1
|
Zhu Z, McClintock TS, Bieberich E. Transcriptomics analysis reveals potential regulatory role of nSMase2 (Smpd3) in nervous system development and function of middle-aged mouse brains. GENES, BRAIN, AND BEHAVIOR 2024; 23:e12911. [PMID: 39171374 PMCID: PMC11339599 DOI: 10.1111/gbb.12911] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/29/2024] [Revised: 07/22/2024] [Accepted: 07/29/2024] [Indexed: 08/23/2024]
Abstract
Neutral sphingomyelinase-2 (nSMase2), gene name sphingomyelin phosphodiesterase-3 (Smpd3), is a key regulatory enzyme responsible for generating the sphingolipid ceramide. The function of nSMase2 in the brain is still controversial. To better understand the functional roles of nSMase2 in the aging mouse brain, we applied RNA-seq analysis, which identified a total of 1462 differentially abundant mRNAs between +/fro and fro/fro, of which 891 were increased and 571 were decreased in nSMase2-deficient mouse brains. The most strongly enriched GO and KEGG annotation terms among transcripts increased in fro/fro mice included synaptogenesis, synapse development, synaptic signaling, axon development, and axonogenesis. Among decreased transcripts, enriched annotations included ribosome assembly and mitochondrial protein complex functions. KEGG analysis of decreased transcripts also revealed overrepresentation of annotations for Alzheimer's disease (AD), Parkinson's disease (PD), and Huntington disease (HD). Ingenuity Pathway Analysis (IPA) tools predicted lower susceptibility to these neurodegenerative disorders, as well as predictions agreeing with stronger synaptic function, learning, and memory in fro/fro mice. The IPA tools identified signaling proteins, epigenetic regulators, and microRNAs as likely upstream regulators of the broader set of genes encoding the affected transcripts. It also revealed 16 gene networks, each linked to biological processes identified as overrepresented annotations among the affected transcripts by multiple analysis methods. Therefore, the analysis of these RNA-seq data indicates that nSMase2 impacts synaptic function and neural development, and may contribute to the onset and development of neurodegenerative diseases in middle-aged mice.
Collapse
Affiliation(s)
- Zhihui Zhu
- Department of PhysiologyUniversity of Kentucky College of MedicineLexingtonKentuckyUSA
| | - Timothy S. McClintock
- Department of PhysiologyUniversity of Kentucky College of MedicineLexingtonKentuckyUSA
| | - Erhard Bieberich
- Department of PhysiologyUniversity of Kentucky College of MedicineLexingtonKentuckyUSA
- Veterans Affairs Medical CenterLexingtonKentuckyUSA
| |
Collapse
|
2
|
Wang L, Qu F, Yu X, Yang S, Zhao B, Chen Y, Li P, Zhang Z, Zhang J, Han X, Wei D. Cortical lipid metabolic pathway alteration of early Alzheimer's disease and candidate drugs screen. Eur J Med Res 2024; 29:199. [PMID: 38528586 DOI: 10.1186/s40001-024-01730-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2022] [Accepted: 02/12/2024] [Indexed: 03/27/2024] Open
Abstract
BACKGROUND Lipid metabolism changes occur in early Alzheimer's disease (AD) patients. Yet little is known about metabolic gene changes in early AD cortex. METHODS The lipid metabolic genes selected from two datasets (GSE39420 and GSE118553) were analyzed with enrichment analysis. Protein-protein interaction network construction and correlation analyses were used to screen core genes. Literature analysis and molecular docking were applied to explore potential therapeutic drugs. RESULTS 60 lipid metabolic genes differentially expressed in early AD patients' cortex were screened. Bioinformatics analyses revealed that up-regulated genes were mainly focused on mitochondrial fatty acid oxidation and mediating the activation of long-chain fatty acids, phosphoproteins, and cholesterol metabolism. Down-regulated genes were mainly focused on lipid transport, carboxylic acid metabolic process, and neuron apoptotic process. Literature reviews and molecular docking results indicated that ACSL1, ACSBG2, ACAA2, FABP3, ALDH5A1, and FFAR4 were core targets for lipid metabolism disorder and had a high binding affinity with compounds including adenosine phosphate, oxidized Photinus luciferin, BMS-488043, and candidate therapeutic drugs especially bisphenol A, benzo(a)pyrene, ethinyl estradiol. CONCLUSIONS AD cortical lipid metabolism disorder was associated with the dysregulation of the PPAR signaling pathway, glycerophospholipid metabolism, adipocytokine signaling pathway, fatty acid biosynthesis, fatty acid degradation, ferroptosis, biosynthesis of unsaturated fatty acids, and fatty acid elongation. Candidate drugs including bisphenol A, benzo(a)pyrene, ethinyl estradiol, and active compounds including adenosine phosphate, oxidized Photinus luciferin, and BMS-488043 have potential therapeutic effects on cortical lipid metabolism disorder of early AD.
Collapse
Affiliation(s)
- Linshuang Wang
- Institute of Basic Research in Clinical Medicine, China Academy of Chinese Medical Sciences, Beijing, 100700, China
| | - Fengxue Qu
- Beijing Anzhen Hospital, Capital Medical University, Beijing, 100029, China
| | - Xueyun Yu
- Institute of Basic Research in Clinical Medicine, China Academy of Chinese Medical Sciences, Beijing, 100700, China
| | - Sixia Yang
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, 510515, China
| | - Binbin Zhao
- Institute of Gerontology, Hubei University of Chinese Medicine, Wuhan, 430065, China
| | - Yaojing Chen
- State Key Laboratory of Cognitive Neuroscience and Learning & IDG/McGovern Institute for Brain Research, Beijing Normal University, Beijing, 100875, China
- BABRI Centre, Beijing Normal University, Beijing, 100875, China
| | - Pengbo Li
- State Key Laboratory of Cognitive Neuroscience and Learning & IDG/McGovern Institute for Brain Research, Beijing Normal University, Beijing, 100875, China
- BABRI Centre, Beijing Normal University, Beijing, 100875, China
| | - Zhanjun Zhang
- State Key Laboratory of Cognitive Neuroscience and Learning & IDG/McGovern Institute for Brain Research, Beijing Normal University, Beijing, 100875, China
- BABRI Centre, Beijing Normal University, Beijing, 100875, China
| | - Junying Zhang
- Institute of Basic Research in Clinical Medicine, China Academy of Chinese Medical Sciences, Beijing, 100700, China.
