1
|
Mu F, Wang M, Zeng X, Liu L, Wang F. A predictive model of pregnancy loss using pre-pregnancy endocrine and immunological parameters in women with abnormal glucose/lipid metabolism and previous pregnancy loss. Endocrine 2024; 86:441-450. [PMID: 38898223 PMCID: PMC11445311 DOI: 10.1007/s12020-024-03937-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/12/2024] [Accepted: 06/17/2024] [Indexed: 06/21/2024]
Abstract
OBJECTIVE To investigate the clinical and endocrine risk factors for pregnancy loss in women with abnormal glucose/lipid metabolism and a history of pregnancy loss, and to develop a predictive model to assess the risk of pregnancy loss in these women's subsequent pregnancies. METHODS Patients with a history of pregnancy loss who had abnormal glucose/lipid metabolism were retrospectively included in this study, and their pre-pregnancy baseline and clinical characteristics were collected. A predictive nomogram was constructed based on the results of the multivariable logistic regression model analysis, and its calibration and discriminatory capabilities were evaluated. The internal validation was then performed and the net benefits were assessed by the clinical decision curve. RESULTS The predictive model was eventually incorporated eight variables, including maternal age, previous pregnancy losses, anticardiolipin antibody (aCL) IgG, aCL IgM, thyroid peroxidase antibody, complement 4, free thyroxine and total cholesterol. The area under the curve (AUC) of the nomogram was 0.709, and Chi-square value and P value of the Hosmer-Lemeshow test were 12.786 and 0.119, respectively, indicating that the nomogram had a satisfactory calibration and discriminatory performance. The validation cohort showed a similar result for the discrimination of the nomogram (AUC = 0.715). The clinical decision curve demonstrated the nomogram had good positive net benefits. CONCLUSIONS This is the first study to predict the risks of subsequent pregnancy loss in women with abnormal glucose/lipid metabolism and history of pregnancy loss using pre-pregnancy clinical and endocrine parameters. This predictive nomogram may provide clinicians assistance to personalize the management of subsequent pregnancies in these patients.
Collapse
Affiliation(s)
- Fangxiang Mu
- Department of Reproductive Medicine, Lanzhou University Second Hospital, Lanzhou, 730030, China
| | - Mei Wang
- Department of Reproductive Medicine, Lanzhou University Second Hospital, Lanzhou, 730030, China
| | - Xianghui Zeng
- Department of Reproductive Medicine, Lanzhou University Second Hospital, Lanzhou, 730030, China
| | - Lin Liu
- Department of Reproductive Medicine, Lanzhou University Second Hospital, Lanzhou, 730030, China
| | - Fang Wang
- Department of Reproductive Medicine, Lanzhou University Second Hospital, Lanzhou, 730030, China.
| |
Collapse
|
2
|
He K, Zhao Z, Zhang J, Li D, Wang S, Liu Q. Cholesterol Metabolism in Neurodegenerative Diseases. Antioxid Redox Signal 2024. [PMID: 38842175 DOI: 10.1089/ars.2024.0674] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 06/07/2024]
Abstract
Significance: Cholesterol plays a crucial role in the brain, where it is highly concentrated and tightly regulated to support normal brain functions. It serves as a vital component of cell membranes, ensuring their integrity, and acts as a key regulator of various brain processes. Dysregulation of cholesterol metabolism in the brain has been linked to impaired brain function and the onset of neurodegenerative diseases such as Alzheimer's disease (AD), Parkinson's disease, and Huntington's disease. Recent Advances: A significant advancement has been the identification of astrocyte-derived apoliprotein E as a key regulator of de novo cholesterol biosynthesis in neurons, providing insights into how extracellular signals influence neuronal cholesterol levels. In addition, the development of antibody-based therapies, particularly for AD, presents promising opportunities for therapeutic interventions. Critical Issues: Despite significant research, the association between cholesterol and neurodegenerative diseases remains inconclusive. It is crucial to distinguish between plasma cholesterol and brain cholesterol, as these pools are relatively independent. This differentiation should be considered when evaluating statin-based treatment approaches. Furthermore, assessing not only the total cholesterol content in the brain but also its distribution among different types of brain cells is essential. Future Direction: Establishing a causal link between changes in brain/plasma cholesterol levels and the onset of brain dysfunction/neurodegenerative diseases remains a key objective. In addition, conducting cell-specific analyses of cholesterol homeostasis in various types of brain cells under pathological conditions will enhance our understanding of cholesterol metabolism in neurodegenerative diseases. Manipulating cholesterol levels to restore homeostasis may represent a novel approach for alleviating neurological symptoms.
Collapse
Affiliation(s)
- Keqiang He
- Department of Anesthesiology, The First Affiliated Hospital of USTC, Center for Advanced Interdisciplinary Science and Biomedicine of IHM, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230001, China
| | - Zhiwei Zhao
- Department of Cardiovascular Surgery, the First Affiliated Hospital of USTC, Hefei National Laboratory for Physical Sciences at the Microscale, Division of Life Sciences and Medicine, University of Science and Technology of China
| | - Juan Zhang
- Department of Neurology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, Hefei National Research Center for Physical Sciences at the Microscale, University of Science and Technology of China, Hefei, China
- CAS Key Laboratory of Brain Function and Diseases, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
- Institute on Aging and Brain Disorders, The First Affiliated Hospital of USTC, University of Science and Technology of China, Hefei, China
| | - Dingfeng Li
- Department of Neurology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, Hefei National Research Center for Physical Sciences at the Microscale, University of Science and Technology of China, Hefei, China
- CAS Key Laboratory of Brain Function and Diseases, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
- Institute on Aging and Brain Disorders, The First Affiliated Hospital of USTC, University of Science and Technology of China, Hefei, China
| | - Sheng Wang
- Department of Anesthesiology, The First Affiliated Hospital of USTC, Center for Advanced Interdisciplinary Science and Biomedicine of IHM, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230001, China
| | - Qiang Liu
- Department of Anesthesiology, The First Affiliated Hospital of USTC, Center for Advanced Interdisciplinary Science and Biomedicine of IHM, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230001, China
- Department of Neurology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, Hefei National Research Center for Physical Sciences at the Microscale, University of Science and Technology of China, Hefei, China
- CAS Key Laboratory of Brain Function and Diseases, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
- Institute on Aging and Brain Disorders, The First Affiliated Hospital of USTC, University of Science and Technology of China, Hefei, China
- Neurodegenerative Disorder Research Center, Anhui Province Key Laboratory of Biomedical Aging Research, University of Science and Technology of China, Hefei, China
- Key Laboratory of Immune Response and Immunotherapy, University of Science and Technology of China, Hefei, China
| |
Collapse
|
3
|
Hoock JGF, Rossetti C, Bilgin M, Depta L, Enemark-Rasmussen K, Christianson JC, Laraia L. Identification of non-conventional small molecule degraders and stabilizers of squalene synthase. Chem Sci 2023; 14:12973-12983. [PMID: 38023519 PMCID: PMC10664564 DOI: 10.1039/d3sc04064j] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2023] [Accepted: 10/15/2023] [Indexed: 12/01/2023] Open
Abstract
Squalene synthase (SQS) is an essential enzyme in the mevalonate pathway, which controls cholesterol biosynthesis and homeostasis. Although catalytic inhibitors of SQS have been developed, none have been approved for therapeutic use so far. Herein we sought to develop SQS degraders using targeted protein degradation (TPD) to lower overall cellular cholesterol content. We found that KY02111, a small molecule ligand of SQS, selectively causes SQS to degrade in a proteasome-dependent manner. Unexpectedly, compounds based on the same scaffold linked to E3 ligase recruiting ligands led to SQS stabilization. Proteomic analysis found KY02111 to reduce only the levels of SQS, while lipidomic analysis determined that KY02111-induced degradation lowered cellular cholesteryl ester content. Stabilizers shielded SQS from its natural turnover without recruiting their matching E3 ligase or affecting enzymatic target activity. Our work shows that degradation of SQS is possible despite a challenging biological setting and provides the first chemical tools to degrade and stabilize SQS.
Collapse
Affiliation(s)
- Joseph G F Hoock
- Department of Chemistry, Technical University of Denmark Kemitorvet 207 Kongens Lyngby 2800 Denmark
| | - Cecilia Rossetti
- Department of Chemistry, Technical University of Denmark Kemitorvet 207 Kongens Lyngby 2800 Denmark
| | - Mesut Bilgin
- Lipidomics Core Facility, Danish Cancer Institute Strandboulevarden 49 Copenhagen 2100 Denmark
| | - Laura Depta
- Department of Chemistry, Technical University of Denmark Kemitorvet 207 Kongens Lyngby 2800 Denmark
| | - Kasper Enemark-Rasmussen
- Department of Chemistry, Technical University of Denmark Kemitorvet 207 Kongens Lyngby 2800 Denmark
| | - John C Christianson
- Nuffield Department of Rheumatology, Orthopaedics, and Musculoskeletal Sciences, Botnar Research Centre, University of Oxford Headington Oxford OX3 7LD UK
| | - Luca Laraia
- Department of Chemistry, Technical University of Denmark Kemitorvet 207 Kongens Lyngby 2800 Denmark
| |
Collapse
|
4
|
Patel P, Selvaraju V, Babu JR, Wang X, Geetha T. Novel Differentially Methylated Regions Identified by Genome-Wide DNA Methylation Analyses Contribute to Racial Disparities in Childhood Obesity. Genes (Basel) 2023; 14:genes14051098. [PMID: 37239458 DOI: 10.3390/genes14051098] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2023] [Revised: 05/10/2023] [Accepted: 05/15/2023] [Indexed: 05/28/2023] Open
Abstract
The magnitude of the childhood obesity epidemic and its effects on public health has accelerated the pursuit of practical preventative measures. Epigenetics is one subject that holds a lot of promise, despite being relatively new. The study of potentially heritable variations in gene expression that do not require modifications to the underlying DNA sequence is known as epigenetics. Here, we used Illumina MethylationEPIC BeadChip Array to identify differentially methylated regions in DNA isolated from saliva between normal weight (NW) and overweight/obese (OW/OB) children and between European American (EA) and African American (AA) children. A total of 3133 target IDs (associated with 2313 genes) were differentially methylated (p < 0.05) between NW and OW/OB children. In OW/OB children, 792 target IDs were hypermethylated and 2341 were hypomethylated compared to NW. Similarly, in the racial groups EA and AA, a total of 1239 target IDs corresponding to 739 genes were significantly differentially methylated in which 643 target IDs were hypermethylated and 596 were hypomethylated in the AA compared to EA participants. Along with this, the study identified novel genes that could contribute to the epigenetic regulation of childhood obesity.
Collapse
Affiliation(s)
- Priyadarshni Patel
- Department of Nutritional Sciences, Auburn University, Auburn, AL 36849, USA
| | | | - Jeganathan Ramesh Babu
- Department of Nutritional Sciences, Auburn University, Auburn, AL 36849, USA
- Boshell Metabolic Diseases and Diabetes Program, Auburn University, Auburn, AL 36849, USA
- Alabama Agricultural Experiment Station, Auburn University, Auburn, AL 36849, USA
| | - Xu Wang
- Alabama Agricultural Experiment Station, Auburn University, Auburn, AL 36849, USA
- Department of Pathobiology, College of Veterinary Medicine, Auburn University, Auburn, AL 36849, USA
- HudsonAlpha Institute for Biotechnology, Huntsville, AL 35806, USA
| | - Thangiah Geetha
- Department of Nutritional Sciences, Auburn University, Auburn, AL 36849, USA
- Boshell Metabolic Diseases and Diabetes Program, Auburn University, Auburn, AL 36849, USA
- Alabama Agricultural Experiment Station, Auburn University, Auburn, AL 36849, USA
| |
Collapse
|
5
|
Oike A, Iwata S, Hirayama A, Ono Y, Nagasato Y, Kawabata Y, Takai S, Sanematsu K, Wada N, Shigemura N. Bisphosphonate affects the behavioral responses to HCl by disrupting farnesyl diphosphate synthase in mouse taste bud and tongue epithelial cells. Sci Rep 2022; 12:21246. [PMID: 36481783 PMCID: PMC9732047 DOI: 10.1038/s41598-022-25755-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Accepted: 12/05/2022] [Indexed: 12/13/2022] Open
Abstract
Little is known about the molecular mechanisms underlying drug-induced taste disorders, which can cause malnutrition and reduce quality of life. One of taste disorders is known adverse effects of bisphosphonates, which are administered as anti-osteoporotic drugs. Therefore, the present study evaluated the effects of risedronate (a bisphosphonate) on taste bud cells. Expression analyses revealed that farnesyl diphosphate synthase (FDPS, a key enzyme in the mevalonate pathway) was present in a subset of mouse taste bud and tongue epithelial cells, especially type III sour-sensitive taste cells. Other mevalonate pathway-associated molecules were also detected in mouse taste buds. Behavioral analyses revealed that mice administered risedronate exhibited a significantly enhanced aversion to HCl but not for other basic taste solutions, whereas the taste nerve responses were not affected by risedronate. Additionally, the taste buds of mice administered risedronate exhibited significantly lower mRNA expression of desmoglein-2, an integral component of desmosomes. Taken together, these findings suggest that risedronate may interact directly with FDPS to inhibit the mevalonate pathway in taste bud and tongue epithelial cells, thereby affecting the expression of desmoglein-2 related with epithelial barrier function, which may lead to alterations in behavioral responses to HCl via somatosensory nerves.
