty of Mathematics, Physics and Informatics F1, Comenius University in Bratislava, Mlynska dolina 6280, 842 48 Bratislava, Slovakia
| | - Libusa Sikurova
- Faculty of Mathematics, Physics and Informatics F1, Comenius University in Bratislava, Mlynska dolina 6280, 842 48 Bratislava, Slovakia
| | - Iva Jancikova
- Faculty of Mathematics and Physics, Charles University, Ke Karlovu 2027/3, 121 16 Prague, Czech Republic
| | - Dana Gaskova
- Faculty of Mathematics and Physics, Charles University, Ke Karlovu 2027/3, 121 16 Prague, Czech Republic
| | - Yvetta Gbelska
- Faculty of Sciences, Department of Microbiology and Virology, Comenius University in Bratislava, Ilkovicova 6, 842 15 Bratislava, Slovakia
| |
Collapse
|
33
|
Chatuphonprasert W, Jarukamjorn K, Ellinger I. Physiology and Pathophysiology of Steroid Biosynthesis, Transport and Metabolism in the Human Placenta. Front Pharmacol 2018; 9:1027. [PMID: 30258364 PMCID: PMC6144938 DOI: 10.3389/fphar.2018.01027] [Citation(s) in RCA: 153] [Impact Index Per Article: 25.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2018] [Accepted: 08/24/2018] [Indexed: 12/11/2022] Open
Abstract
The steroid hormones progestagens, estrogens, androgens, and glucocorticoids as well as their precursor cholesterol are required for successful establishment and maintenance of pregnancy and proper development of the fetus. The human placenta forms at the interface of maternal and fetal circulation. It participates in biosynthesis and metabolism of steroids as well as their regulated exchange between maternal and fetal compartment. This review outlines the mechanisms of human placental handling of steroid compounds. Cholesterol is transported from mother to offspring involving lipoprotein receptors such as low-density lipoprotein receptor (LDLR) and scavenger receptor class B type I (SRB1) as well as ATP-binding cassette (ABC)-transporters, ABCA1 and ABCG1. Additionally, cholesterol is also a precursor for placental progesterone and estrogen synthesis. Hormone synthesis is predominantly performed by members of the cytochrome P-450 (CYP) enzyme family including CYP11A1 or CYP19A1 and hydroxysteroid dehydrogenases (HSDs) such as 3β-HSD and 17β-HSD. Placental estrogen synthesis requires delivery of sulfate-conjugated precursor molecules from fetal and maternal serum. Placental uptake of these precursors is mediated by members of the solute carrier (SLC) family including sodium-dependent organic anion transporter (SOAT), organic anion transporter 4 (OAT4), and organic anion transporting polypeptide 2B1 (OATP2B1). Maternal-fetal glucocorticoid transport has to be tightly regulated in order to ensure healthy fetal growth and development. For that purpose, the placenta expresses the enzymes 11β-HSD 1 and 2 as well as the transporter ABCB1. This article also summarizes the impact of diverse compounds and diseases on the expression level and activity of the involved transporters, receptors, and metabolizing enzymes and concludes that the regulatory mechanisms changing the physiological to a pathophysiological state are barely explored. The structure and the cellular composition of the human placental barrier are introduced. While steroid production, metabolism and transport in the placental syncytiotrophoblast have been explored for decades, few information is available for the role of placental-fetal endothelial cells in these processes. With regard to placental structure and function, significant differences exist between species. To further decipher physiologic pathways and their pathologic alterations in placental steroid handling, proper model systems are mandatory.
Collapse
Affiliation(s)
- Waranya Chatuphonprasert
- Pathophysiology of the Placenta, Department of Pathophysiology and Allergy Research, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria.,Faculty of Medicine, Mahasarakham University, Maha Sarakham, Thailand
| | - Kanokwan Jarukamjorn
- Research Group for Pharmaceutical Activities of Natural Products Using Pharmaceutical Biotechnology (PANPB), Faculty of Pharmaceutical Sciences, Khon Kaen University, Khon Kaen, Thailand
| | - Isabella Ellinger
- Pathophysiology of the Placenta, Department of Pathophysiology and Allergy Research, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
34
|
Genaro-Mattos TC, Tallman KA, Allen LB, Anderson A, Mirnics K, Korade Z, Porter NA. Dichlorophenyl piperazines, including a recently-approved atypical antipsychotic, are potent inhibitors of DHCR7, the last enzyme in cholesterol biosynthesis. Toxicol Appl Pharmacol 2018; 349:21-28. [PMID: 29698737 DOI: 10.1016/j.taap.2018.04.029] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2018] [Revised: 04/20/2018] [Accepted: 04/21/2018] [Indexed: 12/21/2022]
Abstract
While antipsychotic medications provide important relief from debilitating psychotic symptoms, they also have significant adverse side effects, which might have relevant impact on human health. Several research studies, including ours, have shown that commonly used antipsychotics such as haloperidol and aripiprazole affect cholesterol biosynthesis at the conversion of 7-dehydrocholesterol (7-DHC) to cholesterol. This transformation is promoted by the enzyme DHCR7 and its inhibition causes increases in plasma and tissue levels of 7-DHC. The inhibition of this enzymatic step by mutations in the Dhcr7 gene leads to Smith-Lemli-Opitz syndrome, a devastating human condition that can be replicated in rats by small molecule inhibitors of DHCR7. The fact that two compounds, brexpiprazole and cariprazine, that were recently approved by the FDA have substructural elements in common with the DHCR7 inhibitor aripiprazole, prompted us to evaluate the effect of brexpiprazole and cariprazine on cholesterol biosynthesis. We report that cariprazine affects levels of 7-DHC and cholesterol in cell culture incubations at concentrations as low as 5 nM. Furthermore, a common metabolite of cariprazine and aripiprazole, 2,3-(dichlorophenyl) piperazine, inhibits DHCR7 activity at concentrations comparable to those of the potent teratogen AY9944. The cell culture experiments were corroborated in mice in studies showing that treatment with cariprazine elevated 7-DHC in brain and serum. The consequences of sterol inhibition by antipsychotics in the developing nervous system and the safety of their use during pregnancy remains to be established.