- State Key Laboratory of Cognitive Neuroscience and Learning & IDG/McGovern Institute for Brain Research, Beijing Normal University, Beijing, 100875, China.
- BABRI Centre, Beijing Normal University, Beijing, 100875, China.
| | - Xuejie Han
- Institute of Basic Research in Clinical Medicine, China Academy of Chinese Medical Sciences, Beijing, 100700, China.
| | - Dongfeng Wei
- Institute of Basic Research in Clinical Medicine, China Academy of Chinese Medical Sciences, Beijing, 100700, China.
| |
Collapse
|
3
|
He XY, Frackowiak J, Dobkin C, Brown WT, Yang SY. Involvement of Type 10 17β-Hydroxysteroid Dehydrogenase in the Pathogenesis of Infantile Neurodegeneration and Alzheimer's Disease. Int J Mol Sci 2023; 24:17604. [PMID: 38139430 PMCID: PMC10743717 DOI: 10.3390/ijms242417604] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2023] [Revised: 12/02/2023] [Accepted: 12/09/2023] [Indexed: 12/24/2023] Open
Abstract
Type 10 17β-hydroxysteroid dehydrogenase (17β-HSD10) is the HSD17B10 gene product playing an appreciable role in cognitive functions. It is the main hub of exercise-upregulated mitochondrial proteins and is involved in a variety of metabolic pathways including neurosteroid metabolism to regulate allopregnanolone homeostasis. Deacetylation of 17β-HSD10 by sirtuins helps regulate its catalytic activities. 17β-HSD10 may also play a critical role in the control of mitochondrial structure, morphology and dynamics by acting as a member of the Parkin/PINK1 pathway, and by binding to cyclophilin D to open mitochondrial permeability pore. 17β-HSD10 also serves as a component of RNase P necessary for mitochondrial tRNA maturation. This dehydrogenase can bind with the Aβ peptide thereby enhancing neurotoxicity to brain cells. Even in the absence of Aβ, its quantitative and qualitative variations can result in neurodegeneration. Since elevated levels of 17β-HSD10 were found in brain cells of Alzheimer's disease (AD) patients and mouse AD models, it is considered to be a key factor in AD pathogenesis. Since data underlying Aβ-binding-alcohol dehydrogenase (ABAD) were not secured from reported experiments, ABAD appears to be a fabricated alternative term for the HSD17B10 gene product. Results of this study would encourage researchers to solve the question why elevated levels of 17β-HSD10 are present in brains of AD patients and mouse AD models. Searching specific inhibitors of 17β-HSD10 may find candidates to reduce senile neurodegeneration and open new approaches for the treatment of AD.
Collapse
Affiliation(s)
- Xue-Ying He
- Department of Molecular Biology, NYS Institute for Basic Research in Developmental Disabilities, Staten Island, NY 10314, USA
| | - Jannusz Frackowiak
- Department of Developmental Neurobiology, NYS Institute for Basic Research in Developmental Disabilities, Staten Island, NY 10314, USA
| | - Carl Dobkin
- Department of Human Genetics, NYS Institute for Basic Research in Developmental Disabilities, Staten Island, NY 10314, USA
| | - William Ted Brown
- Department of Human Genetics, NYS Institute for Basic Research in Developmental Disabilities, Staten Island, NY 10314, USA
| | - Song-Yu Yang
- Department of Molecular Biology, NYS Institute for Basic Research in Developmental Disabilities, Staten Island, NY 10314, USA
- Ph.D. Program in Biology-Neuroscience, Graduate Center of the City, University of New York, New York, NY 10016, USA
| |
Collapse
|
4
|
Hanzlova M, Miskerikova MS, Rotterova A, Chalupova K, Jurkova K, Hamsikova M, Andrys R, Haleckova A, Svobodova J, Schmidt M, Benek O, Musilek K. Nanomolar Benzothiazole-Based Inhibitors of 17β-HSD10 with Cellular Bioactivity. ACS Med Chem Lett 2023; 14:1724-1732. [PMID: 38116418 PMCID: PMC10726454 DOI: 10.1021/acsmedchemlett.3c00355] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2023] [Revised: 11/03/2023] [Accepted: 11/07/2023] [Indexed: 12/21/2023] Open
Abstract
Multifunctional mitochondrial enzyme 17β-hydroxysteroid dehydrogenase type 10 (17β-HSD10) is a potential drug target for the treatment of various pathologies. The most discussed is the pathology associated with Alzheimer's disease (AD), where 17β-HSD10 overexpression and its interaction with amyloid-β peptide contribute to mitochondrial dysfunction and neuronal stress. In this work, a series of new benzothiazole-derived 17β-HSD10 inhibitors were designed based on the structure-activity relationship analysis of formerly published inhibitors. A set of enzyme-based and cell-based methods were used to evaluate the inhibitory potency of new compounds, their interaction with the enzyme, and their cytotoxicity. Most compounds exhibited significantly a higher inhibitory potential compared to published benzothiazolyl ureas and good target engagement in a cellular environment accompanied by low cytotoxicity. The best hits displayed mixed-type inhibition with half maximal inhibitory concentration (IC50) values in the nanomolar range for the purified enzyme (3-7, 15) and/or low micromolar IC50 values in the cell-based assay (6, 13-16).