Collapse
Affiliation(s)
- Asami Oike
- grid.177174.30000 0001 2242 4849Section of Oral Neuroscience, Graduate School of Dental Science, Kyushu University, Fukuoka, Japan ,grid.177174.30000 0001 2242 4849Section of Interdisciplinary Dentistry, Graduate School of Dental Science, Kyushu University, Fukuoka, Japan
| | - Shusuke Iwata
- grid.177174.30000 0001 2242 4849Section of Oral Neuroscience, Graduate School of Dental Science, Kyushu University, Fukuoka, Japan ,grid.177174.30000 0001 2242 4849Research and Development Center for Five-Sense Devices, Kyushu University, Fukuoka, Japan
| | - Ayaka Hirayama
- grid.177174.30000 0001 2242 4849Section of Oral Neuroscience, Graduate School of Dental Science, Kyushu University, Fukuoka, Japan
| | - Yurika Ono
- grid.177174.30000 0001 2242 4849Section of Oral Neuroscience, Graduate School of Dental Science, Kyushu University, Fukuoka, Japan
| | - Yuki Nagasato
- grid.177174.30000 0001 2242 4849Section of Oral Neuroscience, Graduate School of Dental Science, Kyushu University, Fukuoka, Japan
| | - Yuko Kawabata
- grid.177174.30000 0001 2242 4849Department of Cell Biology, Aging Science, and Pharmacology, Division of Oral Biological Sciences, Faculty of Dental Science, Kyushu University, Fukuoka, Japan
| | - Shingo Takai
- grid.177174.30000 0001 2242 4849Section of Oral Neuroscience, Graduate School of Dental Science, Kyushu University, Fukuoka, Japan
| | - Keisuke Sanematsu
- grid.177174.30000 0001 2242 4849Section of Oral Neuroscience, Graduate School of Dental Science, Kyushu University, Fukuoka, Japan ,grid.177174.30000 0001 2242 4849Research and Development Center for Five-Sense Devices, Kyushu University, Fukuoka, Japan ,grid.177174.30000 0001 2242 4849Oral Health/Brain Health/Total Health Research Center, Faculty of Dental Science, Kyushu University, Fukuoka, Japan
| | - Naohisa Wada
- grid.177174.30000 0001 2242 4849Section of Interdisciplinary Dentistry, Graduate School of Dental Science, Kyushu University, Fukuoka, Japan
| | - Noriatsu Shigemura
- grid.177174.30000 0001 2242 4849Section of Oral Neuroscience, Graduate School of Dental Science, Kyushu University, Fukuoka, Japan ,grid.177174.30000 0001 2242 4849Research and Development Center for Five-Sense Devices, Kyushu University, Fukuoka, Japan
| |
Collapse
|
6
|
Nourse JL, Leung VM, Abuwarda H, Evans EL, Izquierdo-Ortiz E, Ly AT, Truong N, Smith S, Bhavsar H, Bertaccini G, Monuki ES, Panicker MM, Pathak MM. Piezo1 regulates cholesterol biosynthesis to influence neural stem cell fate during brain development. J Gen Physiol 2022; 154:213449. [PMID: 36069933 PMCID: PMC9458470 DOI: 10.1085/jgp.202213084] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2022] [Accepted: 07/28/2022] [Indexed: 02/02/2023] Open
Abstract
Mechanical forces and tissue mechanics influence the morphology of the developing brain, but the underlying molecular mechanisms have been elusive. Here, we examine the role of mechanotransduction in brain development by focusing on Piezo1, a mechanically activated ion channel. We find that Piezo1 deletion results in a thinner neuroepithelial layer, disrupts pseudostratification, and reduces neurogenesis in E10.5 mouse embryos. Proliferation and differentiation of Piezo1 knockout (KO) mouse neural stem cells (NSCs) isolated from E10.5 embryos are reduced in vitro compared to littermate WT NSCs. Transcriptome analysis of E10.5 Piezo1 KO brains reveals downregulation of the cholesterol biosynthesis superpathway, in which 16 genes, including Hmgcr, the gene encoding the rate-limiting enzyme of the cholesterol biosynthesis pathway, are downregulated by 1.5-fold or more. Consistent with this finding, membrane lipid composition is altered, and the cholesterol levels are reduced in Piezo1 KO NSCs. Cholesterol supplementation of Piezo1 KO NSCs partially rescues the phenotype in vitro. These findings demonstrate a role for Piezo1 in the neurodevelopmental process that modulates the quantity, quality, and organization of cells by influencing cellular cholesterol metabolism. Our study establishes a direct link in NSCs between PIEZO1, intracellular cholesterol levels, and neural development.
Collapse
Affiliation(s)
- Jamison L. Nourse
- Department of Physiology and Biophysics, University of California, Irvine, Irvine, CA,Sue and Bill Gross Stem Cell Research Center, University of California, Irvine, Irvine, CA
| | - Vivian M. Leung
- Department of Physiology and Biophysics, University of California, Irvine, Irvine, CA,Sue and Bill Gross Stem Cell Research Center, University of California, Irvine, Irvine, CA
| | - Hamid Abuwarda
- Department of Physiology and Biophysics, University of California, Irvine, Irvine, CA,Sue and Bill Gross Stem Cell Research Center, University of California, Irvine, Irvine, CA
| | - Elizabeth L. Evans
- Department of Physiology and Biophysics, University of California, Irvine, Irvine, CA,Sue and Bill Gross Stem Cell Research Center, University of California, Irvine, Irvine, CA
| | - Esmeralda Izquierdo-Ortiz
- Department of Physiology and Biophysics, University of California, Irvine, Irvine, CA,Sue and Bill Gross Stem Cell Research Center, University of California, Irvine, Irvine, CA
| | - Alan T. Ly
- Department of Physiology and Biophysics, University of California, Irvine, Irvine, CA,Sue and Bill Gross Stem Cell Research Center, University of California, Irvine, Irvine, CA
| | - Nguyen Truong
- Department of Physiology and Biophysics, University of California, Irvine, Irvine, CA,Sue and Bill Gross Stem Cell Research Center, University of California, Irvine, Irvine, CA
| | - Samantha Smith
- Department of Physiology and Biophysics, University of California, Irvine, Irvine, CA,Sue and Bill Gross Stem Cell Research Center, University of California, Irvine, Irvine, CA
| | - Harsh Bhavsar
- Department of Physiology and Biophysics, University of California, Irvine, Irvine, CA,Sue and Bill Gross Stem Cell Research Center, University of California, Irvine, Irvine, CA
| | - Gabriella Bertaccini
- Department of Physiology and Biophysics, University of California, Irvine, Irvine, CA,Sue and Bill Gross Stem Cell Research Center, University of California, Irvine, Irvine, CA
| | - Edwin S. Monuki
- Sue and Bill Gross Stem Cell Research Center, University of California, Irvine, Irvine, CA,Department of Pathology and Laboratory Medicine, University of California, Irvine, Irvine, CA
| | - Mitradas M. Panicker
- Department of Physiology and Biophysics, University of California, Irvine, Irvine, CA,Sue and Bill Gross Stem Cell Research Center, University of California, Irvine, Irvine, CA
| | - Medha M. Pathak
- Department of Physiology and Biophysics, University of California, Irvine, Irvine, CA,Sue and Bill Gross Stem Cell Research Center, University of California, Irvine, Irvine, CA,Center for Complex Biological Systems, University of California, Irvine, Irvine, CA,Department of Biomedical Engineering, University of California, Irvine, Irvine, CA,Correspondence to Medha M. Pathak:
| |
Collapse
|
7
|
Abe RJ, Abe JI, Nguyen MTH, Olmsted-Davis EA, Mamun A, Banerjee P, Cooke JP, Fang L, Pownall H, Le NT. Free Cholesterol Bioavailability and Atherosclerosis. Curr Atheroscler Rep 2022; 24:323-336. [PMID: 35332444 PMCID: PMC9050774 DOI: 10.1007/s11883-022-01011-z] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/02/2022] [Indexed: 11/30/2022]
Abstract
PURPOSE OF REVIEW As both a cholesterol acceptor and carrier in the reverse cholesterol transport (RCT) pathway, high-density lipoprotein (HDL) is putatively atheroprotective. However, current pharmacological therapies to increase plasma HDL cholesterol (HDL-c) concentration have paradoxically failed to prevent or reduce atherosclerosis and cardiovascular disease (CVD). Given that free cholesterol (FC) transfer between surfaces of lipoproteins and cells is reversible, excess plasma FC can be transferred to the cells of peripheral tissue sites resulting in atherosclerosis. Here, we summarize potential mechanisms contributing to this paradox and highlight the role of excess free cholesterol (FC) bioavailability in atherosclerosis vs. atheroprotection. RECENT FINDINGS Recent findings have established a complex relationship between HDL-c concentration and atherosclerosis. Systemic scavenger receptor class B type 1 (SR-B1) knock out (KO) mice exhibit with increased diet-induced atherosclerosis despite having an elevated plasma HDL-c concentration compared to wild type (WT) mice. The greater bioavailability of HDL-FC in SR-B1 vs. WT mice is associated with a higher FC content in multiple cell types and tissue sites. These results suggest that dysfunctional HDL with high FC bioavailability is atheroprone despite high HDL-c concentration. Past oversimplification of HDL-c involvement in cholesterol transport has led to the failures in HDL targeted therapy. Evidence suggests that FC-mediated functionality of HDL is of higher importance than its quantity; as a result, deciphering the regulatory mechanisms by which HDL-FC bioavailability can induce atherosclerosis can have far-reaching clinical implications.
Collapse
Affiliation(s)
- Rei J Abe
- Center for Cardiovascular Sciences, Houston Methodist Research Institute, Houston, TX, USA
- Department of Physiology and Biophysics, Weill Cornell Medicine, New York, NY, USA
| | - Jun-Ichi Abe
- Department of Cardiology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Minh T H Nguyen
- Center for Cardiovascular Sciences, Houston Methodist Research Institute, Houston, TX, USA
- University of Science and Technology of Hanoi, Vietnam Academy of Science and Technology, Hanoi, Vietnam
| | | | - Abrar Mamun
- Center for Cardiovascular Sciences, Houston Methodist Research Institute, Houston, TX, USA
| | - Priyanka Banerjee
- Center for Cardiovascular Sciences, Houston Methodist Research Institute, Houston, TX, USA
| | - John P Cooke
- Center for Cardiovascular Sciences, Houston Methodist Research Institute, Houston, TX, USA
- Weill Cornell Medicine, New York, NY, USA
| | - Longhou Fang
- Center for Cardiovascular Sciences, Houston Methodist Research Institute, Houston, TX, USA
- Weill Cornell Medicine, New York, NY, USA
| | - Henry Pownall
- Weill Cornell Medicine, New York, NY, USA
- Center for Bioenergetics, Department of Medicine, Houston Methodist Research Institute, Houston, TX, USA
| | - Nhat-Tu Le
- Center for Cardiovascular Sciences, Houston Methodist Research Institute, Houston, TX, USA.
- Weill Cornell Medicine, New York, NY, USA.
| |
Collapse
|
8
|
Doerfler AM, Han J, Jarrett KE, Tang L, Jain A, Saltzman A, De Giorgi M, Chuecos M, Hurley AE, Li A, Morand P, Ayala C, Goodlett DR, Malovannaya A, Martin JF, de Aguiar Vallim TQ, Shroyer N, Lagor WR. Intestinal Deletion of 3-Hydroxy-3-Methylglutaryl-Coenzyme A Reductase Promotes Expansion of the Resident Stem Cell Compartment. Arterioscler Thromb Vasc Biol 2022; 42:381-394. [PMID: 35172604 PMCID: PMC8957608 DOI: 10.1161/atvbaha.122.317320] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2020] [Accepted: 01/21/2022] [Indexed: 11/16/2022]
Abstract
BACKGROUND The intestine occupies the critical interface between cholesterol absorption and excretion. Surprisingly little is known about the role of de novo cholesterol synthesis in this organ, and its relationship to whole body cholesterol homeostasis. Here, we investigate the physiological importance of this pathway through genetic deletion of the rate-limiting enzyme. METHODS Mice lacking 3-hydroxy-3-methylglutaryl-coenzyme A reductase (Hmgcr) in intestinal villus and crypt epithelial cells were generated using a Villin-Cre transgene. Plasma lipids, intestinal morphology, mevalonate pathway metabolites, and gene expression were analyzed. RESULTS Mice with intestine-specific loss of Hmgcr were markedly smaller at birth, but gain weight at a rate similar to wild-type littermates, and are viable and fertile into adulthood. Intestine lengths and weights were greater relative to body weight in both male and female Hmgcr intestinal knockout mice. Male intestinal knockout had decreased plasma cholesterol levels, whereas fasting triglycerides were lower in both sexes. Lipidomics revealed substantial reductions in numerous nonsterol isoprenoids and sterol intermediates within the epithelial layer, but cholesterol levels were preserved. Hmgcr intestinal knockout mice also showed robust activation of SREBP-2 (sterol-regulatory element binding protein-2) target genes in the epithelium, including the LDLR (low-density lipoprotein receptor). At the cellular level, loss of Hmgcr is compensated for quickly after birth through a dramatic expansion of the stem cell compartment, which persists into adulthood. CONCLUSIONS Loss of Hmgcr in the intestine is compatible with life through compensatory increases in intestinal absorptive surface area, LDLR expression, and expansion of the resident stem cell compartment.