Collapse
Affiliation(s)
- Thiago C Genaro-Mattos
- Department of Chemistry, Vanderbilt Institute of Chemical Biology, Nashville, TN, United States
| | - Keri A Tallman
- Department of Chemistry, Vanderbilt Institute of Chemical Biology, Nashville, TN, United States
| | - Luke B Allen
- Department of Pediatrics and Biochemistry, Molecular Biology, UNMC, Omaha, NE 68198, United States
| | - Allison Anderson
- Munroe-Meyer Institute for Genetics and Rehabilitation, Omaha, NE 68198, United States
| | - Karoly Mirnics
- Munroe-Meyer Institute for Genetics and Rehabilitation, Omaha, NE 68198, United States
| | - Zeljka Korade
- Department of Pediatrics and Biochemistry, Molecular Biology, UNMC, Omaha, NE 68198, United States
| | - Ned A Porter
- Department of Chemistry, Vanderbilt Institute of Chemical Biology, Nashville, TN, United States; Vanderbilt Kennedy Center for Research on Human Development, Nashville, TN, United States.
| |
Collapse
|
35
|
Ce O, Rs P, Ab W, S D, Cj W, Qm M, D L. Potential Link Between Proprotein Convertase Subtilisin/Kexin Type 9 and Alzheimer's Disease. ACTA ACUST UNITED AC 2018; 1. [PMID: 32352077 DOI: 10.31531/2581-4745.1000106] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Alzheimer's disease [AD] is not only the most common neurodegenerative disease but is also currently incurable. Proprotein convertase subtilisin/kexin-9 [PCSK9] is an indirect regulator of plasma low density lipoprotein [LDL] levels controlling LDL receptor expression at the plasma membrane. PCSK9 also appears to regulate the development of glucose intolerance, insulin resistance, abdominal obesity, inflammation, and hypertension, conditions that have been identified as risk factors for AD. PCSK9 levels also depend on age, sex, and ethnic background, factors associated with AD. Herein, we will review indirect evidence that suggests a link between PCSK9 levels and AD.
Collapse
Affiliation(s)
- Oldham Ce
- Department of Pharmaceutical Sciences, Biomanufacturing Research Institute and Technology Enterprise [BRITE], College of Arts and Sciences, North Carolina Central University, Durham, USA
| | - Powell Rs
- Department of Pharmaceutical Sciences, Biomanufacturing Research Institute and Technology Enterprise [BRITE], College of Arts and Sciences, North Carolina Central University, Durham, USA
| | - Williams Ab
- Department of Pharmaceutical Sciences, Biomanufacturing Research Institute and Technology Enterprise [BRITE], College of Arts and Sciences, North Carolina Central University, Durham, USA
| | - Dixon S
- Department of Pharmaceutical Sciences, Biomanufacturing Research Institute and Technology Enterprise [BRITE], College of Arts and Sciences, North Carolina Central University, Durham, USA
| | - Wooten Cj
- Department of Pharmaceutical Sciences, Biomanufacturing Research Institute and Technology Enterprise [BRITE], College of Arts and Sciences, North Carolina Central University, Durham, USA
| | - Melendez Qm
- Department of Pharmaceutical Sciences, Biomanufacturing Research Institute and Technology Enterprise [BRITE], College of Arts and Sciences, North Carolina Central University, Durham, USA
| | - Lopez D
- Department of Pharmaceutical Sciences, Biomanufacturing Research Institute and Technology Enterprise [BRITE], College of Arts and Sciences, North Carolina Central University, Durham, USA
| |
Collapse
|
36
|
Korade Z, Genaro-Mattos TC, Tallman KA, Liu W, Garbett KA, Koczok K, Balogh I, Mirnics K, Porter NA. Vulnerability of DHCR7+/- mutation carriers to aripiprazole and trazodone exposure. J Lipid Res 2017; 58:2139-2146. [PMID: 28972118 DOI: 10.1194/jlr.m079475] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2017] [Revised: 09/19/2017] [Indexed: 12/20/2022] Open
Abstract
Smith-Lemli-Opitz syndrome is a recessive disorder caused by mutations in 7-dehydrocholesterol reductase (DHCR)7 with a heterozygous (HET) carrier frequency of 1-3%. A defective DHCR7 causes accumulation of 7-dehydrocholesterol (DHC), which is a highly oxidizable and toxic compound. Recent studies suggest that several antipsychotics, including the highly prescribed pharmaceuticals, aripiprazole (ARI) and trazodone (TRZ), increase 7-DHC levels in vitro and in humans. Our investigation was designed to compare the effects of ARI and TRZ on cholesterol (Chol) synthesis in fibroblasts from DHCR7+/- human carriers and controls (CTRs). Six matched pairs of fibroblasts were treated and their sterol profile analyzed by LC-MS. Significantly, upon treatment with ARI and TRZ, the total accumulation of 7-DHC was higher in DHCR7-HET cells than in CTR fibroblasts. The same set of experiments was repeated in the presence of 13C-lanosterol to determine residual Chol synthesis, revealing that ARI and TRZ strongly inhibit de novo Chol biosynthesis. The results suggest that DHCR7 carriers have increased vulnerability to both ARI and TRZ exposure compared with CTRs. Thus, the 1-3% of the population who are DHCR7 carriers may be more likely to sustain deleterious health consequences on exposure to compounds like ARI and TRZ that increase levels of 7-DHC, especially during brain development.