Collapse
Affiliation(s)
| | | | | | - Katarina Chalupova
- Faculty of Science, Department
of Chemistry, University of Hradec Kralove, Rokitanskeho 62, 500 03 Hradec Kralove, Czech Republic
| | - Katarina Jurkova
- Faculty of Science, Department
of Chemistry, University of Hradec Kralove, Rokitanskeho 62, 500 03 Hradec Kralove, Czech Republic
| | - Marie Hamsikova
- Faculty of Science, Department
of Chemistry, University of Hradec Kralove, Rokitanskeho 62, 500 03 Hradec Kralove, Czech Republic
| | - Rudolf Andrys
- Faculty of Science, Department
of Chemistry, University of Hradec Kralove, Rokitanskeho 62, 500 03 Hradec Kralove, Czech Republic
| | - Annamaria Haleckova
- Faculty of Science, Department
of Chemistry, University of Hradec Kralove, Rokitanskeho 62, 500 03 Hradec Kralove, Czech Republic
| | - Jana Svobodova
- Faculty of Science, Department
of Chemistry, University of Hradec Kralove, Rokitanskeho 62, 500 03 Hradec Kralove, Czech Republic
| | - Monika Schmidt
- Faculty of Science, Department
of Chemistry, University of Hradec Kralove, Rokitanskeho 62, 500 03 Hradec Kralove, Czech Republic
| | - Ondrej Benek
- Faculty of Science, Department
of Chemistry, University of Hradec Kralove, Rokitanskeho 62, 500 03 Hradec Kralove, Czech Republic
| | - Kamil Musilek
- Faculty of Science, Department
of Chemistry, University of Hradec Kralove, Rokitanskeho 62, 500 03 Hradec Kralove, Czech Republic
| |
Collapse
|
5
|
He XY, Dobkin C, Brown WT, Yang SY. Infantile Neurodegeneration Results from Mutants of 17β-Hydroxysteroid Dehydrogenase Type 10 Rather Than Aβ-Binding Alcohol Dehydrogenase. Int J Mol Sci 2023; 24:ijms24108487. [PMID: 37239833 DOI: 10.3390/ijms24108487] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Revised: 04/27/2023] [Accepted: 05/02/2023] [Indexed: 05/28/2023] Open
Abstract
Type 10 17β-hydroxysteroid dehydrogenase (17β-HSD10), a homo-tetrameric multifunctional protein with 1044 residues encoded by the HSD17B10 gene, is necessary for brain cognitive function. Missense mutations result in infantile neurodegeneration, an inborn error in isoleucine metabolism. A 5-methylcytosine hotspot underlying a 388-T transition leads to the HSD10 (p.R130C) mutant to be responsible for approximately half of all cases suffering with this mitochondrial disease. Fewer females suffer with this disease due to X-inactivation. The binding capability of this dehydrogenase to Aβ-peptide may play a role in Alzheimer's disease, but it appears unrelated to infantile neurodegeneration. Research on this enzyme was complicated by reports of a purported Aβ-peptide-binding alcohol dehydrogenase (ABAD), formerly referred to as endoplasmic-reticulum-associated Aβ-binding protein (ERAB). Reports concerning both ABAD and ERAB in the literature reflect features inconsistent with the known functions of 17β-HSD10. It is clarified here that ERAB is reportedly a longer subunit of 17β-HSD10 (262 residues). 17β-HSD10 exhibits L-3-hydroxyacyl-CoA dehydrogenase activity and is thus also referred to in the literature as short-chain 3-hydorxyacyl-CoA dehydrogenase or type II 3-hydorxyacyl-CoA dehydrogenase. However, 17β-HSD10 is not involved in ketone body metabolism, as reported in the literature for ABAD. Reports in the literature referring to ABAD (i.e., 17β-HSD10) as a generalized alcohol dehydrogenase, relying on data underlying ABAD's activities, were found to be unreproducible. Furthermore, the rediscovery of ABAD/ERAB's mitochondrial localization did not cite any published research on 17β-HSD10. Clarification of the purported ABAD/ERAB function derived from these reports on ABAD/ERAB may invigorate this research field and encourage new approaches to the understanding and treatment of HSD17B10-gene-related disorders. We establish here that infantile neurodegeneration is caused by mutants of 17β-HSD10 but not ABAD, and so we conclude that ABAD represents a misnomer employed in high-impact journals.
Collapse
Affiliation(s)
- Xue-Ying He
- Department of Molecular Biology, NYS Institute for Basic Research in Developmental Disabilities, Staten Island, NY 10314, USA
| | - Carl Dobkin
- Department of Human Genetics, NYS Institute for Basic Research in Developmental Disabilities, Staten Island, NY 10314, USA
| | - William Ted Brown
- Central Clinical School, University of Sydney, Sydney 2006, Australia
| | - Song-Yu Yang
- Department of Molecular Biology, NYS Institute for Basic Research in Developmental Disabilities, Staten Island, NY 10314, USA
- Ph.D. Program in Biology-Neuroscience, Graduate Center, City University of New York, New York, NY 10016, USA
| |
Collapse
|
6
|
A High-Throughput Search for SFXN1 Physical Partners Led to the Identification of ATAD3, HSD10 and TIM50. BIOLOGY 2022; 11:biology11091298. [PMID: 36138777 PMCID: PMC9495560 DOI: 10.3390/biology11091298] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Revised: 08/23/2022] [Accepted: 08/28/2022] [Indexed: 11/25/2022]
Abstract
Simple Summary Mitochondria are central players in cell fate and cell death. Indeed, mitochondrial dysfunction has been observed in many diseases, including neurodegenerative diseases. The activity of these organelles relies on numerous mitochondrial transporters, among which the sideroflexins have received little attention to date despite their emerging importance in human health. To better understand the cellular functions of these transporters and their associations with diseases, we herein investigated the molecular partners of one human sideroflexin, SFXN1. Several proteins capable of interacting with SFXN1 were identified, including ATAD3 and HSD10, two mitochondrial proteins linked to neuronal disorders. Abstract Sideroflexins (SFXN, SLC56) are a family of evolutionarily conserved mitochondrial carriers potentially involved in iron homeostasis. One member of the SFXN family is SFXN1, recently identified as a human mitochondrial serine transporter. However, little is known about the SFXN1 interactome, necessitating a high-throughput search to better characterize SFXN1 mitochondrial functions. Via co-immunoprecipitation followed by shotgun mass spectrometry (coIP-MS), we identified 96 putative SFXN1 interactors in the MCF7 human cell line. Our in silico analysis of the SFXN1 interactome highlights biological processes linked to mitochondrial organization, electron transport chains and transmembrane transport. Among the potential physical partners, ATAD3A and 17β-HSD10, two proteins associated with neurological disorders, were confirmed using different human cell lines. Nevertheless, further work will be needed to investigate the significance of these interactions.