Collapse
Affiliation(s)
- Alexandria M. Doerfler
- Molecular Physiology and Biophysics Graduate Program, Baylor College of Medicine, Houston, Texas, USA
- Department of Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, Texas, USA
| | - Jun Han
- University of Victoria - Genome British Columbia Proteomics Centre, Victoria, British Columbia, Canada
- Division of Medical Sciences, University of Victoria, Victoria, British Columbia, Canada
| | - Kelsey E. Jarrett
- Department of Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, Texas, USA
- Integrative Molecular and Biomedical Sciences Graduate Program, Baylor College of Medicine, Houston, Texas, USA
- Department of Medicine, Division of Cardiology, University of California Los Angeles, Los Angeles, USA
| | - Li Tang
- Department of Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, Texas, USA
- Hunan Provincial Key Lab on Bioinformatics, School of Computer Science and Engineering, Central South University, Changsha 410083, China
| | - Antrix Jain
- Mass Spectrometry Proteomics Core, Baylor College of Medicine, Houston, Texas, USA
| | - Alexander Saltzman
- Mass Spectrometry Proteomics Core, Baylor College of Medicine, Houston, Texas, USA
| | - Marco De Giorgi
- Department of Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, Texas, USA
| | - Marcel Chuecos
- Department of Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, Texas, USA
- Translational Biology and Molecular Medicine Graduate Program, Baylor College of Medicine, Houston, Texas, USA
| | - Ayrea E. Hurley
- Department of Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, Texas, USA
| | - Ang Li
- Department of Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, Texas, USA
- Department of Bioengineering, Rice University, Houston, Texas, USA
| | - Pauline Morand
- Department of Biological Chemistry, University of California Los Angeles, Los Angeles, USA
| | - Claudia Ayala
- Department of Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, Texas, USA
| | - David R. Goodlett
- University of Victoria - Genome British Columbia Proteomics Centre, Victoria, British Columbia, Canada
- Department of Biochemistry & Microbiology, University of Victoria, Victoria, British Columbia, Canada
| | - Anna Malovannaya
- Mass Spectrometry Proteomics Core, Baylor College of Medicine, Houston, Texas, USA
- Verna and Marrs McLean Department of Biochemistry and Molecular Biology, Baylor College of Medicine, Houston, Texas, USA
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas, USA
- Dan L Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, Texas, USA
| | - James F. Martin
- Molecular Physiology and Biophysics Graduate Program, Baylor College of Medicine, Houston, Texas, USA
- Department of Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, Texas, USA
- Integrative Molecular and Biomedical Sciences Graduate Program, Baylor College of Medicine, Houston, Texas, USA
- Translational Biology and Molecular Medicine Graduate Program, Baylor College of Medicine, Houston, Texas, USA
- Cardiomyocyte Renewal Laboratory, Texas Heart Institute, Houston, Texas, USA
- Program in Developmental Biology, Baylor College of Medicine, Houston, Texas, USA
- Cardiovascular Research Institute, Baylor College of Medicine, Houston, Texas USA
| | - Thomas Q. de Aguiar Vallim
- Department of Medicine, Division of Cardiology, University of California Los Angeles, Los Angeles, USA
- Department of Biological Chemistry, University of California Los Angeles, Los Angeles, USA
- Molecular Biology Institute, University of California Los Angeles, Los Angeles, USA
- Johnsson Comprehensive Cancer Center, University of California Los Angeles, Los Angeles, USA
| | - Noah Shroyer
- Integrative Molecular and Biomedical Sciences Graduate Program, Baylor College of Medicine, Houston, Texas, USA
- Translational Biology and Molecular Medicine Graduate Program, Baylor College of Medicine, Houston, Texas, USA
- Department of Medicine, Section of Gastroenterology and Hepatology, Baylor College of Medicine, Houston, Texas, USA
| | - William R. Lagor
- Molecular Physiology and Biophysics Graduate Program, Baylor College of Medicine, Houston, Texas, USA
- Department of Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, Texas, USA
- Integrative Molecular and Biomedical Sciences Graduate Program, Baylor College of Medicine, Houston, Texas, USA
- Translational Biology and Molecular Medicine Graduate Program, Baylor College of Medicine, Houston, Texas, USA
- Department of Bioengineering, Rice University, Houston, Texas, USA
- Cardiovascular Research Institute, Baylor College of Medicine, Houston, Texas USA
| |
Collapse
|
9
|
Zhang Y, Day K, Absher DM. STAT3-mediated allelic imbalance of novel genetic variant Rs1047643 and B-cell-specific super-enhancer in association with systemic lupus erythematosus. eLife 2022; 11:72837. [PMID: 35188103 PMCID: PMC8884724 DOI: 10.7554/elife.72837] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2021] [Accepted: 02/18/2022] [Indexed: 11/24/2022] Open
Abstract
Mapping of allelic imbalance (AI) at heterozygous loci has the potential to establish links between genetic risk for disease and biological function. Leveraging multi-omics data for AI analysis and functional annotation, we discovered a novel functional risk variant rs1047643 at 8p23 in association with systemic lupus erythematosus (SLE). This variant displays dynamic AI of chromatin accessibility and allelic expression on FDFT1 gene in B cells with SLE. We further found a B-cell restricted super-enhancer (SE) that physically contacts with this SNP-residing locus, an interaction that also appears specifically in B cells. Quantitative analysis of chromatin accessibility and DNA methylation profiles further demonstrated that the SE exhibits aberrant activity in B cell development with SLE. Functional studies identified that STAT3, a master factor associated with autoimmune diseases, directly regulates both the AI of risk variant and the activity of SE in cultured B cells. Our study reveals that STAT3-mediated SE activity and cis-regulatory effects of SNP rs1047643 at 8p23 locus are associated with B cell deregulation in SLE.
Collapse
Affiliation(s)
- Yanfeng Zhang
- HudsonAlpha Institute for Biotechnology, Huntsville, United States
| | | | - Devin M Absher
- HudsonAlpha Institute for Biotechnology, Huntsville, United States
| |
Collapse
|
10
|
Lauschke K, Dalgaard MD, Emnéus J, Vinggaard AM. Transcriptomic changes upon epoxiconazole exposure in a human stem cell-based model of developmental toxicity. CHEMOSPHERE 2021; 284:131225. [PMID: 34182286 DOI: 10.1016/j.chemosphere.2021.131225] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/12/2021] [Revised: 06/07/2021] [Accepted: 06/11/2021] [Indexed: 06/13/2023]
Abstract
Conazole fungicides such as epoxiconazole are mostly used on cereals of crops to inhibit fungal growth through direct inhibition of sterol 14α-demethylase (CYP51A1). However, this enzyme is highly conserved and in humans it is part of the steroid hormone biosynthesis pathway. Endocrine disrupting effects of epoxiconazole have been shown in rodents and have been substantiated by in vitro data, however, the underlying molecular mechanisms are not clear. We took advantage of a human stem cell based in vitro model for developmental toxicity to study the molecular effects of epoxiconazole. This model is based on 3D cultures of embryoid bodies and differentiation into cardiomyocytes, which mimics the early stages of embryonic development. We have previously shown that epoxiconazole impairs differentiation of these embryoid bodies and therefore has the potential to affect human embryonic development. We employed global transcriptome analysis using RNA sequencing and found that the steroid biosynthesis pathway including CYP51A1, the human sterol 14α-demethylase, was highly deregulated by epoxiconazole in our model. We confirmed that most genes of the steroid biosynthesis pathway were upregulated, including CYP51A1, suggesting a compensatory mechanism at the gene expression level. Our data suggest that epoxiconazole acts mainly by decreasing cholesterol biosynthesis in the cells. We conclude that epoxiconazole bears the potential to harm human embryonic development through inhibition of the steroid biosynthesis pathway. As this may be a common feature of compounds that target sterol 14α-demethylase, we add evidence to the assumption that conazole fungicides may be human developmental toxicants.
Collapse
Affiliation(s)
- Karin Lauschke
- National Food Institute, Technical University of Denmark, Denmark; Department for Biotechnology and Biomedicine, Technical University of Denmark, Denmark
| | | | - Jenny Emnéus
- Department for Biotechnology and Biomedicine, Technical University of Denmark, Denmark
| | | |
Collapse
|
11
|
Takemoto Y, Kadota S, Minami I, Otsuka S, Okuda S, Abo M, Punzalan LL, Shen Y, Shiba Y, Uesugi M. Chemical Genetics Reveals a Role of Squalene Synthase in TGFβ Signaling and Cardiomyogenesis. Angew Chem Int Ed Engl 2021. [DOI: 10.1002/ange.202100523] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Affiliation(s)
- Yasushi Takemoto
- Institute for Chemical Research (ICR) Kyoto University Uji Kyoto 611-0011 Japan
| | - Shin Kadota
- Institute for Biomedical Sciences Shinshu University Matsumoto, Nagano 390-8621 Japan
| | - Itsunari Minami
- Institute for Integrated Cell-Material Sciences (WPI-iCeMS) Kyoto University Kyoto 606-8501 Japan
| | - Shinya Otsuka
- Institute for Chemical Research (ICR) Kyoto University Uji Kyoto 611-0011 Japan
| | - Satoshi Okuda
- Institute for Chemical Research (ICR) Kyoto University Uji Kyoto 611-0011 Japan
| | - Masahiro Abo
- Institute for Chemical Research (ICR) Kyoto University Uji Kyoto 611-0011 Japan
| | - Louvy Lynn Punzalan
- Institute for Chemical Research (ICR) Kyoto University Uji Kyoto 611-0011 Japan
| | - Yan Shen
- Institute for Chemical Research (ICR) Kyoto University Uji Kyoto 611-0011 Japan
| | - Yuji Shiba
- Institute for Biomedical Sciences Shinshu University Matsumoto, Nagano 390-8621 Japan
| | - Motonari Uesugi
- Institute for Chemical Research (ICR) Kyoto University Uji Kyoto 611-0011 Japan
- Institute for Integrated Cell-Material Sciences (WPI-iCeMS) Kyoto University Kyoto 606-8501 Japan
- School of Pharmacy Fudan University Shanghai 201203 China
| |
Collapse
|
12
|
Takemoto Y, Kadota S, Minami I, Otsuka S, Okuda S, Abo M, Punzalan LL, Shen Y, Shiba Y, Uesugi M. Chemical Genetics Reveals a Role of Squalene Synthase in TGFβ Signaling and Cardiomyogenesis. Angew Chem Int Ed Engl 2021; 60:21824-21831. [PMID: 34374184 DOI: 10.1002/anie.202100523] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2021] [Indexed: 11/11/2022]
Abstract
KY02111 is a widely used small molecule that boosts cardiomyogenesis of the mesoderm cells derived from pluripotent stem cells, yet its molecular mechanism of action remains elusive. The present study resolves the initially perplexing effects of KY02111 on Wnt signaling and subsequently identifies squalene synthase (SQS) as a molecular target of KY02111 and its optimized version, KY-I. By disrupting the interaction of SQS with cardiac ER-membrane protein TMEM43, KY02111 impairs TGFβ signaling, but not Wnt signaling, and thereby recapitulates the clinical mutation of TMEM43 that causes arrhythmogenic right ventricular cardiomyopathy (ARVC), an inherited heart disease that involves a substitution of myocardium with fatty tissue. These findings reveal a heretofore undescribed role of SQS in TGFβ signaling and cardiomyogenesis. KY02111 may find its use in ARVC modeling as well as serve as a chemical tool for studying TGFβ/SMAD signaling.
Collapse
Affiliation(s)
- Yasushi Takemoto
- Institute for Chemical Research (ICR), Kyoto University, Uji, Kyoto 611-0011, Japan
| | - Shin Kadota
- Institute for Biomedical Sciences, Shinshu University, Matsumoto, Nagano, 390-8621, Japan
| | - Itsunari Minami
- Institute for Integrated Cell-Material Sciences (WPI-iCeMS), Kyoto University, Kyoto, 606-8501, Japan
| | - Shinya Otsuka
- Institute for Chemical Research (ICR), Kyoto University, Uji, Kyoto 611-0011, Japan
| | - Satoshi Okuda
- Institute for Chemical Research (ICR), Kyoto University, Uji, Kyoto 611-0011, Japan
| | - Masahiro Abo
- Institute for Chemical Research (ICR), Kyoto University, Uji, Kyoto 611-0011, Japan
| | - Louvy Lynn Punzalan
- Institute for Chemical Research (ICR), Kyoto University, Uji, Kyoto 611-0011, Japan
| | - Yan Shen
- Institute for Chemical Research (ICR), Kyoto University, Uji, Kyoto 611-0011, Japan
| | - Yuji Shiba
- Institute for Biomedical Sciences, Shinshu University, Matsumoto, Nagano, 390-8621, Japan
| | - Motonari Uesugi
- Institute for Chemical Research (ICR), Kyoto University, Uji, Kyoto 611-0011, Japan.,Institute for Integrated Cell-Material Sciences (WPI-iCeMS), Kyoto University, Kyoto, 606-8501, Japan.,School of Pharmacy, Fudan University, Shanghai, 201203, China
| |
Collapse
|
13
|
Dang E, Man G, Lee D, Crumrine DA, Mauro TM, Elias PM, Man MQ. Mutations in 3β-hydroxysteroid-δ8, δ7-isomerase paradoxically benefit epidermal permeability barrier homeostasis in mice. Exp Dermatol 2020; 30:384-389. [PMID: 33205489 DOI: 10.1111/exd.14236] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2020] [Revised: 11/03/2020] [Accepted: 11/09/2020] [Indexed: 11/27/2022]
Abstract
Inherited or acquired blockade of distal steps in the cholesterol synthetic pathway results in ichthyosis, due to reduced cholesterol production and/or the accumulation of toxic metabolic precursors, while inhibition of epidermal cholesterol synthesis compromises epidermal permeability barrier homeostasis. We showed here that 3β-hydroxysteroid-δ8, δ7-isomerase-deficient mice (TD), an analog for CHILD syndrome in humans, exhibited not only lower basal transepidermal water loss rates, but also accelerated permeability barrier recovery despite the lower expression levels of mRNA for epidermal differentiation marker-related proteins and lipid synthetic enzymes. Moreover, TD mice displayed low skin surface pH, paralleled by increased expression levels of mRNA for sodium/hydrogen exchanger 1 (NHE1) and increased antimicrobial peptide expression, compared with wild-type (WT) mice, which may compensate for the decreased differentiation and lipid synthesis. Additionally, in comparison with WT controls, TD mice showed a significant reduction in ear thickness following challenges with either phorbol ester or oxazolone. However, TD mice exhibited growth retardation. Together, these results demonstrate that 3β-hydroxysteroid-δ8, δ7-isomerase deficiency does not compromise epidermal permeability barrier in mice, suggesting that alterations in epidermal function depend on which step of the cholesterol synthetic pathway is interrupted. But whether these findings in mice could be mirrored in humans remains to be determined.
Collapse
Affiliation(s)
- Erle Dang
- Dermatology Services, Veterans Affairs Medical Center, University of California San Francisco, CA, USA.,Department of Dermatology, Xijing Hospital, The Fourth Military Medical University, Xi'an, China
| | - George Man
- Dermatology Services, Veterans Affairs Medical Center, University of California San Francisco, CA, USA
| | - Dale Lee
- Dermatology Services, Veterans Affairs Medical Center, University of California San Francisco, CA, USA
| | - Debbie A Crumrine
- Dermatology Services, Veterans Affairs Medical Center, University of California San Francisco, CA, USA
| | - Theodora M Mauro
- Dermatology Services, Veterans Affairs Medical Center, University of California San Francisco, CA, USA
| | - Peter M Elias
- Dermatology Services, Veterans Affairs Medical Center, University of California San Francisco, CA, USA
| | - Mao-Qiang Man
- Dermatology Services, Veterans Affairs Medical Center, University of California San Francisco, CA, USA
| |
Collapse
|
14
|
Role of Metabolism in Bone Development and Homeostasis. Int J Mol Sci 2020; 21:ijms21238992. [PMID: 33256181 PMCID: PMC7729585 DOI: 10.3390/ijms21238992] [Citation(s) in RCA: 52] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2020] [Revised: 11/22/2020] [Accepted: 11/25/2020] [Indexed: 02/07/2023] Open
Abstract
Carbohydrates, fats, and proteins are the underlying energy sources for animals and are catabolized through specific biochemical cascades involving numerous enzymes. The catabolites and metabolites in these metabolic pathways are crucial for many cellular functions; therefore, an imbalance and/or dysregulation of these pathways causes cellular dysfunction, resulting in various metabolic diseases. Bone, a highly mineralized organ that serves as a skeleton of the body, undergoes continuous active turnover, which is required for the maintenance of healthy bony components through the deposition and resorption of bone matrix and minerals. This highly coordinated event is regulated throughout life by bone cells such as osteoblasts, osteoclasts, and osteocytes, and requires synchronized activities from different metabolic pathways. Here, we aim to provide a comprehensive review of the cellular metabolism involved in bone development and homeostasis, as revealed by mouse genetic studies.