Collapse
Affiliation(s)
- Zeljka Korade
- Departments of Pediatrics and Biochemistry and Molecular Biology University of Nebraska Medical Center, Omaha, NE 68198
| | - Thiago C Genaro-Mattos
- Department of Chemistry and Vanderbilt Institute of Chemical Biology Vanderbilt University, Nashville, TN 37235
| | - Keri A Tallman
- Department of Chemistry and Vanderbilt Institute of Chemical Biology Vanderbilt University, Nashville, TN 37235
| | - Wei Liu
- Department of Chemistry and Vanderbilt Institute of Chemical Biology Vanderbilt University, Nashville, TN 37235
| | | | - Katalin Koczok
- Department of Laboratory Medicine, Division of Clinical Genetics, University of Debrecen, Debrecen 4032, Hungary
| | - Istvan Balogh
- Department of Laboratory Medicine, Division of Clinical Genetics, University of Debrecen, Debrecen 4032, Hungary
| | - Karoly Mirnics
- Munroe-Meyer Institute for Genetics and Rehabilitation, University of Nebraska Medical Center, Omaha, NE 68198
| | - Ned A Porter
- Department of Chemistry and Vanderbilt Institute of Chemical Biology Vanderbilt University, Nashville, TN 37235
| |
Collapse
|
37
|
Jeong HS, Funari T, Gordon K, Richard G, Agim NG. Concurrent Chondrodysplasia Punctata Type 2 (Conradi-Hunermann-Happle Syndrome) and Ichthyosis Vulgaris in Teenaged Twin Girls. Pediatr Dermatol 2017; 34:e245-e248. [PMID: 28730607 DOI: 10.1111/pde.13227] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
We present concurrent X-linked chondrodysplasia punctata and ichthyosis vulgaris in adolescent fraternal twin girls, notable for initial presentation with dry skin in adolescence, characterized by dark-brown scale typical of ichthyosis vulgaris and blaschkolinear, atrophic, scaly plaques. This constellation of findings prompted further genetic investigation. Using a multigene approach to examine 39 genes associated with congenital ichthyosis, next-generation sequencing revealed a novel heterozygous missense mutation at a mutational hotspot in the EBP gene c.439C>T (p.R147C) in conjunction with a single nonsense mutation in the FLG gene (p.R501X) in both sisters. These individuals highlight the clinical variability of Conradi-Hunermann-Happle syndrome, illustrate the possibility of co-occurrence of rare and common forms of ichthyosis, and demonstrate the utility of multigene analysis.
Collapse
Affiliation(s)
- Haneol S Jeong
- Department of Dermatology, University of Texas Southwestern Medical Center, Dallas, Texas
| | | | - Katherine Gordon
- Department of Dermatology, University of Texas Southwestern Medical Center, Dallas, Texas.,Pediatric Dermatology, Children's Medical Center, Dallas, Texas
| | | | - Nnenna G Agim
- Department of Dermatology, University of Texas Southwestern Medical Center, Dallas, Texas.,Pediatric Dermatology, Children's Medical Center, Dallas, Texas
| |
Collapse
|
38
|
Corso G, Dello Russo A, Gelzo M. Liver and the defects of cholesterol and bile acids biosynthesis: Rare disorders many diagnostic pitfalls. World J Gastroenterol 2017; 23:5257-5265. [PMID: 28839426 PMCID: PMC5550775 DOI: 10.3748/wjg.v23.i29.5257] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/18/2017] [Revised: 05/01/2017] [Accepted: 07/04/2017] [Indexed: 02/06/2023] Open
Abstract
In recent decades, biotechnology produced a growth of knowledge on the causes and mechanisms of metabolic diseases that have formed the basis for their study, diagnosis and treatment. Unfortunately, it is well known that the clinical features of metabolic diseases can manifest themselves with very different characteristics and escape early detection. Also, it is well known that the prognosis of many metabolic diseases is excellent if diagnosed and treated early. In this editorial we briefly summarized two groups of inherited metabolic diseases, the defects of cholesterol biosynthesis and those of bile acids. Both groups show variable clinical manifestations but some clinical signs and symptoms are common in both the defects of cholesterol and bile acids. The differential diagnosis can be made analyzing sterol profiles in blood and/or bile acids in blood and urine by chromatographic techniques (GC-MS and LC-MS/MS). Several defects of both biosynthetic pathways are treatable so early diagnosis is crucial. Unfortunately their diagnosis is made too late, due either to the clinical heterogeneity of the syndromes (severe, mild and very mild) that to the scarcity of scientific dissemination of these rare diseases. Therefore, the delay in diagnosis leads the patient to the medical observation when the disease has produced irreversible damages to the body. Here, we highlighted simple clinical and laboratory descriptions that can potentially make you to suspect a defect in cholesterol biosynthesis and/or bile acids, as well, we suggest appropriate request of the laboratory tests that along with common clinical features can help to diagnose these defects.
Collapse
|
39
|
Frey AJ, Park BY, Schriver ER, Feldman DR, Parry S, Croen LA, Fallin DM, Hertz-Picciotto I, Newschaffer CJ, Snyder NW. Differences in testosterone and its precursors by sex of the offspring in meconium. J Steroid Biochem Mol Biol 2017; 167:78-85. [PMID: 27871978 PMCID: PMC5292277 DOI: 10.1016/j.jsbmb.2016.11.010] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/27/2016] [Revised: 11/07/2016] [Accepted: 11/14/2016] [Indexed: 01/13/2023]
Abstract
Prenatal metabolism exerts profound effects on development. The first stool of the newborn, meconium, provides a window into the prenatal metabolic environment. The objective of this study was to examine the feasibility of meconium as a novel matrix to quantify prenatal steroid levels. We quantified parameters of analytical interest regarding the use of meconium, including sample stability. We hypothesized that meconium steroid content would differ by sex, prompting analysis of meconium to test effects of prenatal steroid metabolism. Meconium from 193 newborns enrolled in the Early Autism Risk Longitudinal Investigation (EARLI) study, including 107 males, and 86 females, were analyzed by isotope dilution-liquid chromatography-high resolution mass spectrometry (ID-LC-HRMS) while blinded to identity for testosterone (T), androstenedione (AD), and dehydroepiandrosterone (DHEA). Steroid levels were compared by sex, and investigations of potential trends resulting from sample storage or processing was conducted. The unconjugated steroid content of meconium in ng/g (mean, standard deviation) was for males: T (2.67, 8.99), AD (20.01, 28.12), DHEA (13.96, 23.57) and for females: T (0.82, 1.63), AD (22.32, 24.38), DHEA (21.06, 43.49). T was higher in meconium from males (p=0.0333), and DHEA was higher in meconium from females (p=0.0202). 6 female and 3 male T values were below the limit of detection. No extreme variability in hydration or trend in steroid levels by storage time was detected. Sexually dimorphic levels of hormones may reflect gestational differentiation, and future studies should consider meconium analysis.