Collapse
|
7
|
Yin X, Chan LS, Bose D, Jackson AU, VandeHaar P, Locke AE, Fuchsberger C, Stringham HM, Welch R, Yu K, Fernandes Silva L, Service SK, Zhang D, Hector EC, Young E, Ganel L, Das I, Abel H, Erdos MR, Bonnycastle LL, Kuusisto J, Stitziel NO, Hall IM, Wagner GR, Kang J, Morrison J, Burant CF, Collins FS, Ripatti S, Palotie A, Freimer NB, Mohlke KL, Scott LJ, Wen X, Fauman EB, Laakso M, Boehnke M. Genome-wide association studies of metabolites in Finnish men identify disease-relevant loci. Nat Commun 2022; 13:1644. [PMID: 35347128 PMCID: PMC8960770 DOI: 10.1038/s41467-022-29143-5] [Citation(s) in RCA: 77] [Impact Index Per Article: 25.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2021] [Accepted: 02/23/2022] [Indexed: 01/13/2023] Open
Abstract
Few studies have explored the impact of rare variants (minor allele frequency < 1%) on highly heritable plasma metabolites identified in metabolomic screens. The Finnish population provides an ideal opportunity for such explorations, given the multiple bottlenecks and expansions that have shaped its history, and the enrichment for many otherwise rare alleles that has resulted. Here, we report genetic associations for 1391 plasma metabolites in 6136 men from the late-settlement region of Finland. We identify 303 novel association signals, more than one third at variants rare or enriched in Finns. Many of these signals identify genes not previously implicated in metabolite genome-wide association studies and suggest mechanisms for diseases and disease-related traits.
Collapse
Affiliation(s)
- Xianyong Yin
- Department of Biostatistics and Center for Statistical Genetics, University of Michigan School of Public Health, Ann Arbor, MI, 48109, USA
| | - Lap Sum Chan
- Department of Biostatistics and Center for Statistical Genetics, University of Michigan School of Public Health, Ann Arbor, MI, 48109, USA
| | - Debraj Bose
- Department of Biostatistics and Center for Statistical Genetics, University of Michigan School of Public Health, Ann Arbor, MI, 48109, USA
| | - Anne U Jackson
- Department of Biostatistics and Center for Statistical Genetics, University of Michigan School of Public Health, Ann Arbor, MI, 48109, USA
| | - Peter VandeHaar
- Department of Biostatistics and Center for Statistical Genetics, University of Michigan School of Public Health, Ann Arbor, MI, 48109, USA
| | - Adam E Locke
- McDonnell Genome Institute, Washington University School of Medicine, St Louis, MO, 63108, USA
| | - Christian Fuchsberger
- Department of Biostatistics and Center for Statistical Genetics, University of Michigan School of Public Health, Ann Arbor, MI, 48109, USA
- Institute for Biomedicine, Eurac Research, Bolzano, 39100, Italy
| | - Heather M Stringham
- Department of Biostatistics and Center for Statistical Genetics, University of Michigan School of Public Health, Ann Arbor, MI, 48109, USA
| | - Ryan Welch
- Department of Biostatistics and Center for Statistical Genetics, University of Michigan School of Public Health, Ann Arbor, MI, 48109, USA
| | - Ketian Yu
- Department of Biostatistics and Center for Statistical Genetics, University of Michigan School of Public Health, Ann Arbor, MI, 48109, USA
| | - Lilian Fernandes Silva
- Institute of Clinical Medicine, Internal Medicine, University of Eastern Finland, Kuopio, 70210, Finland
| | - Susan K Service
- Center for Neurobehavioral Genetics, Jane and Terry Semel Institute for Neuroscience and Human Behavior, University of California Los Angeles, Los Angeles, CA, 90024, USA
| | - Daiwei Zhang
- Department of Biostatistics and Center for Statistical Genetics, University of Michigan School of Public Health, Ann Arbor, MI, 48109, USA
- Department of Biostatistics, Epidemiology and Informatics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, 19104, USA
| | - Emily C Hector
- Department of Statistics, North Carolina State University, Raleigh, NC, 27695, USA
| | - Erica Young
- McDonnell Genome Institute, Washington University School of Medicine, St Louis, MO, 63108, USA
- Cardiovascular Division, Department of Medicine, Washington University School of Medicine, St Louis, MO, 63110, USA
| | - Liron Ganel
- McDonnell Genome Institute, Washington University School of Medicine, St Louis, MO, 63108, USA
| | - Indraniel Das
- McDonnell Genome Institute, Washington University School of Medicine, St Louis, MO, 63108, USA
| | - Haley Abel
- Department of Medicine, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Michael R Erdos
- Molecular Genetics Section, Center for Precision Health Research, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Lori L Bonnycastle
- Molecular Genetics Section, Center for Precision Health Research, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Johanna Kuusisto