Collapse
|
15
|
Şahİn G, Tellİ M, ÜnlÜ ES, Pehlİvan KarakaŞ F. Effects of moderate high temperature and UV-B on accumulation of withanolides and relative expression of the squalene synthase gene in Physalis peruviana. ACTA ACUST UNITED AC 2020; 44:295-303. [PMID: 33110367 PMCID: PMC7585162 DOI: 10.3906/biy-2002-69] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2020] [Accepted: 06/05/2020] [Indexed: 11/21/2022]
Abstract
Physalis peruviana
L. (Cape gooseberry) is a source for a variety of phytocompounds such as withanolides, withanone, withaferin A, and withanolide A. These withanolides are high-value drug candidates due to their various pharmacological properties. To meet the increasing demands for these compounds, plant cell technology offers a reliable alternative. Exogenous addition of elicitors is considered the most effective strategy for enhanced production of secondary metabolites. In this study, we investigated changes in withanolide accumulation and characterized the gene expression level changes of squalene synthase enzyme in
P. peruviana
shoot cultures exposed to mild nonlethal heat stress (45°C for 2 and 5 h) and UV-B radiation (313 nm for 15 min and 3 h). We demonstrated significant changes in withanolide content with 7.86- and 12.5-fold increases for 2- and 5-hmild high-temperature exposure times, respectively. Exposure to UV-B also changed the withanolide content by 7.22- and 7-fold increases for 15 min and 3 h exposure times, respectively. The relative expression level of squalene synthase gene showed consistent results with1.80- and 10.13-fold increases in withanolide for 2- and 5-h mild high-temperature exposure times, and 1.34- and 2.01-fold increases with 15 min and 3 h UV-B exposure times, respectively.
Collapse
Affiliation(s)
- Günce Şahİn
- Department of Biology, Faculty of Arts and Science, Bolu Abant İzzet Baysal University, Bolu Turkey
| | - Murat Tellİ
- Department of Biology, Faculty of Arts and Science, Bolu Abant İzzet Baysal University, Bolu Turkey
| | - Ercan Selçuk ÜnlÜ
- Department of Chemisty, Faculty of Arts and Science, Bolu Abant İzzet Baysal University, Bolu Turkey
| | - Fatma Pehlİvan KarakaŞ
- Department of Biology, Faculty of Arts and Science, Bolu Abant İzzet Baysal University, Bolu Turkey
| |
Collapse
|
16
|
Ha NT, Lee CH. Roles of Farnesyl-Diphosphate Farnesyltransferase 1 in Tumour and Tumour Microenvironments. Cells 2020; 9:cells9112352. [PMID: 33113804 PMCID: PMC7693003 DOI: 10.3390/cells9112352] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Revised: 10/23/2020] [Accepted: 10/24/2020] [Indexed: 12/14/2022] Open
Abstract
Farnesyl-diphosphate farnesyltransferase 1 (FDFT1, squalene synthase), a membrane-associated enzyme, synthesizes squalene via condensation of two molecules of farnesyl pyrophosphate. Accumulating evidence has noted that FDFT1 plays a critical role in cancer, particularly in metabolic reprogramming, cell proliferation, and invasion. Based on these advances in our knowledge, FDFT1 could be a potential target for cancer treatment. This review focuses on the contribution of FDFT1 to the hallmarks of cancer, and further, we discuss the applicability of FDFT1 as a cancer prognostic marker and target for anticancer therapy.
Collapse
|
17
|
Sherwood WB, Kothalawala DM, Kadalayil L, Ewart S, Zhang H, Karmaus W, Arshad SH, Holloway JW, Rezwan FI. Epigenome-Wide Association Study Reveals Duration of Breastfeeding Is Associated with Epigenetic Differences in Children. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2020; 17:E3569. [PMID: 32443666 PMCID: PMC7277240 DOI: 10.3390/ijerph17103569] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/20/2020] [Revised: 05/15/2020] [Accepted: 05/17/2020] [Indexed: 12/21/2022]
Abstract
Several small studies have shown associations between breastfeeding and genome-wide DNA methylation (DNAm). We performed a comprehensive Epigenome-Wide Association Study (EWAS) to identify associations between breastfeeding and DNAm patterns in childhood. We analysed DNAm data from the Isle of Wight Birth Cohort at birth, 10, 18 and 26 years. The feeding method was categorized as breastfeeding duration >3 months and >6 months, and exclusive breastfeeding duration >3 months. EWASs using robust linear regression were performed to identify differentially methylated positions (DMPs) in breastfed and non-breastfed children at age 10 (false discovery rate of 5%). Differentially methylated regions (DMRs) were identified using comb-p. The persistence of significant associations was evaluated in neonates and individuals at 18 and 26 years. Two DMPs, in genes SNX25 and LINC00840, were significantly associated with breastfeeding duration >6 months at 10 years and was replicated for >3 months of exclusive breastfeeding. Additionally, a significant DMR spanning the gene FDFT1 was identified in 10-year-old children who were exposed to a breastfeeding duration >3 months. None of these signals persisted to 18 or 26 years. This study lends further support for a suggestive role of DNAm in the known benefits of breastfeeding on a child's future health.
Collapse
Affiliation(s)
- William B. Sherwood
- Human Development and Health, Faculty of Medicine, University of Southampton, Southampton SO16 6YD, UK; (W.B.S.); (D.M.K.); (L.K.); (F.I.R.)
| | - Dilini M. Kothalawala
- Human Development and Health, Faculty of Medicine, University of Southampton, Southampton SO16 6YD, UK; (W.B.S.); (D.M.K.); (L.K.); (F.I.R.)
- NIHR Southampton Biomedical Research Centre, University Hospital Southampton NHS Foundation Trust, Southampton SO16 6YD, UK;
| | - Latha Kadalayil
- Human Development and Health, Faculty of Medicine, University of Southampton, Southampton SO16 6YD, UK; (W.B.S.); (D.M.K.); (L.K.); (F.I.R.)
| | - Susan Ewart
- College of Veterinary Medicine, Michigan State University, East Lansing, MI 48824, USA;
| | - Hongmei Zhang
- Division of Epidemiology, Biostatistics and Environmental Health, School of Public Health, University of Memphis, 236A Robison Hall, Memphis, TN 38152, USA; (H.Z.); (W.K.)
| | - Wilfried Karmaus
- Division of Epidemiology, Biostatistics and Environmental Health, School of Public Health, University of Memphis, 236A Robison Hall, Memphis, TN 38152, USA; (H.Z.); (W.K.)
| | - S. Hasan Arshad
- NIHR Southampton Biomedical Research Centre, University Hospital Southampton NHS Foundation Trust, Southampton SO16 6YD, UK;
- Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton SO16 6YD, UK
- The David Hide Asthma and Allergy Research Centre, St Mary’s Hospital, Isle of Wight PO30 5TG UK
| | - John W. Holloway
- Human Development and Health, Faculty of Medicine, University of Southampton, Southampton SO16 6YD, UK; (W.B.S.); (D.M.K.); (L.K.); (F.I.R.)
- NIHR Southampton Biomedical Research Centre, University Hospital Southampton NHS Foundation Trust, Southampton SO16 6YD, UK;
| | - Faisal I. Rezwan
- Human Development and Health, Faculty of Medicine, University of Southampton, Southampton SO16 6YD, UK; (W.B.S.); (D.M.K.); (L.K.); (F.I.R.)
- School of Water, Energy and Environment, Cranfield University, Cranfield, Bedfordshire MK43 0AL, UK
| |
Collapse
|
18
|
Rewiring Neuronal Glycerolipid Metabolism Determines the Extent of Axon Regeneration. Neuron 2019; 105:276-292.e5. [PMID: 31786011 PMCID: PMC6975164 DOI: 10.1016/j.neuron.2019.10.009] [Citation(s) in RCA: 75] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2019] [Revised: 09/11/2019] [Accepted: 10/03/2019] [Indexed: 12/21/2022]
Abstract
How adult neurons coordinate lipid metabolism to regenerate axons remains elusive. We found that depleting neuronal lipin1, a key enzyme controlling the balanced synthesis of glycerolipids through the glycerol phosphate pathway, enhanced axon regeneration after optic nerve injury. Axotomy elevated lipin1 in retinal ganglion cells, which contributed to regeneration failure in the CNS by favorably producing triglyceride (TG) storage lipids rather than phospholipid (PL) membrane lipids in neurons. Regrowth induced by lipin1 depletion required TG hydrolysis and PL synthesis. Decreasing TG synthesis by deleting neuronal diglyceride acyltransferases (DGATs) and enhancing PL synthesis through the Kennedy pathway promoted axon regeneration. In addition, peripheral neurons adopted this mechanism for their spontaneous axon regeneration. Our study reveals a critical role of lipin1 and DGATs as intrinsic regulators of glycerolipid metabolism in neurons and indicates that directing neuronal lipid synthesis away from TG synthesis and toward PL synthesis may promote axon regeneration. Injury-elevated Lipin1 and DGAT in retinal ganglion cells suppress regeneration Neuronal lipin1 and DGATs increase triglyceride and decrease phospholipids Redirecting triacylglyceride to phospholipid synthesis promotes axon regeneration
Collapse
|
19
|
Besnard T, Sloboda N, Goldenberg A, Küry S, Cogné B, Breheret F, Trochu E, Conrad S, Vincent M, Deb W, Balguerie X, Barbarot S, Baujat G, Ben-Omran T, Bursztejn AC, Carmignac V, Datta AN, Delignières A, Faivre L, Gardie B, Guéant JL, Kuentz P, Lenglet M, Nassogne MC, Ramaekers V, Schnur RE, Si Y, Torti E, Thevenon J, Vabres P, Van Maldergem L, Wand D, Wiedemann A, Cariou B, Redon R, Lamazière A, Bézieau S, Feillet F, Isidor B. Biallelic pathogenic variants in the lanosterol synthase gene LSS involved in the cholesterol biosynthesis cause alopecia with intellectual disability, a rare recessive neuroectodermal syndrome. Genet Med 2019; 21:2025-2035. [DOI: 10.1038/s41436-019-0445-x] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2018] [Accepted: 01/14/2019] [Indexed: 12/18/2022] Open
|
20
|
Coman D, Vissers LE, Riley LG, Kwint MP, Hauck R, Koster J, Geuer S, Hopkins S, Hallinan B, Sweetman L, Engelke UF, Burrow TA, Cardinal J, McGill J, Inwood A, Gurnsey C, Waterham HR, Christodoulou J, Wevers RA, Pitt J. Squalene Synthase Deficiency: Clinical, Biochemical, and Molecular Characterization of a Defect in Cholesterol Biosynthesis. Am J Hum Genet 2018; 103:125-130. [PMID: 29909962 DOI: 10.1016/j.ajhg.2018.05.004] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2018] [Accepted: 05/11/2018] [Indexed: 01/12/2023] Open
Abstract
Mendelian disorders of cholesterol biosynthesis typically result in multi-system clinical phenotypes, underlining the importance of cholesterol in embryogenesis and development. FDFT1 encodes for an evolutionarily conserved enzyme, squalene synthase (SS, farnesyl-pyrophosphate farnesyl-transferase 1), which catalyzes the first committed step in cholesterol biosynthesis. We report three individuals with profound developmental delay, brain abnormalities, 2-3 syndactyly of the toes, and facial dysmorphisms, resembling Smith-Lemli-Opitz syndrome, the most common cholesterol biogenesis defect. The metabolite profile in plasma and urine suggested that their defect was at the level of squalene synthase. Whole-exome sequencing was used to identify recessive disease-causing variants in FDFT1. Functional characterization of one variant demonstrated a partial splicing defect and altered promoter and/or enhancer activity, reflecting essential mechanisms for regulating cholesterol biosynthesis/uptake in steady state.
Collapse
|
21
|
Carson RA, Rudine AC, Tally SJ, Franks AL, Frahm KA, Waldman JK, Silswal N, Burale S, Phan JV, Chandran UR, Monaghan AP, DeFranco DB. Statins impact primary embryonic mouse neural stem cell survival, cell death, and fate through distinct mechanisms. PLoS One 2018; 13:e0196387. [PMID: 29738536 PMCID: PMC5940229 DOI: 10.1371/journal.pone.0196387] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2017] [Accepted: 04/12/2018] [Indexed: 12/11/2022] Open
Abstract
Statins inhibit HMG-CoA reductase, the rate-limiting enzyme in the cholesterol biosynthesis pathway (CBP), and are used for the prevention of cardiovascular disease. The anti-inflammatory effects of statins may also provide therapeutic benefits and have led to their use in clinical trials for preeclampsia, a pregnancy-associated inflammatory condition, despite their current classification as category X (i.e. contraindicated during pregnancy). In the developing neocortex, products of the CBP play essential roles in proliferation and differentiation of neural stem-progenitor cells (NSPCs). To understand how statins could impact the developing brain, we studied effects of pravastatin and simvastatin on primary embryonic NSPC survival, proliferation, global transcription, and cell fate in vitro. We found that statins dose dependently decrease NSPC expansion by promoting cell death and autophagy of NSPCs progressing through the G1 phase of the cell cycle. Genome-wide transcriptome analysis demonstrates an increase in expression of CBP genes following pravastatin treatment, through activation of the SREBP2 transcription factor. Co-treatment with farnesyl pyrophosphate (FPP), a CBP metabolite downstream of HMG-CoA reductase, reduces SREBP2 activation and pravastatin-induced PARP cleavage. Finally, pravastatin and simvastatin differentially alter NSPC cell fate and mRNA expression during differentiation, through a non-CBP dependent pathway.