Collapse
Affiliation(s)
- Alexander J Frey
- AJ Drexel Autism Institute, Drexel University, 3020 Market St, Suite 560, Philadelphia, PA 19104, USA
| | - Bo Y Park
- Department of Mental Health, Johns Hopkins Bloomberg School of Public Health, 624 N. Broadway, HH884 Baltimore, MD 21205, USA
| | - Emily R Schriver
- AJ Drexel Autism Institute, Drexel University, 3020 Market St, Suite 560, Philadelphia, PA 19104, USA
| | - Daniel R Feldman
- AJ Drexel Autism Institute, Drexel University, 3020 Market St, Suite 560, Philadelphia, PA 19104, USA
| | - Samuel Parry
- Maternal Fetal Medicine Division, University of Pennsylvania School of Medicine, 2000 Courtyard Building, 3400 Spruce Street, Philadelphia, PA 19104, USA
| | - Lisa A Croen
- Autism Research Program, Kaiser Permanente Division of Research, 2000 Broadway, Oakland, CA 94612, USA
| | - Daniele M Fallin
- Wendy Klag Center for Autism and Developmental Disabilities, Department of Mental Health, Johns Hopkins Bloomberg School of Public Health, 624 N. Broadway, HH 850, Baltimore, MD 21205, USA
| | - Irva Hertz-Picciotto
- Medical Investigation of Neurodevelopmental Disorders (MIND) Institute, Department of Public Health Sciences, School of Medicine, University of California, Davis, USA
| | - Craig J Newschaffer
- AJ Drexel Autism Institute, Drexel University, 3020 Market St, Suite 560, Philadelphia, PA 19104, USA
| | - Nathaniel W Snyder
- AJ Drexel Autism Institute, Drexel University, 3020 Market St, Suite 560, Philadelphia, PA 19104, USA.
| |
Collapse
|
40
|
Tallman KA, Kim HYH, Korade Z, Genaro-Mattos TC, Wages PA, Liu W, Porter NA. Probes for protein adduction in cholesterol biosynthesis disorders: Alkynyl lanosterol as a viable sterol precursor. Redox Biol 2017; 12:182-190. [PMID: 28258022 PMCID: PMC5333532 DOI: 10.1016/j.redox.2017.02.013] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2017] [Indexed: 01/13/2023] Open
Abstract
The formation of lipid electrophile-protein adducts is associated with many disorders that involve perturbations of cellular redox status. The identities of adducted proteins and the effects of adduction on protein function are mostly unknown and an increased understanding of these factors may help to define the pathogenesis of various human disorders involving oxidative stress. 7-Dehydrocholesterol (7-DHC), the immediate biosynthetic precursor to cholesterol, is highly oxidizable and gives electrophilic oxysterols that adduct proteins readily, a sequence of events proposed to occur in Smith-Lemli-Opitz syndrome (SLOS), a human disorder resulting from an error in cholesterol biosynthesis. Alkynyl lanosterol (a-Lan) was synthesized and studied in Neuro2a cells, Dhcr7-deficient Neuro2a cells and human fibroblasts. When incubated in control Neuro2a cells and control human fibroblasts, a-Lan completed the sequence of steps involved in cholesterol biosynthesis and alkynyl-cholesterol (a-Chol) was the major product formed. In Dhcr7-deficient Neuro2a cells or fibroblasts from SLOS patients, the biosynthetic transformation was interrupted at the penultimate step and alkynyl-7-DHC (a-7-DHC) was the major product formed. When a-Lan was incubated in Dhcr7-deficient Neuro2a cells and the alkynyl tag was used to ligate a biotin group to alkyne-containing products, protein-sterol adducts were isolated and identified. In parallel experiments with a-Lan and a-7-DHC in Dhcr7-deficient Neuro2a cells, a-7-DHC was found to adduct to a larger set of proteins (799) than a-Lan (457) with most of the a-Lan protein adducts (423) being common to the larger a-7-DHC set. Of the 423 proteins found common to both experiments, those formed from a-7-DHC were more highly enriched compared to a DMSO control than were those derived from a-Lan. The 423 common proteins were ranked according to the enrichment determined for each protein in the a-Lan and a-7-DHC experiments and there was a very strong correlation of protein ranks for the adducts formed in the parallel experiments.