- Institute of Clinical Medicine, Internal Medicine, University of Eastern Finland, Kuopio, 70210, Finland
- Center for Medicine and Clinical Research, Kuopio University Hospital, Kuopio, 70210, Finland
| | - Nathan O Stitziel
- McDonnell Genome Institute, Washington University School of Medicine, St Louis, MO, 63108, USA
- Cardiovascular Division, Department of Medicine, Washington University School of Medicine, St Louis, MO, 63110, USA
- Department of Genetics, Washington University School of Medicine, St Louis, MO, 63110, USA
| | - Ira M Hall
- Center for Genomic Health, Department of Genetics, Yale University, New Haven, CT, 06510, USA
| | | | - Jian Kang
- Department of Biostatistics and Center for Statistical Genetics, University of Michigan School of Public Health, Ann Arbor, MI, 48109, USA
| | - Jean Morrison
- Department of Biostatistics and Center for Statistical Genetics, University of Michigan School of Public Health, Ann Arbor, MI, 48109, USA
| | - Charles F Burant
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Francis S Collins
- Molecular Genetics Section, Center for Precision Health Research, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Samuli Ripatti
- Institute for Molecular Medicine Finland, FIMM, HiLIFE, University of Helsinki, Helsinki, 00290, Finland
- Department of Public Health, University of Helsinki, Helsinki, 00014, Finland
- Broad Institute of MIT & Harvard, Cambridge, MA, 02142, USA
| | - Aarno Palotie
- Institute for Molecular Medicine Finland, FIMM, HiLIFE, University of Helsinki, Helsinki, 00290, Finland
- Department of Public Health, University of Helsinki, Helsinki, 00014, Finland
- Analytic and Translational Genetics Unit, Department of Medicine, Department of Neurology, and Department of Psychiatry, Massachusetts General Hospital, Boston, MA, 02114, USA
| | - Nelson B Freimer
- Center for Neurobehavioral Genetics, Jane and Terry Semel Institute for Neuroscience and Human Behavior, University of California Los Angeles, Los Angeles, CA, 90024, USA
| | - Karen L Mohlke
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Laura J Scott
- Department of Biostatistics and Center for Statistical Genetics, University of Michigan School of Public Health, Ann Arbor, MI, 48109, USA
| | - Xiaoquan Wen
- Department of Biostatistics and Center for Statistical Genetics, University of Michigan School of Public Health, Ann Arbor, MI, 48109, USA
| | - Eric B Fauman
- Internal Medicine Research Unit, Pfizer Worldwide Research, Development and Medical, Cambridge, MA, 02139, USA.
| | - Markku Laakso
- Institute of Clinical Medicine, Internal Medicine, University of Eastern Finland, Kuopio, 70210, Finland.
| | - Michael Boehnke
- Department of Biostatistics and Center for Statistical Genetics, University of Michigan School of Public Health, Ann Arbor, MI, 48109, USA.
| |
Collapse
|
8
|
He XY, Dobkin C, Brown W, Yang SY. 3-Hydroxyacyl-CoA and Alcohol Dehydrogenase Activities of Mitochondrial Type 10 17β-Hydroxysteroid Dehydrogenase in Neurodegeneration Study. J Alzheimers Dis 2022; 88:1487-1497. [PMID: 35786658 PMCID: PMC9484088 DOI: 10.3233/jad-220481] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/31/2022] [Indexed: 11/15/2022]
Abstract
BACKGROUND Mitochondrial 17β-hydroxysteroid dehydrogenase type 10 (17β-HSD10) is necessary for brain cognitive function, but its studies were confounded by reports of Aβ-peptide binding alcohol dehydrogenase (ABAD), formerly endoplasmic reticulum-associated Aβ-peptide binding protein (ERAB), for two decades so long as ABAD serves as the alternative term of 17β-HSD10. OBJECTIVE To determine whether those ABAD reports are true or false, even if they were published in prestigious journals. METHODS 6xHis-tagged 17β-HSD10 was prepared and characterized by well-established experimental procedures. RESULTS The N-terminal 6xHis tag did not significantly interfere with the dehydrogenase activities of 17β-HSD10, but the kinetic constants of its 3-hydroxyacyl-CoA dehydrogenase activity are drastically distinct from those of ABAD, and it was not involved in ketone body metabolism as previously reported for ABAD. Furthermore, it was impossible to measure its generalized alcohol dehydrogenase activities underlying the concept of ABAD because the experimental procedures described in ABAD reports violated basic chemical and/or biochemical principles. More incredibly, both authors and journals had not yet agreed to make any corrigenda of ABAD reports. CONCLUSION Brain 17β-HSD10 plays a key role in neurosteroid metabolism and further studies in this area may lead to potential treatments of neurodegeneration including AD.