Collapse
Affiliation(s)
- Ross A. Carson
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States of America
| | - Anthony C. Rudine
- Department of Pediatrics, Division of Newborn Medicine, Children’s Hospital of Pittsburgh, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States of America
| | - Serena J. Tally
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States of America
| | - Alexis L. Franks
- Department of Pediatrics, Division of Child Neurology, Children’s Hospital of Pittsburgh, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States of America
| | - Krystle A. Frahm
- Department of Medicine, Division of Endocrinology and Metabolism, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States of America
| | - Jacob K. Waldman
- Department of Biomedical Informatics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States of America
| | - Neerupma Silswal
- Department of Biomedical Sciences, University of Missouri Kansas City School of Medicine, Kansas City, Missouri, United States of America
| | - Suban Burale
- Department of Biomedical Sciences, University of Missouri Kansas City School of Medicine, Kansas City, Missouri, United States of America
| | - James V. Phan
- Department of Biomedical Sciences, University of Missouri Kansas City School of Medicine, Kansas City, Missouri, United States of America
| | - Uma R. Chandran
- Department of Biomedical Informatics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States of America
| | - A. Paula Monaghan
- Department of Biomedical Sciences, University of Missouri Kansas City School of Medicine, Kansas City, Missouri, United States of America
| | - Donald B. DeFranco
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States of America
- * E-mail:
| |
Collapse
|
22
|
Ibrahim MF, Abdelaziz N. Assessment of squalene eligibility in bettering some maternal and fetal disorders instigated by gamma irradiation of rats at mid gestation. Int J Radiat Biol 2018; 94:229-237. [PMID: 29310516 DOI: 10.1080/09553002.2018.1425806] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
PURPOSE Squalene is an eminent vital part of the synthesis of steroid hormones in the body as well as the first specific intermediate in cholesterol biosynthesis that plays an essential role in normal embryogenesis. The present work was designed to test the maternal and embryonic response to the modulating capacity of squalene (0.4 ml/kg/d), when supplemented to rats from days 1 to 18 of pregnancy, against the damaging consequences induced by maternal subjection to 3 Gy gamma irradiation on day 10 post-conception. MATERIALS AND METHODS The experimental protocol comprised of four different pregnant groups, namely: (1) control, (2) squalene supplemented, (3) irradiated and (4) squalene supplemented + irradiated. RESULTS It has been detected that radiation has increased the maternal blood lactate dehydrogenase (as a marker of tissue injury), cholesterol, triglycerides, estradiol and progesterone and has also provoked the oxidative stress that has been demonstrated by the increased malondialdehyde (MDA) and the decreased glutathione (GSH) and superoxide dismutase (SOD). These maternal changes were associated with high embryonic lethality, growth retardation, severe developmental abnormalities and defective neural tube closure expressed by exencephaly. However, squalene treatment has significantly improved the radiation imposed maternal variations and reduced the embryonic mortality, although it has not been able to attenuate the embryonic neural tube defects. CONCLUSIONS It has been presumed that the maternal mid-gestational irradiation (day 10) has affected the fetal nervous system development with concomitant maternal oxidative stress, hyperlipidemia, and increased progesterone and estradiol levels. Squalene uptake has improved the maternal variations and reduced the embryonic mortality while could not stop or improve the embryonic neural tube defects imposed by radiation at this exact radiation timing.
Collapse
Affiliation(s)
- Maha Fawzy Ibrahim
- a Radiation Biology Department , National Centre for Radiation Research and Technology (NCRRT), Atomic Energy Authority , Cairo , Egypt
| | - Nahed Abdelaziz
- a Radiation Biology Department , National Centre for Radiation Research and Technology (NCRRT), Atomic Energy Authority , Cairo , Egypt
| |
Collapse
|
23
|
Alarcon VB, Marikawa Y. Statins inhibit blastocyst formation by preventing geranylgeranylation. Mol Hum Reprod 2016; 22:350-63. [PMID: 26908642 DOI: 10.1093/molehr/gaw011] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2015] [Accepted: 01/29/2016] [Indexed: 12/14/2022] Open
Abstract
STUDY HYPOTHESIS Statins, inhibitors of 3-hydroxy-3-methylglutaryl-coenzyme A (HMG-CoA) reductase of the mevalonate pathway and prescription drugs that treat hypercholesterolemia, compromise preimplantation mouse development via modulation of HIPPO signaling. STUDY FINDING HMG-CoA reductase activity is required for trophectoderm specification, namely blastocyst cavity formation and Yes-associated protein (YAP) nuclear localization, through the production of isoprenoid geranylgeranyl pyrophosphate (GGPP) and the action of geranylgeranyl transferase. WHAT IS KNOWN ALREADY Previous studies have shown that treatment of mouse embryos with mevastatin prevents blastocyst formation, but how HMG-CoA reductase is involved in preimplantation development is unknown. HIPPO signaling regulates specification of the trophectoderm lineage of the mouse blastocyst by controlling the nuclear localization of YAP. In human cell lines, the mevalonate pathway regulates YAP to mediate self-renewal and survival through geranylgeranylation of RHO proteins. These studies suggest that in preimplantation development, statins may act through HIPPO pathway to interfere with trophectoderm specification and thereby inhibit blastocyst formation. STUDY DESIGN, SAMPLES/MATERIALS, METHODS Eight-cell stage (E2.5) mouse embryos were treated in hanging drop culture with chemical agents, namely statins (lovastatin, atorvastatin, cerivastatin and pravastatin), mevalonic acid (MVA), cholesterol, squalene, farnesyl pyrophosphate (FPP), geranylgeranyl pyrophosphate (GGPP), geranylgeranyltransferase inhibitor GGTI-298, RHO inhibitor I, and squalene synthase inhibitor YM-53601, up to the late blastocyst stage (E4.5). Efficiency of blastocyst formation was assessed based on gross morphology and the measurement of the cavity size using an image analysis software. Effects on cell lineages and HIPPO signaling were analyzed using immunohistochemistry with confocal microscopy based on the expression patterns of the lineage-specific markers and the nuclear accumulation of YAP. Effects on cell lineages were also examined by quantitative RT-PCR based on the transcript levels of the lineage-specific marker genes. Data were analyzed using one-way ANOVA and two-sample t-test. MAIN RESULTS AND THE ROLE OF CHANCE All four statins examined inhibited blastocyst formation. The adverse impact of statins was rescued by supplementation of MVA (P < 0.01) or GGPP (P < 0.01) but not squalene nor cholesterol. Blastocyst formation was also prevented by GGTI-298 (P < 0.01). These results indicate that HMG-CoA reductase activity is required for blastocyst formation mainly through the production of GGPP but not cholesterol. Inhibition of RHO proteins, known targets of geranylgeranylation, impaired blastocyst formation, which was not reversed by GGPP supplementation. Nuclear localization of YAP was diminished by statin treatment but fully restored by supplementation of MVA (P < 0.01) or GGPP (P < 0.01). This suggests that HIPPO signaling is regulated by GGPP-dependent mechanisms, possibly geranylgeranylation of RHO, to enable trophectoderm formation. YM-53601 prevented blastocyst formation (P < 0.01), but its adverse impact was not rescued by supplementation of squalene or cholesterol, suggesting that squalene synthesis inhibition was not the cause of blastocyst defects. LIMITATIONS, REASONS FOR CAUTION Analyses were conducted on embryos cultured ex vivo, but they enable the determination of specific concentrations that impair embryo development which can be compared with drug concentrations in the reproductive tract when testing in vivo impact of statins through animal experimentations. Also, analyses were conducted in only one species, the mouse. Epidemiological studies on the effects of various types of statins on the fertility of women are necessary. WIDER IMPLICATIONS OF THE FINDINGS Our study reveals how the mevalonate pathway is required for blastocyst formation and intersects with HIPPO pathway to provide a mechanistic basis for the embryotoxic effect of statins. This bears relevance for women who are taking statins while trying to conceive, since statins have potential to prevent the conceptus from reaching the blastocyst stage and to cause early conceptus demise. LARGE SCALE DATA Not applicable. STUDY FUNDING AND COMPETING INTERESTS This study was supported by grants from the George F. Straub Trust of the Hawaii Community Foundation (13ADVC-60315 to V.B.A.) and the National Institutes of Health, USA (P20GM103457 to V.B.A.). The authors have no conflict of interest to declare.
Collapse
Affiliation(s)
- Vernadeth B Alarcon
- Institute for Biogenesis Research, Department of Anatomy, Biochemistry and Physiology, John A. Burns School of Medicine, University of Hawaii, Honolulu, HI 96813, USA
| | - Yusuke Marikawa
- Institute for Biogenesis Research, Department of Anatomy, Biochemistry and Physiology, John A. Burns School of Medicine, University of Hawaii, Honolulu, HI 96813, USA
| |
Collapse
|
24
|
Rhim JH, Luo X, Xu X, Gao D, Zhou T, Li F, Qin L, Wang P, Xia X, Wong STC. A High-content screen identifies compounds promoting the neuronal differentiation and the midbrain dopamine neuron specification of human neural progenitor cells. Sci Rep 2015; 5:16237. [PMID: 26542303 PMCID: PMC4635364 DOI: 10.1038/srep16237] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2015] [Accepted: 10/14/2015] [Indexed: 12/30/2022] Open
Abstract
Small molecule compounds promoting the neuronal differentiation of stem/progenitor cells are of pivotal importance to regenerative medicine. We carried out a high-content screen to systematically characterize known bioactive compounds, on their effects on the neuronal differentiation and the midbrain dopamine (mDA) neuron specification of neural progenitor cells (NPCs) derived from the ventral mesencephalon of human fetal brain. Among the promoting compounds three major pharmacological classes were identified including the statins, TGF-βRI inhibitors, and GSK-3 inhibitors. The function of each class was also shown to be distinct, either to promote both the neuronal differentiation and mDA neuron specification, or selectively the latter, or promote the former but suppress the latter. We then carried out initial investigation on the possible mechanisms underlying, and demonstrated their applications on NPCs derived from human pluripotent stem cells (PSCs). Our study revealed the potential of several small molecule compounds for use in the directed differentiation of human NPCs. The screening result also provided insight into the signaling network regulating the differentiation of human NPCs.
Collapse
Affiliation(s)
- Ji Heon Rhim
- Chao Center for BRAIN, Department of Systems Medicine and Bioengineering, Houston Methodist Research Institute, 6670 Bertner Avenue, Houston, TX 77030
| | - Xiangjian Luo
- Chao Center for BRAIN, Department of Systems Medicine and Bioengineering, Houston Methodist Research Institute, 6670 Bertner Avenue, Houston, TX 77030
| | - Xiaoyun Xu
- Chao Center for BRAIN, Department of Systems Medicine and Bioengineering, Houston Methodist Research Institute, 6670 Bertner Avenue, Houston, TX 77030
| | - Dongbing Gao
- Chao Center for BRAIN, Department of Systems Medicine and Bioengineering, Houston Methodist Research Institute, 6670 Bertner Avenue, Houston, TX 77030
| | - Tieling Zhou
- Chao Center for BRAIN, Department of Systems Medicine and Bioengineering, Houston Methodist Research Institute, 6670 Bertner Avenue, Houston, TX 77030
| | - Fuhai Li
- Chao Center for BRAIN, Department of Systems Medicine and Bioengineering, Houston Methodist Research Institute, 6670 Bertner Avenue, Houston, TX 77030.,Weill Cornell Medical College, Cornell University, New York, NY 10065
| | - Lidong Qin
- Department of Nanomedicine, Houston Methodist Research Institute, 6670 Bertner Avenue, Houston, TX 77030.,Weill Cornell Medical College, Cornell University, New York, NY 10065
| | - Ping Wang
- Department of Pathology and Genomic Medicine, Houston Methodist Research Institute, Houston, TX 77030.,Weill Cornell Medical College, Cornell University, New York, NY 10065
| | - Xiaofeng Xia
- Chao Center for BRAIN, Department of Systems Medicine and Bioengineering, Houston Methodist Research Institute, 6670 Bertner Avenue, Houston, TX 77030.,Weill Cornell Medical College, Cornell University, New York, NY 10065
| | - Stephen T C Wong
- Chao Center for BRAIN, Department of Systems Medicine and Bioengineering, Houston Methodist Research Institute, 6670 Bertner Avenue, Houston, TX 77030.,Weill Cornell Medical College, Cornell University, New York, NY 10065
| |
Collapse
|
25
|
Marcuzzi A, Piscianz E, Valencic E, Monasta L, Vecchi Brumatti L, Tommasini A. To Extinguish the Fire from Outside the Cell or to Shutdown the Gas Valve Inside? Novel Trends in Anti-Inflammatory Therapies. Int J Mol Sci 2015; 16:21277-93. [PMID: 26370962 PMCID: PMC4613252 DOI: 10.3390/ijms160921277] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2015] [Revised: 08/19/2015] [Accepted: 08/31/2015] [Indexed: 12/26/2022] Open
Abstract
Cytokines are the most important soluble mediators of inflammation. Rare pediatric diseases provided exemplar conditions to study the anti-inflammatory efficacy of new generation therapies (biologics/biopharmaceuticals) selectively targeting single cytokines. Monoclonal antibodies and recombinant proteins have revolutionized anti-inflammatory therapies in the last two decades, allowing the specific targeting of single cytokines. They are very effective in extinguishing inflammation from outside the cell, even with the risk of an excessive and prolonged immunosuppression. Small molecules can enter the cell and shutdown the valve of inflammation by directly targeting signal proteins involved in cytokine release or in response to cytokines. They are orally-administrable drugs whose dosage can be easily adjusted to obtain the desired anti-inflammatory effect. This could make these drugs more suitable for a wide range of diseases as stroke, gout, or neurological impairment, where inflammatory activation plays a pivotal role as trigger. Autoinflammatory diseases, which have previously put anti-cytokine proteins in the limelight, can again provide a valuable model to measure the real potential of small inhibitors as anti-inflammatory agents.