Collapse
Affiliation(s)
- Keri A Tallman
- Department of Chemistry and Vanderbilt Institute of Chemical Biology, Vanderbilt University, Nashville, TN 37235, United States
| | - Hye-Young H Kim
- Department of Chemistry and Vanderbilt Institute of Chemical Biology, Vanderbilt University, Nashville, TN 37235, United States
| | - Zeljka Korade
- Vanderbilt Kennedy Center for Research on Human Development, Vanderbilt University, Nashville, TN 37235, United States; Department of Psychiatry, Vanderbilt University, Nashville, TN 37235, United States
| | - Thiago C Genaro-Mattos
- Department of Chemistry and Vanderbilt Institute of Chemical Biology, Vanderbilt University, Nashville, TN 37235, United States
| | - Phillip A Wages
- Department of Chemistry and Vanderbilt Institute of Chemical Biology, Vanderbilt University, Nashville, TN 37235, United States
| | - Wei Liu
- Department of Chemistry and Vanderbilt Institute of Chemical Biology, Vanderbilt University, Nashville, TN 37235, United States
| | - Ned A Porter
- Department of Chemistry and Vanderbilt Institute of Chemical Biology, Vanderbilt University, Nashville, TN 37235, United States; Vanderbilt Kennedy Center for Research on Human Development, Vanderbilt University, Nashville, TN 37235, United States.
| |
Collapse
|
41
|
Aslan A, Borcek AO, Pamukcuoglu S, Baykaner MK. Intracranial undifferentiated malign neuroglial tumor in Smith-Lemli-Opitz syndrome: A theory of a possible predisposing factor for primary brain tumors via a case report. Childs Nerv Syst 2017; 33:171-177. [PMID: 27526097 DOI: 10.1007/s00381-016-3214-z] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/22/2016] [Accepted: 08/03/2016] [Indexed: 12/17/2022]
Abstract
BACKGROUND Smith-Lemli-Opitz Syndrome (SLOS) is a rare hereditary autosomal recessive disorder with broken cholesterol synthesis causing by 7-dehydrocholesterol reductase deficiency. Although the clinical features and pathogenesis is well-defined, it is unknown whether there is a relationship between SLOS and neoplastic processes, especially brain neoplasms. PURPOSE We aimed to attract the attentions to any possibility of relation between SLOS and intracranial tumor development via a pediatric case with both intracranial high-grade neuroglial tumor and SLOS, and thus to contribute an additional data to the literature on togetherness of these two clinical conditions. METHOD In our clinic, we experienced an interesting case of a 10-year-old child with both SLOS and huge brain tumor as rarely seen. Here, we reviewed the features and pathophysiology of SLOS and brain tumors via this case. RESULTS The patient was operated in our clinic, after, his brain tumor had been diagnosed, and his histopathology was resulted in undifferentiated malignant neuroglial WHO grade 4 tumor. CONCLUSION According to current literature, our case is the first report on coexisting of SLOS and intracranial undifferentiated malignant neuroglial tumor. Common pathways like impaired sonic hedgehog (Shh) signaling pathway may be considered for pathogenesis of a probable link between SLOS and brain tumors in further studies.
Collapse
Affiliation(s)
- Ayfer Aslan
- Neurosurgery Department, Faculty of Medicine, Gazi University, Ankara, Turkey.
| | - Alp Ozgun Borcek
- Division of Pediatric Neurosurgery, Faculty of Medicine, Gazi University, Ankara, Turkey
| | - Selma Pamukcuoglu
- Department of Pathology, Gazi University Faculty of Medicine, Ankara, Turkey
| | - M Kemali Baykaner
- Division of Pediatric Neurosurgery, Faculty of Medicine, Gazi University, Ankara, Turkey
| |
Collapse
|
42
|
Jafurulla M, Chattopadhyay A. Structural Stringency of Cholesterol for Membrane Protein Function Utilizing Stereoisomers as Novel Tools: A Review. Methods Mol Biol 2017; 1583:21-39. [PMID: 28205164 DOI: 10.1007/978-1-4939-6875-6_3] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/06/2023]
Abstract
Cholesterol is an important lipid in the context of membrane protein function. The function of a number of membrane proteins, including G protein-coupled receptors (GPCRs) and ion channels, has been shown to be dependent on membrane cholesterol. However, the molecular mechanism underlying such regulation is still being explored. In some cases, specific interaction between cholesterol and the protein has been implicated. In other cases, the effect of cholesterol on the membrane properties has been attributed for the regulation of protein function. In this article, we have provided an overview of experimental approaches that are useful for determining the degree of structural stringency of cholesterol for membrane protein function. In the process, we have highlighted the role of immediate precursors in cholesterol biosynthetic pathway in the function of membrane proteins. Special emphasis has been given to the application of stereoisomers of cholesterol in deciphering the structural stringency required for regulation of membrane protein function. A comprehensive examination of these processes would help in understanding the molecular basis of cholesterol regulation of membrane proteins in subtle details.
Collapse
Affiliation(s)
- Md Jafurulla
- CSIR-Centre for Cellular and Molecular Biology, Uppal Road, Hyderabad, 500 007, India
| | | |
Collapse
|
43
|
Accumulation of specific sterol precursors targets a MAP kinase cascade mediating cell-cell recognition and fusion. Proc Natl Acad Sci U S A 2016; 113:11877-11882. [PMID: 27708165 DOI: 10.1073/pnas.1610527113] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
Sterols are vital components of eukaryotic cell membranes. Defects in sterol biosynthesis, which result in the accumulation of precursor molecules, are commonly associated with cellular disorders and disease. However, the effects of these sterol precursors on the metabolism, signaling, and behavior of cells are only poorly understood. In this study, we show that the accumulation of only ergosterol precursors with a conjugated double bond in their aliphatic side chain specifically disrupts cell-cell communication and fusion in the fungus Neurospora crassa Genetically identical germinating spores of this fungus undergo cell-cell fusion, thereby forming a highly interconnected supracellular network during colony initiation. Before fusion, the cells use an unusual signaling mechanism that involves the coordinated and alternating switching between signal sending and receiving states of the two fusion partners. Accumulation of only ergosterol precursors with a conjugated double bond in their aliphatic side chain disrupts this coordinated cell-cell communication and suppresses cell fusion. These specific sterol precursors target a single ERK-like mitogen-activated protein (MAP) kinase (MAK-1)-signaling cascade, whereas a second MAP kinase pathway (MAK-2), which is also involved in cell fusion, is unaffected. These observations indicate that a minor specific change in sterol structure can exert a strong detrimental effect on a key signaling pathway of the cell, resulting in the absence of cell fusion.