Collapse
Affiliation(s)
- Xue-Ying He
- Department of Molecular Biology, NYS Institute for Basic Research in Developmental Disabilities, Staten Island, NY, USA
| | - Carl Dobkin
- Department of Human Genetics, NYS Institute for Basic Research in Developmental Disabilities, Staten Island, NY, USA
| | - W.Ted Brown
- Department of Human Genetics, NYS Institute for Basic Research in Developmental Disabilities, Staten Island, NY, USA
- Central Clinical School, University of Sydney, Sydney, Australia
| | - Song-Yu Yang
- Department of Molecular Biology, NYS Institute for Basic Research in Developmental Disabilities, Staten Island, NY, USA
- Ph.D. Program in Biology-Neuroscience, Graduate Center of the City University of New York, New York, NY, USA
| |
Collapse
|
9
|
Casciaro F, Persico G, Rusin M, Amatori S, Montgomery C, Rutkowsky JR, Ramsey JJ, Cortopassi G, Fanelli M, Giorgio M. The Histone H3 K4me3, K27me3, and K27ac Genome-Wide Distributions Are Differently Influenced by Sex in Brain Cortexes and Gastrocnemius of the Alzheimer’s Disease PSAPP Mouse Model. EPIGENOMES 2021; 5:epigenomes5040026. [PMID: 34968250 PMCID: PMC8715457 DOI: 10.3390/epigenomes5040026] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2021] [Revised: 11/21/2021] [Accepted: 11/22/2021] [Indexed: 11/24/2022] Open
Abstract
Background: Women represent the majority of Alzheimer’s disease patients and show typical symptoms. Genetic, hormonal, and behavioral mechanisms have been proposed to explain sex differences in dementia prevalence. However, whether sex differences exist in the epigenetic landscape of neuronal tissue during the progression of the disease is still unknown. Methods: To investigate the differences of histone H3 modifications involved in transcription, we determined the genome-wide profiles of H3K4me3, H3K27ac, and H3K27me3 in brain cortexes of an Alzheimer mouse model (PSAPP). Gastrocnemius muscles were also tested since they are known to be different in the two sexes and are affected during the disease progression. Results: Correlation analysis distinguished the samples based on sex for H3K4me3 and H3K27me3 but not for H3K27ac. The analysis of transcription starting sites (TSS) signal distribution, and analysis of bounding sites revealed that gastrocnemius is more influenced than brain by sex for the three histone modifications considered, exception made for H3K27me3 distribution on the X chromosome which showed sex-related differences in promoters belonging to behavior and cellular or neuronal spheres in mice cortexes. Conclusions: H3K4me3, H3K27ac, and H3K27me3 signals are slightly affected by sex in brain, with the exception of H3K27me3, while a higher number of differences can be found in gastrocnemius.
Collapse
Affiliation(s)
- Francesca Casciaro
- Department of Biomedical Sciences, University of Padua, Via Ugo Bassi 58/B, 35131 Padova, Italy
| | - Giuseppe Persico
- Department of Experimental Oncology, IRCCS—European Institute of Oncology, Via Adamello 16, 20139 Milano, Italy; (G.P.); (M.R.)
| | - Martina Rusin
- Department of Experimental Oncology, IRCCS—European Institute of Oncology, Via Adamello 16, 20139 Milano, Italy; (G.P.); (M.R.)
- Molecular Pathology Laboratory “PaoLa”, Department of Biomolecular Sciences, University of Urbino Carlo Bo, Via Arco d’Augusto 2, 61032 Fano (PU), Italy; (S.A.); (M.F.)
| | - Stefano Amatori
- Molecular Pathology Laboratory “PaoLa”, Department of Biomolecular Sciences, University of Urbino Carlo Bo, Via Arco d’Augusto 2, 61032 Fano (PU), Italy; (S.A.); (M.F.)
| | - Claire Montgomery
- School of Veterinary Medicine, University of California, Davis, CA 95616, USA; (C.M.); (J.R.R.); (J.J.R.); (G.C.)
| | - Jennifer R. Rutkowsky
- School of Veterinary Medicine, University of California, Davis, CA 95616, USA; (C.M.); (J.R.R.); (J.J.R.); (G.C.)
| | - Jon J. Ramsey
- School of Veterinary Medicine, University of California, Davis, CA 95616, USA; (C.M.); (J.R.R.); (J.J.R.); (G.C.)
| | - Gino Cortopassi
- School of Veterinary Medicine, University of California, Davis, CA 95616, USA; (C.M.); (J.R.R.); (J.J.R.); (G.C.)
| | - Mirco Fanelli
- Molecular Pathology Laboratory “PaoLa”, Department of Biomolecular Sciences, University of Urbino Carlo Bo, Via Arco d’Augusto 2, 61032 Fano (PU), Italy; (S.A.); (M.F.)
| | - Marco Giorgio
- Department of Biomedical Sciences, University of Padua, Via Ugo Bassi 58/B, 35131 Padova, Italy
- Department of Experimental Oncology, IRCCS—European Institute of Oncology, Via Adamello 16, 20139 Milano, Italy; (G.P.); (M.R.)
- Correspondence: ; Tel.: +39-04-9827-6060
| |
Collapse
|
10
|
Fišar Z, Musílek K, Benek O, Hroch L, Vinklářová L, Schmidt M, Hroudová J, Raboch J. Effects of novel 17β-hydroxysteroid dehydrogenase type 10 inhibitors on mitochondrial respiration. Toxicol Lett 2020; 339:12-19. [PMID: 33359020 DOI: 10.1016/j.toxlet.2020.12.012] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2020] [Revised: 12/02/2020] [Accepted: 12/19/2020] [Indexed: 12/29/2022]
Abstract
Mitochondrial enzymes are targets of newly synthesized drugs being tested for the treatment of neurodegenerative disorders, such as Alzheimer's disease (AD). The enzyme 17β-hydroxysteroid dehydrogenase type 10 (HSD10) is a multifunctional mitochondrial protein that is thought to play a role in the pathophysiology of AD and is one of the targets of new potential AD drugs. The in vitro effects of frentizole, riluzole, AG18051, and 42 novel modulators of HSD10 (potential AD drugs) on citrate synthase (CS) activity, monoamine oxidase (MAO) activity, complex I- or complex II-linked mitochondrial respiratory rate, and complex I activity were measured in isolated pig brain mitochondria. Based on their minimal inhibitory effects on the respiratory rate of mitochondria and CS and complex I activity, six novel compounds were selected for further testing. Assuming that inhibition of MAO-B could be a desirable effect of AD drugs, only AG18051 and one new compound met the criteria for MAO-B inhibition with minimal drug-induced effects on mitochondrial respiration. In conclusion, our in vitro screening of mitochondrial effect of novel potential AD drugs has enabled the selection of the most promising molecules for further testing that are relatively safe in terms of drug-induced mitochondrial toxicity.