Collapse
Affiliation(s)
- Annalisa Marcuzzi
- Department of Medicine, Surgery and Health Sciences, University of Trieste, Piazzale Europa 1, Trieste 34128, Italy.
| | - Elisa Piscianz
- Institute for Maternal and Child Health - IRCCS "Burlo Garofolo" - , via dell'Istria, 65/1, Trieste 34137, Italy.
| | - Erica Valencic
- Institute for Maternal and Child Health - IRCCS "Burlo Garofolo" - , via dell'Istria, 65/1, Trieste 34137, Italy.
| | - Lorenzo Monasta
- Institute for Maternal and Child Health - IRCCS "Burlo Garofolo" - , via dell'Istria, 65/1, Trieste 34137, Italy.
| | - Liza Vecchi Brumatti
- Institute for Maternal and Child Health - IRCCS "Burlo Garofolo" - , via dell'Istria, 65/1, Trieste 34137, Italy.
| | - Alberto Tommasini
- Institute for Maternal and Child Health - IRCCS "Burlo Garofolo" - , via dell'Istria, 65/1, Trieste 34137, Italy.
| |
Collapse
|
26
|
Janecek E, Wilk E, Schughart K, Geffers R, Strube C. Microarray gene expression analysis reveals major differences between Toxocara canis and Toxocara cati neurotoxocarosis and involvement of T. canis in lipid biosynthetic processes. Int J Parasitol 2015; 45:495-503. [PMID: 25843806 DOI: 10.1016/j.ijpara.2015.02.009] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2015] [Revised: 02/18/2015] [Accepted: 02/20/2015] [Indexed: 12/27/2022]
Abstract
Toxocara canis and Toxocara cati are globally occurring intestinal nematodes of dogs and cats with a high zoonotic potential. Migrating larvae in the CNS of paratenic hosts, including humans, may cause neurotoxocarosis resulting in a variety of neurological symptoms. Toxocara canis exhibits a stronger affinity to the CNS than T. cati, causing more severe neurological symptoms in the mouse model. Pathomechanisms of neurotoxocarosis as well as host responses towards the respective parasite are mostly unknown. Therefore, the aim of this study was to characterise the pathogenesis at a transcriptional level using whole genome microarray expression analysis and identify differences and similarities between T. canis- and T. cati-infected brains. Microarray analysis was conducted in cerebra and cerebella of infected C57Bl/6J mice 42daysp.i. revealing more differentially transcribed genes for T. canis- than T. cati-infected brains. In cerebra and cerebella of T. canis-infected mice, a total of 2304 and 1954 differentially transcribed genes, respectively, were identified whereas 113 and 760 differentially transcribed genes were determined in cerebra and cerebella of T. cati-infected mice. Functional annotation analysis revealed major differences in host responses in terms of significantly enriched biological modules. Up-regulated genes were mainly associated with the terms "immune and defence response", "sensory perception" as well as "behaviour/taxis" retrieved from the Gene Ontology database. These observations indicate a strong immune response in both infection groups with T. cati-infected brains revealing less severe reactions. Down-regulated genes in T. canis-infected cerebra and cerebella revealed a significant enrichment for the Gene Ontology term "lipid/cholesterol biosynthetic process". Cholesterol is a highly abundant and important component in the brain, representing several functions. Disturbances of synthesis as well as concentration changes may lead to dysfunction in signal transduction and neurodegenerative disease. Overall, only a minor overlap of differentially transcribed genes was observed between the two infection groups in both brain parts. Most genes are regulated individually in each infection group, supporting the evident differences of both roundworm species observed in the paratenic host in previous studies. In summary the present study underlines the differences in pathogenicity of T. canis and T. cati. It furthermore provides a comprehensive basis for future analyses over the course of infection as well as functional tests to identify gene regulatory circuits that are crucial for pathogenesis of neurotoxocarosis. The results of this study provide a promising foundation for further specific research to evaluate the particular pathogenetic mechanisms and to identify possible therapeutic targets.
Collapse
Affiliation(s)
- Elisabeth Janecek
- Institute for Parasitology, University of Veterinary Medicine Hannover, Germany
| | - Esther Wilk
- Department Infection Genetics, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Klaus Schughart
- Department Infection Genetics, Helmholtz Centre for Infection Research, Braunschweig, Germany; University of Veterinary Medicine Hannover, University of Tennessee Health Science Center, USA
| | - Robert Geffers
- Research Group Genome Analytics, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Christina Strube
- Institute for Parasitology, University of Veterinary Medicine Hannover, Germany.
| |
Collapse
|
27
|
Nagashima S, Yagyu H, Tozawa R, Tazoe F, Takahashi M, Kitamine T, Yamamuro D, Sakai K, Sekiya M, Okazaki H, Osuga JI, Honda A, Ishibashi S. Plasma cholesterol-lowering and transient liver dysfunction in mice lacking squalene synthase in the liver. J Lipid Res 2015; 56:998-1005. [PMID: 25755092 DOI: 10.1194/jlr.m057406] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2015] [Indexed: 01/23/2023] Open
Abstract
Squalene synthase (SS) catalyzes the biosynthesis of squalene, the first specific intermediate in the cholesterol biosynthetic pathway. To test the feasibility of lowering plasma cholesterol by inhibiting hepatic SS, we generated mice in which SS is specifically knocked out in the liver (L-SSKO) using Cre-loxP technology. Hepatic SS activity of L-SSKO mice was reduced by >90%. In addition, cholesterol biosynthesis in the liver slices was almost eliminated. Although the hepatic squalene contents were markedly reduced in L-SSKO mice, the hepatic contents of cholesterol and its precursors distal to squalene were indistinguishable from those of control mice, indicating the presence of sufficient centripetal flow of cholesterol and/or its precursors from the extrahepatic tissues. L-SSKO mice showed a transient liver dysfunction with moderate hepatomegaly presumably secondary to increased farnesol production. In a fed state, the plasma total cholesterol and triglyceride were significantly reduced in L-SSKO mice, primarily owing to reduced hepatic VLDL secretion. In a fasted state, the hypolipidemic effect was lost. mRNA expression of liver X receptor α target genes was reduced, while that of sterol-regulatory element binding protein 2 target genes was increased. In conclusion, liver-specific ablation of SS inhibits hepatic cholesterol biosynthesis and induces hypolipidemia without increasing significant mortality.
Collapse
Affiliation(s)
- Shuichi Nagashima
- Division of Endocrinology and Metabolism, School of Medicine, Jichi Medical University, Shimotsuke, Tochigi 329-0498, Japan
| | - Hiroaki Yagyu
- Division of Endocrinology and Metabolism, School of Medicine, Jichi Medical University, Shimotsuke, Tochigi 329-0498, Japan
| | - Ryuichi Tozawa
- Department of Metabolic Diseases, Graduate School of Medicine, University of Tokyo, Tokyo 113-8655, Japan
| | - Fumiko Tazoe
- Division of Endocrinology and Metabolism, School of Medicine, Jichi Medical University, Shimotsuke, Tochigi 329-0498, Japan
| | - Manabu Takahashi
- Division of Endocrinology and Metabolism, School of Medicine, Jichi Medical University, Shimotsuke, Tochigi 329-0498, Japan
| | - Tetsuya Kitamine
- Department of Metabolic Diseases, Graduate School of Medicine, University of Tokyo, Tokyo 113-8655, Japan
| | - Daisuke Yamamuro
- Division of Endocrinology and Metabolism, School of Medicine, Jichi Medical University, Shimotsuke, Tochigi 329-0498, Japan
| | - Kent Sakai
- Division of Endocrinology and Metabolism, School of Medicine, Jichi Medical University, Shimotsuke, Tochigi 329-0498, Japan
| | - Motohiro Sekiya
- Department of Metabolic Diseases, Graduate School of Medicine, University of Tokyo, Tokyo 113-8655, Japan
| | - Hiroaki Okazaki
- Department of Metabolic Diseases, Graduate School of Medicine, University of Tokyo, Tokyo 113-8655, Japan
| | - Jun-ichi Osuga
- Division of Endocrinology and Metabolism, School of Medicine, Jichi Medical University, Shimotsuke, Tochigi 329-0498, Japan
| | - Akira Honda
- Joint Research Center, Tokyo Medical University Ibaraki Medical Center, Ibaraki 300-0395, Japan
| | - Shun Ishibashi
- Division of Endocrinology and Metabolism, School of Medicine, Jichi Medical University, Shimotsuke, Tochigi 329-0498, Japan
| |
Collapse
|
28
|
Hidden disease susceptibility and sexual dimorphism in the heterozygous knockout of Cyp51 from cholesterol synthesis. PLoS One 2014; 9:e112787. [PMID: 25393872 PMCID: PMC4231084 DOI: 10.1371/journal.pone.0112787] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2014] [Accepted: 10/15/2014] [Indexed: 01/16/2023] Open
Abstract
We examined the genotype-phenotype interactions of Cyp51+/- mice carrying one functional allele of lanosterol 14α-demethylase from cholesterol biosynthesis. No distinct developmental or morphological abnormalities were observed by routine visual inspection of Cyp51+/- and Cyp51+/+ mice and fertility was similar. We further collected a large data-set from female and male Cyp51+/- mice and controls fed for 16 weeks with three diets and applied linear regression modeling. We used 3 predictor variables (genotype, sex, diet), and 39 response variables corresponding to the organ characteristics (7), plasma parameters (7), and hepatic gene expression (25). We observed significant differences between Cyp51+/- and wild-type mice in organ characteristics and blood lipid profile. Hepatomegaly was observed in Cyp51+/- males, together with elevated total and low-density lipoprotein cholesterol. Cyp51+/- females fed high-fat, high-cholesterol diet were leaner and had elevated plasma corticosterone compared to controls. We observed elevated hepatocyte apoptosis, mitosis and lipid infiltration in heterozygous knockouts of both sexes. The Cyp51+/- females had a modified lipid storage homeostasis protecting them from weight-gain when fed high-fat high-cholesterol diet. Malfunction of one Cyp51 allele therefore initiates disease pathways towards cholesterol-linked liver pathologies and sex-dependent response to dietary challenge.
Collapse
|
29
|
Lipids in the nervous system: from biochemistry and molecular biology to patho-physiology. Biochim Biophys Acta Mol Cell Biol Lipids 2014; 1851:51-60. [PMID: 25150974 DOI: 10.1016/j.bbalip.2014.08.011] [Citation(s) in RCA: 73] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2014] [Revised: 08/08/2014] [Accepted: 08/12/2014] [Indexed: 12/16/2022]
Abstract
Lipids in the nervous system accomplish a great number of key functions, from synaptogenesis to impulse conduction, and more. Most of the lipids of the nervous system are localized in myelin sheaths. It has long been known that myelin structure and brain homeostasis rely on specific lipid-protein interactions and on specific cell-to-cell signaling. In more recent years, the growing advances in large-scale technologies and genetically modified animal models have provided valuable insights into the role of lipids in the nervous system. Key findings recently emerged in these areas are here summarized. In addition, we briefly discuss how this new knowledge can open novel approaches for the treatment of diseases associated with alteration of lipid metabolism/homeostasis in the nervous system. This article is part of a Special Issue entitled Linking transcription to physiology in lipidomics.
Collapse
|
30
|
Eisa-Beygi S, Ekker M, Moon TW, Macdonald RL, Wen XY. Developmental processes regulated by the 3-hydroxy-3-methylglutaryl-CoA reductase (HMGCR) pathway: highlights from animal studies. Reprod Toxicol 2014; 46:115-20. [PMID: 24732207 DOI: 10.1016/j.reprotox.2014.04.001] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2013] [Revised: 03/13/2014] [Accepted: 04/02/2014] [Indexed: 12/20/2022]
Abstract
The 3-hydroxy-3-methylglutaryl coenzyme A reductase (HMGCR) is the rate-limiting enzyme in the biosynthesis of cholesterol and isoprenoids, which are substrates required for post-translational modification of signalling proteins that can potentially regulate various aspects of embryonic development. The HMGCR transcripts are detectable during early embryogenesis in both invertebrates and vertebrates, which suggests a conserved developmental requirement for mevalonate derivatives. Consistently, recent animal and in vitro studies have yielded valuable insights into potential morphogenic parameters that are modulated by HMGCR activity. These developmental end-points include brain and craniofacial morphogenesis, PGC migration and survival, myocardial epithelial migration and fusion, EC migration and survival, and vascular stabilization. By providing a synthesis of these studies, we hope that this review will highlight the need to comprehensively examine the entire suite of developmental processes regulated by HMGCR.
Collapse
Affiliation(s)
- Shahram Eisa-Beygi
- Department of Biology, Centre for Advanced Research in Environmental Genomics (CAREG), University of Ottawa, ON, Canada; Zebrafish Centre for Advanced Drug Discovery, Keenan Research Centre of the Li Ka Shing Knowledge Institute of St. Michael's Hospital, Toronto, ON, Canada; Institute of Medical Science & Department of Medicine, University of Toronto, ON, Canada.
| | - Marc Ekker
- Department of Biology, Centre for Advanced Research in Environmental Genomics (CAREG), University of Ottawa, ON, Canada
| | - Thomas W Moon
- Department of Biology, Centre for Advanced Research in Environmental Genomics (CAREG), University of Ottawa, ON, Canada
| | - R Loch Macdonald
- Zebrafish Centre for Advanced Drug Discovery, Keenan Research Centre of the Li Ka Shing Knowledge Institute of St. Michael's Hospital, Toronto, ON, Canada; Institute of Medical Science & Department of Medicine, University of Toronto, ON, Canada; Division of Neurosurgery, St. Michael's Hospital, Labatt Family Centre of Excellence in Brain Injury and Trauma Research, Keenan Research Centre of the Li Ka Shing Knowledge Institute of St. Michael's Hospital, Toronto, ON, Canada
| | - Xiao-Yan Wen
- Zebrafish Centre for Advanced Drug Discovery, Keenan Research Centre of the Li Ka Shing Knowledge Institute of St. Michael's Hospital, Toronto, ON, Canada; Institute of Medical Science & Department of Medicine, University of Toronto, ON, Canada
| |
Collapse
|
31
|
Mevalonate kinase deficiency and neuroinflammation: balance between apoptosis and pyroptosis. Int J Mol Sci 2013; 14:23274-88. [PMID: 24287904 PMCID: PMC3876043 DOI: 10.3390/ijms141223274] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2013] [Revised: 11/08/2013] [Accepted: 11/13/2013] [Indexed: 12/20/2022] Open
Abstract
Mevalonic aciduria, a rare autosomal recessive disease, represents the most severe form of the periodic fever, known as Mevalonate Kinase Deficiency. This disease is caused by the mutation of the MVK gene, which codes for the enzyme mevalonate kinase, along the cholesterol pathway. Mevalonic aciduria patients show recurrent fever episodes with associated inflammatory symptoms, severe neurologic impairments, or death, in early childhood. The typical neurodegeneration occurring in mevalonic aciduria is linked both to the intrinsic apoptosis pathway (caspase-3 and -9), which is triggered by mitochondrial damage, and to pyroptosis (caspase-1). These cell death mechanisms seem to be also related to the assembly of the inflammasome, which may, in turn, activate pro-inflammatory cytokines and chemokines. Thus, this particular molecular platform may play a crucial role in neuroinflammation mechanisms. Nowadays, a specific therapy is still lacking and the pathogenic mechanisms involving neuroinflammation and neuronal dysfunction have not yet been completely understood, making mevalonic aciduria an orphan drug disease. This review aims to analyze the relationship among neuroinflammation, mitochondrial damage, programmed cell death, and neurodegeneration. Targeting inflammation and degeneration in the central nervous system might help identify promising treatment approaches for mevalonic aciduria or other diseases in which these mechanisms are involved.