Collapse
|
44
|
Tulodziecka K, Diaz-Rohrer BB, Farley MM, Chan RB, Di Paolo G, Levental KR, Waxham MN, Levental I. Remodeling of the postsynaptic plasma membrane during neural development. Mol Biol Cell 2016; 27:3480-3489. [PMID: 27535429 PMCID: PMC5221582 DOI: 10.1091/mbc.e16-06-0420] [Citation(s) in RCA: 81] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2016] [Accepted: 08/04/2016] [Indexed: 11/29/2022] Open
Abstract
Neuronal synapses require precise regulation, particularly of membrane components. The composition and organization of synaptic membranes are dramatically remodeled during development, including accumulation of lipids associated with raft domains, and concomitant palmitoylation of PSD-95, suggesting recruitment of domains via scaffold lipidation. Neuronal synapses are the fundamental units of neural signal transduction and must maintain exquisite signal fidelity while also accommodating the plasticity that underlies learning and development. To achieve these goals, the molecular composition and spatial organization of synaptic terminals must be tightly regulated; however, little is known about the regulation of lipid composition and organization in synaptic membranes. Here we quantify the comprehensive lipidome of rat synaptic membranes during postnatal development and observe dramatic developmental lipidomic remodeling during the first 60 postnatal days, including progressive accumulation of cholesterol, plasmalogens, and sphingolipids. Further analysis of membranes associated with isolated postsynaptic densities (PSDs) suggests the PSD-associated postsynaptic plasma membrane (PSD-PM) as one specific location of synaptic remodeling. We analyze the biophysical consequences of developmental remodeling in reconstituted synaptic membranes and observe remarkably stable microdomains, with the stability of domains increasing with developmental age. We rationalize the developmental accumulation of microdomain-forming lipids in synapses by proposing a mechanism by which palmitoylation of the immobilized scaffold protein PSD-95 nucleates domains at the postsynaptic plasma membrane. These results reveal developmental changes in lipid composition and palmitoylation that facilitate the formation of postsynaptic membrane microdomains, which may serve key roles in the function of the neuronal synapse.
Collapse
Affiliation(s)
- Karolina Tulodziecka
- Department of Integrative Biology and Pharmacology, McGovern Medical School at the University of Texas Health Science Center at Houston, Houston, TX 77030
| | - Barbara B Diaz-Rohrer
- Department of Integrative Biology and Pharmacology, McGovern Medical School at the University of Texas Health Science Center at Houston, Houston, TX 77030
| | - Madeline M Farley
- Department of Neurobiology and Anatomy, McGovern Medical School at the University of Texas Health Science Center at Houston, Houston, TX 77030
| | - Robin B Chan
- Department of Pathology and Cell Biology, Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University Medical Center, New York, NY 10032
| | - Gilbert Di Paolo
- Department of Pathology and Cell Biology, Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University Medical Center, New York, NY 10032
| | - Kandice R Levental
- Department of Integrative Biology and Pharmacology, McGovern Medical School at the University of Texas Health Science Center at Houston, Houston, TX 77030
| | - M Neal Waxham
- Department of Neurobiology and Anatomy, McGovern Medical School at the University of Texas Health Science Center at Houston, Houston, TX 77030
| | - Ilya Levental
- Department of Integrative Biology and Pharmacology, McGovern Medical School at the University of Texas Health Science Center at Houston, Houston, TX 77030
| |
Collapse
|
45
|
Kyle SM, Saha PK, Brown HM, Chan LC, Justice MJ. MeCP2 co-ordinates liver lipid metabolism with the NCoR1/HDAC3 corepressor complex. Hum Mol Genet 2016; 25:3029-3041. [PMID: 27288453 PMCID: PMC5181597 DOI: 10.1093/hmg/ddw156] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2016] [Revised: 05/02/2016] [Accepted: 05/18/2016] [Indexed: 01/11/2023] Open
Abstract
Rett syndrome (RTT; OMIM 312750), a progressive neurological disorder, is caused by mutations in methyl-CpG-binding protein 2 (MECP2; OMIM 300005), a ubiquitously expressed factor. A genetic suppressor screen designed to identify therapeutic targets surprisingly revealed that downregulation of the cholesterol biosynthesis pathway improves neurological phenotypes in Mecp2 mutant mice. Here, we show that MeCP2 plays a direct role in regulating lipid metabolism. Mecp2 deletion in mice results in a host of severe metabolic defects caused by lipid accumulation, including insulin resistance, fatty liver, perturbed energy utilization, and adipose inflammation by macrophage infiltration. We show that MeCP2 regulates lipid homeostasis by anchoring the repressor complex containing NCoR1 and HDAC3 to its lipogenesis targets in hepatocytes. Consistently, we find that liver targeted deletion of Mecp2 causes fatty liver disease and dyslipidemia similar to HDAC3 liver-specific deletion. These findings position MeCP2 as a novel component in metabolic homeostasis. Rett syndrome patients also show signs of peripheral dyslipidemia; thus, together these data suggest that RTT should be classified as a neurological disorder with systemic metabolic components. We previously showed that treatment of Mecp2 mice with statin drugs alleviated motor symptoms and improved health and longevity. Lipid metabolism is a highly treatable target; therefore, our results shed light on new metabolic pathways for treatment of Rett syndrome.