Collapse
Affiliation(s)
- Zdeněk Fišar
- Charles University and General University Hospital in Prague, First Faculty of Medicine, Department of Psychiatry, Ke Karlovu 11, 120 00, Prague 2, Czech Republic.
| | - Kamil Musílek
- University of Hradec Kralove, Faculty of Science, Department of Chemistry, Rokitanskeho 62, 500 03, Hradec Kralove, Czech Republic.
| | - Ondřej Benek
- University of Hradec Kralove, Faculty of Science, Department of Chemistry, Rokitanskeho 62, 500 03, Hradec Kralove, Czech Republic
| | - Lukáš Hroch
- University Hospital in Hradec Kralove, Sokolska 581, 500 05, Hradec Kralove, Czech Republic
| | - Lucie Vinklářová
- University of Hradec Kralove, Faculty of Science, Department of Chemistry, Rokitanskeho 62, 500 03, Hradec Kralove, Czech Republic
| | - Monika Schmidt
- University of Hradec Kralove, Faculty of Science, Department of Chemistry, Rokitanskeho 62, 500 03, Hradec Kralove, Czech Republic
| | - Jana Hroudová
- Charles University and General University Hospital in Prague, First Faculty of Medicine, Department of Psychiatry, Ke Karlovu 11, 120 00, Prague 2, Czech Republic
| | - Jiří Raboch
- Charles University and General University Hospital in Prague, First Faculty of Medicine, Department of Psychiatry, Ke Karlovu 11, 120 00, Prague 2, Czech Republic
| |
Collapse
|
11
|
Boutin S, Maltais R, Roy J, Poirier D. Synthesis of 17β-hydroxysteroid dehydrogenase type 10 steroidal inhibitors: Selectivity, metabolic stability and enhanced potency. Eur J Med Chem 2020; 209:112909. [PMID: 33081987 DOI: 10.1016/j.ejmech.2020.112909] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2020] [Revised: 09/28/2020] [Accepted: 10/03/2020] [Indexed: 12/14/2022]
Abstract
17beta-Hydroxysteroid dehydrogenase type 10 (17β-HSD10) is the only mitochondrial member of 17β-HSD family. This enzyme can oxidize estradiol (E2) into estrone (E1), thus reducing concentration of this neuroprotective steroid. Since 17β-HSD10 possesses properties that suggest a possible role in Alzheimer's disease, its inhibition appears to be a therapeutic strategy. After we identified the androsterone (ADT) derivative 1 as a first steroidal inhibitor of 17β-HSD10, new analogs were synthesized to increase the metabolic stability, to improve the selectivity of inhibition over 17β-HSD3 and to optimize the inhibitory potency. From six D-ring derivatives of 1 (17-CO), two compounds (17β-H/17α-OH and 17β-OH/17α-CCH) were more metabolically stable and did not inhibit the 17β-HSD3. Moreover, solid phase synthesis was used to extend the molecular diversity on the 3β-piperazinylmethyl group of the steroid base core. Eight over 120 new derivatives were more potent inhibitors than 1 for the transformation of E2 to E1, with the 4-(4-trifluoromethyl-3-methoxybenzyl)piperazin-1-ylmethyl-ADT (D-3,7) being 16 times more potent (IC50 = 0.14 μM). Finally, D-ring modification of D-3,7 provided 17β-OH/17α-CCH derivative 25 and 17β-H/17α-OH derivative 26, which were more potent inhibitor than 1 (1.8 and 2.4 times, respectively).
Collapse
Affiliation(s)
- Sophie Boutin
- Laboratory of Medicinal Chemistry, Endocrinology and Nephrology Unit, CHU de Québec - Research Center, Québec, QC, Canada; Department of Molecular Medicine, Faculty of Medicine, Université Laval, Québec, QC, Canada
| | - René Maltais
- Laboratory of Medicinal Chemistry, Endocrinology and Nephrology Unit, CHU de Québec - Research Center, Québec, QC, Canada
| | - Jenny Roy
- Laboratory of Medicinal Chemistry, Endocrinology and Nephrology Unit, CHU de Québec - Research Center, Québec, QC, Canada
| | - Donald Poirier
- Laboratory of Medicinal Chemistry, Endocrinology and Nephrology Unit, CHU de Québec - Research Center, Québec, QC, Canada; Department of Molecular Medicine, Faculty of Medicine, Université Laval, Québec, QC, Canada.
| |
Collapse
|
12
|
Pomeshchik Y, Klementieva O, Gil J, Martinsson I, Hansen MG, de Vries T, Sancho-Balsells A, Russ K, Savchenko E, Collin A, Vaz AR, Bagnoli S, Nacmias B, Rampon C, Sorbi S, Brites D, Marko-Varga G, Kokaia Z, Rezeli M, Gouras GK, Roybon L. Human iPSC-Derived Hippocampal Spheroids: An Innovative Tool for Stratifying Alzheimer Disease Patient-Specific Cellular Phenotypes and Developing Therapies. Stem Cell Reports 2020; 15:256-273. [PMID: 32589876 PMCID: PMC7363942 DOI: 10.1016/j.stemcr.2020.06.001] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2019] [Revised: 06/01/2020] [Accepted: 06/01/2020] [Indexed: 12/12/2022] Open
Abstract
The hippocampus is important for memory formation and is severely affected in the brain with Alzheimer disease (AD). Our understanding of early pathogenic processes occurring in hippocampi in AD is limited due to tissue unavailability. Here, we report a chemical approach to rapidly generate free-floating hippocampal spheroids (HSs), from human induced pluripotent stem cells. When used to model AD, both APP and atypical PS1 variant HSs displayed increased Aβ42/Aβ40 peptide ratios and decreased synaptic protein levels, which are common features of AD. However, the two variants differed in tau hyperphosphorylation, protein aggregation, and protein network alterations. NeuroD1-mediated gene therapy in HSs-derived progenitors resulted in modulation of expression of numerous genes, including those involved in synaptic transmission. Thus, HSs can be harnessed to unravel the mechanisms underlying early pathogenic changes in the hippocampi of AD patients, and provide a robust platform for the development of therapeutic strategies targeting early stage AD.