Collapse
|
32
|
Grover A, Samuel G, Bisaria VS, Sundar D. Enhanced withanolide production by overexpression of squalene synthase in Withania somnifera. J Biosci Bioeng 2013; 115:680-5. [DOI: 10.1016/j.jbiosc.2012.12.011] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2012] [Revised: 12/06/2012] [Accepted: 12/12/2012] [Indexed: 10/27/2022]
|
33
|
Robinson JF, Verhoef A, van Beelen VA, Pennings JL, Piersma AH. Dose–response analysis of phthalate effects on gene expression in rat whole embryo culture. Toxicol Appl Pharmacol 2012; 264:32-41. [DOI: 10.1016/j.taap.2012.07.015] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2012] [Revised: 07/06/2012] [Accepted: 07/07/2012] [Indexed: 01/05/2023]
|
34
|
Abstract
Cholesterol is an essential membrane component enriched in plasma membranes, growth cones, and synapses. The brain normally synthesizes all cholesterol locally, but the contribution of individual cell types to brain cholesterol metabolism is unknown. To investigate whether cortical projection neurons in vivo essentially require cholesterol biosynthesis and which cell types support neurons, we have conditionally ablated the cholesterol biosynthesis in these neurons in mice either embryonically or postnatally. We found that cortical projection neurons synthesize cholesterol during their entire lifetime. At all stages, they can also benefit from glial support. Adult neurons that lack cholesterol biosynthesis are mainly supported by astrocytes such that their functional integrity is preserved. In contrast, microglial cells support young neurons. However, compensatory efforts of microglia are only transient leading to layer-specific neuronal death and the reduction of cortical projections. Hence, during the phase of maximal membrane growth and maximal cholesterol demand, neuronal cholesterol biosynthesis is indispensable. Analysis of primary neurons revealed that neurons tolerate only slight alteration in the cholesterol content and plasma membrane tension. This quality control allows neurons to differentiate normally and adjusts the extent of neurite outgrowth, the number of functional growth cones and synapses to the available cholesterol. This study highlights both the flexibility and the limits of horizontal cholesterol transfer in vivo and may have implications for the understanding of neurodegenerative diseases.
Collapse
|
35
|
Nagashima S, Yagyu H, Ohashi K, Tazoe F, Takahashi M, Ohshiro T, Bayasgalan T, Okada K, Sekiya M, Osuga JI, Ishibashi S. Liver-Specific Deletion of 3-Hydroxy-3-Methylglutaryl Coenzyme A Reductase Causes Hepatic Steatosis and Death. Arterioscler Thromb Vasc Biol 2012; 32:1824-31. [DOI: 10.1161/atvbaha.111.240754] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Affiliation(s)
- Shuichi Nagashima
- From the Division of Endocrinology and Metabolism, Department of Medicine, Jichi Medical University, Shimotsuke, Tochigi, Japan (S.N., H.Y., F.T., M.T., T.O., T.B., K. Okada, J.O., S.I.); and Department of Metabolic Diseases, Graduate School of Medicine, University of Tokyo, Tokyo, Japan (K. Ohashi, M.S.)
| | - Hiroaki Yagyu
- From the Division of Endocrinology and Metabolism, Department of Medicine, Jichi Medical University, Shimotsuke, Tochigi, Japan (S.N., H.Y., F.T., M.T., T.O., T.B., K. Okada, J.O., S.I.); and Department of Metabolic Diseases, Graduate School of Medicine, University of Tokyo, Tokyo, Japan (K. Ohashi, M.S.)
| | - Ken Ohashi
- From the Division of Endocrinology and Metabolism, Department of Medicine, Jichi Medical University, Shimotsuke, Tochigi, Japan (S.N., H.Y., F.T., M.T., T.O., T.B., K. Okada, J.O., S.I.); and Department of Metabolic Diseases, Graduate School of Medicine, University of Tokyo, Tokyo, Japan (K. Ohashi, M.S.)
| | - Fumiko Tazoe
- From the Division of Endocrinology and Metabolism, Department of Medicine, Jichi Medical University, Shimotsuke, Tochigi, Japan (S.N., H.Y., F.T., M.T., T.O., T.B., K. Okada, J.O., S.I.); and Department of Metabolic Diseases, Graduate School of Medicine, University of Tokyo, Tokyo, Japan (K. Ohashi, M.S.)
| | - Manabu Takahashi
- From the Division of Endocrinology and Metabolism, Department of Medicine, Jichi Medical University, Shimotsuke, Tochigi, Japan (S.N., H.Y., F.T., M.T., T.O., T.B., K. Okada, J.O., S.I.); and Department of Metabolic Diseases, Graduate School of Medicine, University of Tokyo, Tokyo, Japan (K. Ohashi, M.S.)
| | - Taichi Ohshiro
- From the Division of Endocrinology and Metabolism, Department of Medicine, Jichi Medical University, Shimotsuke, Tochigi, Japan (S.N., H.Y., F.T., M.T., T.O., T.B., K. Okada, J.O., S.I.); and Department of Metabolic Diseases, Graduate School of Medicine, University of Tokyo, Tokyo, Japan (K. Ohashi, M.S.)
| | - Tumenbayar Bayasgalan
- From the Division of Endocrinology and Metabolism, Department of Medicine, Jichi Medical University, Shimotsuke, Tochigi, Japan (S.N., H.Y., F.T., M.T., T.O., T.B., K. Okada, J.O., S.I.); and Department of Metabolic Diseases, Graduate School of Medicine, University of Tokyo, Tokyo, Japan (K. Ohashi, M.S.)
| | - Kenta Okada
- From the Division of Endocrinology and Metabolism, Department of Medicine, Jichi Medical University, Shimotsuke, Tochigi, Japan (S.N., H.Y., F.T., M.T., T.O., T.B., K. Okada, J.O., S.I.); and Department of Metabolic Diseases, Graduate School of Medicine, University of Tokyo, Tokyo, Japan (K. Ohashi, M.S.)
| | - Motohiro Sekiya
- From the Division of Endocrinology and Metabolism, Department of Medicine, Jichi Medical University, Shimotsuke, Tochigi, Japan (S.N., H.Y., F.T., M.T., T.O., T.B., K. Okada, J.O., S.I.); and Department of Metabolic Diseases, Graduate School of Medicine, University of Tokyo, Tokyo, Japan (K. Ohashi, M.S.)
| | - Jun-ichi Osuga
- From the Division of Endocrinology and Metabolism, Department of Medicine, Jichi Medical University, Shimotsuke, Tochigi, Japan (S.N., H.Y., F.T., M.T., T.O., T.B., K. Okada, J.O., S.I.); and Department of Metabolic Diseases, Graduate School of Medicine, University of Tokyo, Tokyo, Japan (K. Ohashi, M.S.)
| | - Shun Ishibashi
- From the Division of Endocrinology and Metabolism, Department of Medicine, Jichi Medical University, Shimotsuke, Tochigi, Japan (S.N., H.Y., F.T., M.T., T.O., T.B., K. Okada, J.O., S.I.); and Department of Metabolic Diseases, Graduate School of Medicine, University of Tokyo, Tokyo, Japan (K. Ohashi, M.S.)
| |
Collapse
|
36
|
Pennings JLA, Theunissen PT, Piersma AH. An optimized gene set for transcriptomics based neurodevelopmental toxicity prediction in the neural embryonic stem cell test. Toxicology 2012; 300:158-67. [PMID: 22760166 DOI: 10.1016/j.tox.2012.06.016] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2012] [Revised: 06/22/2012] [Accepted: 06/23/2012] [Indexed: 11/16/2022]
Abstract
The murine neural embryonic stem cell test (ESTn) is an in vitro model for neurodevelopmental toxicity testing. Recent studies have shown that application of transcriptomics analyses in the ESTn is useful for obtaining more accurate predictions as well as mechanistic insights. Gene expression responses due to stem cell neural differentiation versus toxicant exposure could be distinguished using the Principal Component Analysis based differentiation track algorithm. In this study, we performed a de novo analysis on combined raw data (10 compounds, 19 exposures) from three previous transcriptomics studies to identify an optimized gene set for neurodevelopmental toxicity prediction in the ESTn. By evaluating predictions of 200,000 randomly selected gene sets, we identified genes which significantly contributed to the prediction reliability. A set of 100 genes was obtained, predominantly involved in (neural) development. Further stringency restrictions resulted in a set of 29 genes that allowed for 84% prediction accuracy (area under the curve 94%). We anticipate these gene sets will contribute to further improve ESTn transcriptomics studies aimed at compound risk assessment.
Collapse
Affiliation(s)
- Jeroen L A Pennings
- Laboratory for Health Protection Research (GBO), National Institute for Public Health and the Environment, (RIVM), Bilthoven, The Netherlands.
| | | | | |
Collapse
|
37
|
Saloniemi T, Jokela H, Strauss L, Pakarinen P, Poutanen M. The diversity of sex steroid action: novel functions of hydroxysteroid (17β) dehydrogenases as revealed by genetically modified mouse models. J Endocrinol 2012; 212:27-40. [PMID: 22045753 DOI: 10.1530/joe-11-0315] [Citation(s) in RCA: 63] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Disturbed action of sex steroid hormones, i.e. androgens and estrogens, is involved in the pathogenesis of various severe diseases in humans. Interestingly, recent studies have provided data further supporting the hypothesis that the circulating hormone concentrations do not explain all physiological and pathological processes observed in hormone-dependent tissues, while the intratissue sex steroid concentrations are determined by the expression of steroid metabolising enzymes in the neighbouring cells (paracrine action) and/or by target cells themselves (intracrine action). This local sex steroid production is also a valuable treatment option for developing novel therapies against hormonal diseases. Hydroxysteroid (17β) dehydrogenases (HSD17Bs) compose a family of 14 enzymes that catalyse the conversion between the low-active 17-keto steroids and the highly active 17β-hydroxy steroids. The enzymes frequently expressed in sex steroid target tissues are, thus, potential drug targets in order to lower the local sex steroid concentrations. The present review summarises the recent data obtained for the role of HSD17B1, HSD17B2, HSD17B7 and HSD17B12 enzymes in various metabolic pathways and their physiological and pathophysiological roles as revealed by the recently generated genetically modified mouse models. Our data, together with that provided by others, show that, in addition to having a role in sex steroid metabolism, several of these HSD17B enzymes possess key roles in other metabolic processes: for example, HD17B7 is essential for cholesterol biosynthesis and HSD17B12 is involved in elongation of fatty acids. Additional studies in vitro and in vivo are to be carried out in order to fully define the metabolic role of the HSD17B enzymes and to evaluate their value as drug targets.
Collapse
Affiliation(s)
- Taija Saloniemi
- Department of Physiology, Institute of Biomedicine, University of Turku, Kiinamyllynkatu 10, FI-20014 Turku, Finland
| | | | | | | | | |
Collapse
|
38
|
Valenza M, Cattaneo E. Emerging roles for cholesterol in Huntington's disease. Trends Neurosci 2011; 34:474-86. [DOI: 10.1016/j.tins.2011.06.005] [Citation(s) in RCA: 83] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2010] [Revised: 06/04/2011] [Accepted: 06/08/2011] [Indexed: 01/01/2023]
|
39
|
Keber R, Motaln H, Wagner KD, Debeljak N, Rassoulzadegan M, Ačimovič J, Rozman D, Horvat S. Mouse knockout of the cholesterogenic cytochrome P450 lanosterol 14alpha-demethylase (Cyp51) resembles Antley-Bixler syndrome. J Biol Chem 2011; 286:29086-29097. [PMID: 21705796 DOI: 10.1074/jbc.m111.253245] [Citation(s) in RCA: 72] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Antley-Bixler syndrome (ABS) represents a group of heterogeneous disorders characterized by skeletal, cardiac, and urogenital abnormalities that have frequently been associated with mutations in fibroblast growth factor receptor 2 or cytochrome P450 reductase genes. In some ABS patients, reduced activity of the cholesterogenic cytochrome P450 CYP51A1, an ortholog of the mouse CYP51, and accumulation of lanosterol and 24,25-dihydrolanosterol has been reported, but the role of CYP51A1 in the ABS etiology has remained obscure. To test whether Cyp51 could be involved in generating an ABS-like phenotype, a mouse knock-out model was developed that exhibited several prenatal ABS-like features leading to lethality at embryonic day 15. Cyp51(-/-) mice had no functional Cyp51 mRNA and no immunodetectable CYP51 protein. The two CYP51 enzyme substrates (lanosterol and 24,25-dihydrolanosterol) were markedly accumulated. Cholesterol precursors downstream of the CYP51 enzymatic step were not detected, indicating that the targeting in this study blocked de novo cholesterol synthesis. This was reflected in the up-regulation of 10 cholesterol synthesis genes, with the exception of 7-dehydrocholesterol reductase. Lethality was ascribed to heart failure due to hypoplasia, ventricle septum, and epicardial and vasculogenesis defects, suggesting that Cyp51 deficiency was involved in heart development and coronary vessel formation. As the most likely downstream molecular mechanisms, alterations were identified in the sonic hedgehog and retinoic acid signaling pathways. Cyp51 knock-out mice provide evidence that Cyp51 is essential for embryogenesis and present a potential animal model for studying ABS syndrome in humans.