Collapse
Affiliation(s)
- Stephanie M Kyle
- Genetics and Genome Biology Program, The Hospital for Sick Children, The Peter Gilgan Centre for Research and Learning, Toronto, ON M5G 0A4, Canada.,Department of Molecular and Human Genetics
| | - Pradip K Saha
- Department of Medicine, Division of Diabetes, Endocrinology and Metabolism, Baylor College of Medicine, Houston, TX 77030, USA
| | | | - Lawrence C Chan
- Department of Medicine, Division of Diabetes, Endocrinology and Metabolism, Baylor College of Medicine, Houston, TX 77030, USA
| | - Monica J Justice
- Genetics and Genome Biology Program, The Hospital for Sick Children, The Peter Gilgan Centre for Research and Learning, Toronto, ON M5G 0A4, Canada .,Department of Molecular and Human Genetics.,Department of Molecular Genetics, University of Toronto, Toronto, ON M5S 1A1, Canada
| |
Collapse
|
46
|
Peciuliene S, Burnyte B, Gudaitiene R, Rusoniene S, Drazdiene N, Liubsys A, Utkus A. Perinatal manifestation of mevalonate kinase deficiency and efficacy of anakinra. Pediatr Rheumatol Online J 2016; 14:19. [PMID: 27012807 PMCID: PMC4807578 DOI: 10.1186/s12969-016-0081-9] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/21/2016] [Accepted: 03/21/2016] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND Mevalonate kinase deficiency is a metabolic autoinflammatory syndrome caused by mutations in the MVK gene, mevalonate kinase, the key enzyme in the non-sterol isoprenoid biosynthesis pathway. Two phenotypes of mevalonate kinase deficiency are known based on the level of enzymatic deficiency, mevalonic aciduria and hyperimmunoglobulinemia D syndrome, but a wide spectrum of intermediate phenotypes has been reported. Currently one of the most effective treatments is biological therapy (with interleukin-1 antagonist anakinra or tumour necrosis factor-α inhibitor etanercept). CASE PRESENTATION The patient in this case has a phenotype contributing to a severe disease that caused the symptoms to manifest very early, in the prenatal period. Mevalonate kinase deficiency was suspected on the basis of clinical (hydrops fetalis, hepatosplenomegaly, hypotonia) and laboratory signs (anaemia, intense acute phase reaction, increased urinary excretion of mevalonic acid). Mutation analysis of the MVK gene confirmed the biochemical diagnosis. Treatment with the interleukin-1 antagonist anakinra was started (minimal dose of 1 mg/kg/day) and revealed its efficacy after three days. CONCLUSIONS Our case highlights the need for a very detailed clinical and laboratory assessment in new-borns with any suggestion of autoinflammatory disorders. It is important that patients are diagnosed as early as possible to provide better multidisciplinary follow-up and therapy when needed.
Collapse
Affiliation(s)
- Skaiste Peciuliene
- Neonatology Centre of Vilnius University, Santariškių St. 7, Vilnius, Lithuania.
| | - Birute Burnyte
- Neonatology Centre of Vilnius University, Santariškių St. 7, Vilnius, Lithuania ,Department of Human and Medical Genetics, Faculty of Medicine, Vilnius University, Vilnius, Lithuania
| | - Rymanta Gudaitiene
- Neonatology Centre of Vilnius University, Santariškių St. 7, Vilnius, Lithuania
| | | | - Nijole Drazdiene
- Neonatology Centre of Vilnius University, Santariškių St. 7, Vilnius, Lithuania
| | - Arunas Liubsys
- Neonatology Centre of Vilnius University, Santariškių St. 7, Vilnius, Lithuania
| | - Algirdas Utkus
- Department of Human and Medical Genetics, Faculty of Medicine, Vilnius University, Vilnius, Lithuania
| |
Collapse
|
47
|
Korade Z, Kim HYH, Tallman KA, Liu W, Koczok K, Balogh I, Xu L, Mirnics K, Porter NA. The Effect of Small Molecules on Sterol Homeostasis: Measuring 7-Dehydrocholesterol in Dhcr7-Deficient Neuro2a Cells and Human Fibroblasts. J Med Chem 2016; 59:1102-15. [PMID: 26789657 DOI: 10.1021/acs.jmedchem.5b01696] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Well-established cell culture models were combined with new analytical methods to assess the effects of small molecules on the cholesterol biosynthesis pathway. The analytical protocol, which is based on sterol derivation with the dienolphile PTAD, was found to be reliable for the analysis of 7-DHC and desmosterol. The PTAD method was applied to the screening of a small library of pharmacologically active substances, and the effect of compounds on the cholesterol pathway was determined. Of some 727 compounds, over 30 compounds decreased 7-DHC in Dhcr7-deficient Neuro2a cells. The examination of chemical structures of active molecules in the screen grouped the compounds into distinct categories. In addition to statins, our screen found that SERMs, antifungals, and several antipsychotic medications reduced levels of 7-DHC. The activities of selected compounds were verified in human fibroblasts derived from Smith-Lemli-Opitz syndrome (SLOS) patients and linked to specific transformations in the cholesterol biosynthesis pathway.
Collapse
Affiliation(s)
- Zeljka Korade
- Vanderbilt Kennedy Center for Research on Human Development, Vanderbilt University , Nashville, Tennessee 37235, United States
| | | | | | | | - Katalin Koczok
- Department of Laboratory Medicine, Division of Clinical Genetics, University of Debrecen , Nagyerdei krt. 98, 4032 Debrecen, Hungary
| | - Istvan Balogh
- Department of Laboratory Medicine, Division of Clinical Genetics, University of Debrecen , Nagyerdei krt. 98, 4032 Debrecen, Hungary
| | | | - Karoly Mirnics
- Vanderbilt Kennedy Center for Research on Human Development, Vanderbilt University , Nashville, Tennessee 37235, United States
| | - Ned A Porter
- Vanderbilt Kennedy Center for Research on Human Development, Vanderbilt University , Nashville, Tennessee 37235, United States
| |
Collapse
|
48
|
Peri A. Neuroprotective effects of estrogens: the role of cholesterol. J Endocrinol Invest 2016; 39:11-8. [PMID: 26084445 DOI: 10.1007/s40618-015-0332-5] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/24/2015] [Accepted: 06/01/2015] [Indexed: 10/23/2022]
Abstract
INTRODUCTION Experimental and clinical evidence suggests that estrogens have protective effects in the brain. Nevertheless, their potential role against neurodegenerative diseases, in particular Alzheimer's disease (AD), is still a matter of debate. The identification of the seladin-1 gene (for SELective Alzheimer's Disease INdicator-1), which appeared to be significantly less expressed in brain region affected in AD, opened a new scenario in the field of neuroprotective mechanisms. Seladin-1 was found to have neuroprotective properties through its anti-apoptotic activity. In addition, it was subsequently demonstrated that seladin-1 also has enzymatic activity, because it catalyzes the conversion of desmosterol into cholesterol. Several studies have shown that an appropriate amount of membrane cholesterol plays a pivotal role to protect nerve cells against β-amyloid toxicity in AD and to counteract the synthesis of β-amyloid. METHODS AND RESULTS We demonstrated that the expression of seladin-1, as well as the synthesis of cell cholesterol, is stimulated by estrogens in human neuronal precursor cells. Cholesterol enriched cells became more resistant against oxidative stress and β-amyloid toxicity. We thus hypothesized that seladin-1 might be a mediator of the neuroprotective effects of estrogens. Indeed, in cells in which seladin-1 gene expression had been silenced by siRNA the protective effects of estrogens were lost. This finding indicates that seladin-1 is a crucial mediator of the neuroprotective effects of these hormones, at least in our cell model. CONCLUSIONS In summary, these results establish a new link between estrogens and cholesterol, which is represented by the neuroprotective factor seladin-1.