Collapse
Affiliation(s)
- Rok Keber
- Department of Animal Science, Biotechnical Faculty, University of Ljubljana, 1000 Ljubljana, Slovenia
| | - Helena Motaln
- Department of Animal Science, Biotechnical Faculty, University of Ljubljana, 1000 Ljubljana, Slovenia; Department of Genetic Toxicology and Cancer Biology, National Institute of Biology, 1000 Ljubljana, Slovenia
| | - Kay D Wagner
- INSERM U907, Parc Valrose, Nice, France; Université de Nice, Sophia-Antipolis, Parc Valrose, Nice, France
| | - Nataša Debeljak
- Institute of Biochemistry, Centre for Functional Genomics and Bio-Chips, Faculty of Medicine, University of Ljubljana, 1000 Ljubljana, Slovenia
| | - Minoo Rassoulzadegan
- Université de Nice, Sophia-Antipolis, Parc Valrose, Nice, France; Centre de Biochimie, INSERM U636, Parc Valrose, Nice, France
| | - Jure Ačimovič
- Institute of Biochemistry, Centre for Functional Genomics and Bio-Chips, Faculty of Medicine, University of Ljubljana, 1000 Ljubljana, Slovenia
| | - Damjana Rozman
- Institute of Biochemistry, Centre for Functional Genomics and Bio-Chips, Faculty of Medicine, University of Ljubljana, 1000 Ljubljana, Slovenia
| | - Simon Horvat
- Department of Animal Science, Biotechnical Faculty, University of Ljubljana, 1000 Ljubljana, Slovenia; National Institute of Chemistry, 1000 Ljubljana, Slovenia and.
| |
Collapse
|
40
|
Saher G, Quintes S, Nave KA. Cholesterol: a novel regulatory role in myelin formation. Neuroscientist 2011; 17:79-93. [PMID: 21343408 DOI: 10.1177/1073858410373835] [Citation(s) in RCA: 81] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Myelin consists of tightly compacted membranes that form an insulating sheath around axons. The function of myelin for rapid saltatory nerve conduction is dependent on its unique composition, highly enriched in glycosphingolipids and cholesterol. Cholesterol emerged as the only integral myelin component that is essential and rate limiting for the development of CNS and PNS myelin. Experiments with conditional mouse mutants that lack cholesterol biosynthesis in oligodendrocytes revealed that only minimal changes of the CNS myelin lipid composition are tolerated. In Schwann cells of the PNS, protein trafficking and myelin compaction depend on cholesterol. In this review, the authors summarize the role of cholesterol in myelin biogenesis and myelin disease.
Collapse
Affiliation(s)
- Gesine Saher
- Max Planck Institute of Experimental Medicine, Neurogenetics, Göttingen, Germany.
| | | | | |
Collapse
|
41
|
Horvat S, Mcwhir J, Rozman D. Defects in cholesterol synthesis genes in mouse and in humans: lessons for drug development and safer treatments. Drug Metab Rev 2011; 43:69-90. [DOI: 10.3109/03602532.2010.540580] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
|
42
|
Kim TD, Han JY, Huh GH, Choi YE. Expression and Functional Characterization of Three Squalene Synthase Genes Associated with Saponin Biosynthesis in Panax ginseng. ACTA ACUST UNITED AC 2010; 52:125-37. [DOI: 10.1093/pcp/pcq179] [Citation(s) in RCA: 79] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
|
43
|
Sigurdsson MI, Jamshidi N, Steingrimsson E, Thiele I, Palsson BØ. A detailed genome-wide reconstruction of mouse metabolism based on human Recon 1. BMC SYSTEMS BIOLOGY 2010; 4:140. [PMID: 20959003 PMCID: PMC2978158 DOI: 10.1186/1752-0509-4-140] [Citation(s) in RCA: 115] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/17/2010] [Accepted: 10/19/2010] [Indexed: 12/16/2022]
Abstract
BACKGROUND Well-curated and validated network reconstructions are extremely valuable tools in systems biology. Detailed metabolic reconstructions of mammals have recently emerged, including human reconstructions. They raise the question if the various successful applications of microbial reconstructions can be replicated in complex organisms. RESULTS We mapped the published, detailed reconstruction of human metabolism (Recon 1) to other mammals. By searching for genes homologous to Recon 1 genes within mammalian genomes, we were able to create draft metabolic reconstructions of five mammals, including the mouse. Each draft reconstruction was created in compartmentalized and non-compartmentalized version via two different approaches. Using gap-filling algorithms, we were able to produce all cellular components with three out of four versions of the mouse metabolic reconstruction. We finalized a functional model by iterative testing until it passed a predefined set of 260 validation tests. The reconstruction is the largest, most comprehensive mouse reconstruction to-date, accounting for 1,415 genes coding for 2,212 gene-associated reactions and 1,514 non-gene-associated reactions.We tested the mouse model for phenotype prediction capabilities. The majority of predicted essential genes were also essential in vivo. However, our non-tissue specific model was unable to predict gene essentiality for many of the metabolic genes shown to be essential in vivo. Our knockout simulation of the lipoprotein lipase gene correlated well with experimental results, suggesting that softer phenotypes can also be simulated. CONCLUSIONS We have created a high-quality mouse genome-scale metabolic reconstruction, iMM1415 (Mus Musculus, 1415 genes). We demonstrate that the mouse model can be used to perform phenotype simulations, similar to models of microbe metabolism. Since the mouse is an important experimental organism, this model should become an essential tool for studying metabolic phenotypes in mice, including outcomes from drug screening.
Collapse
Affiliation(s)
- Martin I Sigurdsson
- Department of Biochemistry and Molecular Biology, Faculty of Medicine, University of Iceland, Reykjavik, Iceland
| | | | | | | | | |
Collapse
|
44
|
Robinson JF, van Beelen VA, Verhoef A, Renkens MFJ, Luijten M, van Herwijnen MHM, Westerman A, Pennings JLA, Piersma AH. Embryotoxicant-Specific Transcriptomic Responses in Rat Postimplantation Whole-Embryo Culture. Toxicol Sci 2010; 118:675-85. [DOI: 10.1093/toxsci/kfq292] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
|
45
|
Kappen C, Kruger C, MacGowan J, Salbaum JM. Maternal diet modulates the risk for neural tube defects in a mouse model of diabetic pregnancy. Reprod Toxicol 2010; 31:41-9. [PMID: 20868740 DOI: 10.1016/j.reprotox.2010.09.002] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2010] [Revised: 08/17/2010] [Accepted: 09/04/2010] [Indexed: 02/03/2023]
Abstract
Pregnancies complicated by maternal diabetes have long been known to carry a higher risk for congenital malformations, such as neural tube defects. Using the FVB inbred mouse strain and the Streptozotocin-induced diabetes model, we tested whether the incidence of neural tube defects in diabetic pregnancies can be modulated by maternal diet. In a comparison of two commercial mouse diets, which are considered nutritionally replete, we found that maternal consumption of the unfavorable diet was associated with a more than 3-fold higher rate of neural tube defects. Our results demonstrate that maternal diet can act as a modifier of the risk for abnormal development in high-risk pregnancies, and provide support for the possibility that neural tube defects in human diabetic pregnancies might be preventable by optimized maternal nutrition.
Collapse
Affiliation(s)
- Claudia Kappen
- Department of Developmental Biology, Pennington Biomedical Research Center, Louisiana State University System, 6400 Perkins Road, Baton Rouge, LA 70808, USA.
| | | | | | | |
Collapse
|
46
|
Jokela H, Rantakari P, Lamminen T, Strauss L, Ola R, Mutka AL, Gylling H, Miettinen T, Pakarinen P, Sainio K, Poutanen M. Hydroxysteroid (17beta) dehydrogenase 7 activity is essential for fetal de novo cholesterol synthesis and for neuroectodermal survival and cardiovascular differentiation in early mouse embryos. Endocrinology 2010; 151:1884-92. [PMID: 20185768 DOI: 10.1210/en.2009-0928] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Hydroxysteroid (17beta) dehydrogenase 7 (HSD17B7) has been shown to catalyze the conversion of both estrone to estradiol (17-ketosteroid reductase activity) and zymosterone to zymosterol (3-ketosteroid reductase activity involved in cholesterol biosynthesis) in vitro. To define the metabolic role of the enzyme in vivo, we generated knockout mice deficient in the enzyme activity (HSD17B7KO). The data showed that the lack of HSD17B7 results in a blockage in the de novo cholesterol biosynthesis in mouse embryos in vivo, and HSD17BKO embryos die at embryonic day (E) 10.5. Analysis of neural structures revealed a defect in the development of hemispheres of the front brain with an increased apoptosis in the neuronal tissues. Morphological defects in the cardiovascular system were also observed from E9.5 onward. Mesodermal, endodermal, and hematopoietic cells were all detected by the histological analysis of the visceral yolk sac, whereas no organized vessels were observed in the knockout yolk sac. Immunohistological staining for platelet endothelial cell adhesion molecule-1 indicated that the complexity of the vasculature also was reduced in the HSD17B7KO embryos, particularly in the head capillary plexus and branchial arches. At E8.5-9.5, the heart development and the looping of the heart appeared to be normal in the HSD17B7KO embryos. However, at E10.5 the heart was dilated, and the thickness of the cardiac muscle and pericardium in the HSD17B7KO embryos was markedly reduced, and immunohistochemical staining for GATA-4 revealed that HSD17B7KO embryos had a reduced number of myocardial cells. The septum of the atrium was also defected in the knockout mice.
Collapse
Affiliation(s)
- Heli Jokela
- Department of Physiology, Institute of Biomedicine, University of Turku, Kiinamyllynkatu 10, FIN-20520 Turku, Finland
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Mutka AL, Ikonen E. Genetics and molecular biology: brain cholesterol balance--not such a closed circuit after all. Curr Opin Lipidol 2010; 21:93-4. [PMID: 20101121 DOI: 10.1097/mol.0b013e32833538b7] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
48
|
Cunningham D, Spychala K, McLarren KW, Garza LA, Boerkoel CF, Herman GE. Developmental expression pattern of the cholesterogenic enzyme NSDHL and negative selection of NSDHL-deficient cells in the heterozygous Bpa(1H)/+ mouse. Mol Genet Metab 2009; 98:356-66. [PMID: 19631568 PMCID: PMC2783206 DOI: 10.1016/j.ymgme.2009.06.016] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/07/2009] [Revised: 06/30/2009] [Accepted: 06/30/2009] [Indexed: 12/22/2022]
Abstract
NSDHL (NAD(P)H sterol dehydrogenase-like), is a 3beta-hydroxysterol dehydrogenase thought to function in the demethylation of sterol precursors in one of the later steps of cholesterol biosynthesis. Mutations in the X-linked NSDHL gene cause CHILD syndrome in humans, and the male-lethal bare patches (Bpa) phenotype in mice. The relative level of NSDHL expression among different mouse tissues at several stages of embryogenesis and postnatal development was analyzed by immunohistochemistry. In wild type (WT) embryos, the highest levels of expression were seen in the liver, dorsal root ganglia, central nervous system, retina, adrenal gland and testis. Heterozygous Bpa(1H) females are mosaic for NSDHL expression due to normal random X-inactivation. NSDHL-deficient cells were detected in the developing cerebral cortex and retina of Bpa(1H) female embryos. In postnatal WT and Bpa(1H) animals, we compared the expression pattern of NSDHL in skin, an affected tissue; liver, a main site of cholesterol synthesis; and brain, a tissue dependent on endogenous synthesis of cholesterol due to lack of transport across the blood-brain barrier. Clonal populations of mutant cells were visible in the brain, skin and liver of Bpa(1H) pups. In the liver, the proportion of NSDHL negative cells dropped from approximately 50% at postnatal day 6 to approximately 20% at one year of age. In the brain, which showed the highest expression in cerebral cortical and hippocampal neurons, the proportion of NSDHL negative cells also dropped dramatically over the first year of life. Our results suggest that while NSDHL-deficient cells in the mosaic Bpa(1H) female are able to survive and differentiate during embryonic development, they are subject to negative selection over the life of the animal.
Collapse
Affiliation(s)
- David Cunningham
- The Research Institute at Nationwide Children's Hospital, Columbus, OH 43205, USA.
| | | | | | | | | | | |
Collapse
|
49
|
Büdefeld T, Jezek D, Rozman D, Majdic G. Initiation of Steroidogenesis Precedes Expression of Cholesterologenic Enzymes in the Fetal Mouse Testes. Anat Histol Embryol 2009; 38:461-6. [DOI: 10.1111/j.1439-0264.2009.00975.x] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
50
|
Ablation of cholesterol biosynthesis in neural stem cells increases their VEGF expression and angiogenesis but causes neuron apoptosis. Proc Natl Acad Sci U S A 2009; 106:8350-5. [PMID: 19416849 DOI: 10.1073/pnas.0903541106] [Citation(s) in RCA: 58] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Although sufficient cholesterol supply is known to be crucial for neurons in the developing mammalian brain, the cholesterol requirement of neural stem and progenitor cells in the embryonic central nervous system has not been addressed. Here we have conditionally ablated the activity of squalene synthase (SQS), a key enzyme for endogenous cholesterol production, in the neural stem and progenitor cells of the ventricular zone (VZ) of the embryonic mouse brain. Mutant embryos exhibited a reduced brain size due to the atrophy of the neuronal layers, and died at birth. Analyses of the E11.5-E15.5 dorsal telencephalon and diencephalon revealed that this atrophy was due to massive apoptosis of newborn neurons, implying that this progeny of the SQS-ablated neural stem and progenitor cells was dependent on endogenous cholesterol biosynthesis for survival. Interestingly, the neural stem and progenitor cells of the VZ, the primary target of SQS inactivation, did not undergo significant apoptosis. Instead, vascular endothelial growth factor (VEGF) expression in these cells was strongly upregulated via a hypoxia-inducible factor-1-independent pathway, and angiogenesis in the VZ was increased. Consistent with an increased supply of lipoproteins to these cells, the level of lipid droplets containing triacylglycerides with unsaturated fatty acyl chains was found to be elevated. Our study establishes a direct link between intracellular cholesterol levels, VEGF expression, and angiogenesis. Moreover, our data reveal a hitherto unknown compensatory process by which the neural stem and progenitor cells of the developing mammalian brain evade the detrimental consequences of impaired endogenous cholesterol biosynthesis.
Collapse
|