Collapse
Affiliation(s)
- A Peri
- Endocrine Unit, Department of Experimental and Biomedical Sciences "Mario Serio", Center for Research, Transfer and High Education on Chronic, Inflammatory, Degenerative and Neoplastic Disorders for the Development of Novel Therapies, University of Florence, Viale Pieraccini, 6, 50139, Florence, Italy.
| |
Collapse
|
49
|
Vergnes L, Chin RG, de Aguiar Vallim T, Fong LG, Osborne TF, Young SG, Reue K. SREBP-2-deficient and hypomorphic mice reveal roles for SREBP-2 in embryonic development and SREBP-1c expression. J Lipid Res 2015; 57:410-21. [PMID: 26685326 DOI: 10.1194/jlr.m064022] [Citation(s) in RCA: 49] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2015] [Indexed: 12/31/2022] Open
Abstract
Cholesterol and fatty acid biosynthesis are regulated by the sterol regulatory element-binding proteins (SREBPs), encoded by Srebf1 and Srebf2. We generated mice that were either deficient or hypomorphic for SREBP-2. SREBP-2 deficiency generally caused death during embryonic development. Analyses of Srebf2(-/-) embryos revealed a requirement for SREBP-2 in limb development and expression of morphogenic genes. We encountered only one viable Srebf2(-/-) mouse, which displayed alopecia, attenuated growth, and reduced adipose tissue stores. Hypomorphic SREBP-2 mice (expressing low levels of SREBP-2) survived development, but the female mice exhibited reduced body weight and died between 8 and 12 weeks of age. Male hypomorphic mice were viable but had reduced cholesterol stores in the liver and lower expression of SREBP target genes. Reduced SREBP-2 expression affected SREBP-1 isoforms in a tissue-specific manner. In the liver, reduced SREBP-2 expression nearly abolished Srebf1c transcripts and reduced Srebf1a mRNA levels. In contrast, adipose tissue displayed normal expression of SREBP target genes, likely due to a compensatory increase in Srebf1a expression. Our results establish that SREBP-2 is critical for survival and limb patterning during development. Reduced expression of SREBP-2 from the hypomorphic allele leads to early death in females and reduced cholesterol content in the liver, but not in adipose tissue.
Collapse
Affiliation(s)
- Laurent Vergnes
- Departments of Human Genetics David Geffen School of Medicine at the University of California, Los Angeles, CA 90095
| | - Robert G Chin
- Departments of Human Genetics David Geffen School of Medicine at the University of California, Los Angeles, CA 90095
| | - Thomas de Aguiar Vallim
- Medicine, David Geffen School of Medicine at the University of California, Los Angeles, CA 90095
| | - Loren G Fong
- Medicine, David Geffen School of Medicine at the University of California, Los Angeles, CA 90095
| | - Timothy F Osborne
- Metabolic Disease Program, Sanford-Burnham Medical Research Institute, Orlando, FL 32827
| | - Stephen G Young
- Departments of Human Genetics David Geffen School of Medicine at the University of California, Los Angeles, CA 90095 Medicine, David Geffen School of Medicine at the University of California, Los Angeles, CA 90095 Molecular Biology Institute, University of California, Los Angeles, CA 90095
| | - Karen Reue
- Departments of Human Genetics David Geffen School of Medicine at the University of California, Los Angeles, CA 90095 Medicine, David Geffen School of Medicine at the University of California, Los Angeles, CA 90095 Molecular Biology Institute, University of California, Los Angeles, CA 90095
| |
Collapse
|
50
|
Porter TD. Electron Transfer Pathways in Cholesterol Synthesis. Lipids 2015; 50:927-36. [PMID: 26344922 DOI: 10.1007/s11745-015-4065-1] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2015] [Accepted: 08/21/2015] [Indexed: 12/17/2022]
Abstract
Cholesterol synthesis in the endoplasmic reticulum requires electron input at multiple steps and utilizes both NADH and NADPH as the electron source. Four enzymes catalyzing five steps in the pathway require electron input: squalene monooxygenase, lanosterol demethylase, sterol 4α-methyl oxidase, and sterol C5-desaturase. The electron-donor proteins for these enzymes include cytochrome P450 reductase and the cytochrome b5 pathway. Here I review the evidence for electron donor protein requirements with these enzymes, the evidence for additional electron donor pathways, and the effect of deletion of these redox enzymes on cholesterol and lipid metabolism.
Collapse
Affiliation(s)
- Todd D Porter
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Kentucky, Lexington, KY, 40536-0596, USA.
| |
Collapse
|