Collapse
Affiliation(s)
- Yuriy Pomeshchik
- iPSC Laboratory for CNS Disease Modeling, Department of Experimental Medical Science, BMC D10, Lund University, Lund SE-221 84, Sweden; Strategic Research Area MultiPark, Lund University, Lund SE-221 84, Sweden; Lund Stem Cell Center, Lund University, Lund SE-221 84, Sweden
| | - Oxana Klementieva
- Strategic Research Area MultiPark, Lund University, Lund SE-221 84, Sweden; Medical Microspectroscopy, Department of Experimental Medical Science, BMC B11, Lund University, Lund SE-221 84, Sweden; Experimental Dementia Research Unit, Department of Experimental Medical Science, BMC B11, Lund University, Lund SE-221 84, Sweden
| | - Jeovanis Gil
- Clinical Protein Science and Imaging, Department of Biomedical Engineering, BMC D13, Lund University, Lund SE-221 84, Sweden
| | - Isak Martinsson
- Strategic Research Area MultiPark, Lund University, Lund SE-221 84, Sweden; Experimental Dementia Research Unit, Department of Experimental Medical Science, BMC B11, Lund University, Lund SE-221 84, Sweden
| | - Marita Grønning Hansen
- Lund Stem Cell Center, Lund University, Lund SE-221 84, Sweden; Laboratory of Stem Cells and Restorative Neurology, Department of Clinical Sciences, BMC B10, Lund University, Lund SE-221 84, Sweden
| | - Tessa de Vries
- iPSC Laboratory for CNS Disease Modeling, Department of Experimental Medical Science, BMC D10, Lund University, Lund SE-221 84, Sweden; Strategic Research Area MultiPark, Lund University, Lund SE-221 84, Sweden; Lund Stem Cell Center, Lund University, Lund SE-221 84, Sweden
| | - Anna Sancho-Balsells
- iPSC Laboratory for CNS Disease Modeling, Department of Experimental Medical Science, BMC D10, Lund University, Lund SE-221 84, Sweden; Strategic Research Area MultiPark, Lund University, Lund SE-221 84, Sweden; Lund Stem Cell Center, Lund University, Lund SE-221 84, Sweden
| | - Kaspar Russ
- iPSC Laboratory for CNS Disease Modeling, Department of Experimental Medical Science, BMC D10, Lund University, Lund SE-221 84, Sweden; Strategic Research Area MultiPark, Lund University, Lund SE-221 84, Sweden; Lund Stem Cell Center, Lund University, Lund SE-221 84, Sweden
| | - Ekaterina Savchenko
- iPSC Laboratory for CNS Disease Modeling, Department of Experimental Medical Science, BMC D10, Lund University, Lund SE-221 84, Sweden; Strategic Research Area MultiPark, Lund University, Lund SE-221 84, Sweden; Lund Stem Cell Center, Lund University, Lund SE-221 84, Sweden
| | - Anna Collin
- Department of Clinical Genetics and Pathology, Office for Medical Services, Lund SE-221 85, Sweden
| | - Ana Rita Vaz
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Lisbon, Portugal; Department of Biochemistry and Human Biology, Faculty of Pharmacy, Universidade de Lisboa, Lisbon, Portugal
| | - Silvia Bagnoli
- Laboratorio di Neurogenetica, Dipartimento di Neuroscienze, Psicologia, Area del Farmaco e Salute del Bambino- NEUROFARBA, Università degli Studi di Firenze, Florence 50134, Italy; IRCCS Fondazione Don Carlo Gnocchi, Florence, Italy
| | - Benedetta Nacmias
- Laboratorio di Neurogenetica, Dipartimento di Neuroscienze, Psicologia, Area del Farmaco e Salute del Bambino- NEUROFARBA, Università degli Studi di Firenze, Florence 50134, Italy; IRCCS Fondazione Don Carlo Gnocchi, Florence, Italy
| | - Claire Rampon
- Centre de Recherches sur la Cognition Animale (CRCA), Centre de Biologie Intégrative (CBI), Université de Toulouse; CNRS, UPS, Toulouse Cedex 9, France
| | - Sandro Sorbi
- Laboratorio di Neurogenetica, Dipartimento di Neuroscienze, Psicologia, Area del Farmaco e Salute del Bambino- NEUROFARBA, Università degli Studi di Firenze, Florence 50134, Italy; IRCCS Fondazione Don Carlo Gnocchi, Florence, Italy
| | - Dora Brites
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Lisbon, Portugal; Department of Biochemistry and Human Biology, Faculty of Pharmacy, Universidade de Lisboa, Lisbon, Portugal
| | - György Marko-Varga
- Clinical Protein Science and Imaging, Department of Biomedical Engineering, BMC D13, Lund University, Lund SE-221 84, Sweden
| | - Zaal Kokaia
- Lund Stem Cell Center, Lund University, Lund SE-221 84, Sweden; Laboratory of Stem Cells and Restorative Neurology, Department of Clinical Sciences, BMC B10, Lund University, Lund SE-221 84, Sweden
| | - Melinda Rezeli
- Clinical Protein Science and Imaging, Department of Biomedical Engineering, BMC D13, Lund University, Lund SE-221 84, Sweden
| | - Gunnar K Gouras
- Strategic Research Area MultiPark, Lund University, Lund SE-221 84, Sweden; Experimental Dementia Research Unit, Department of Experimental Medical Science, BMC B11, Lund University, Lund SE-221 84, Sweden
| | - Laurent Roybon
- iPSC Laboratory for CNS Disease Modeling, Department of Experimental Medical Science, BMC D10, Lund University, Lund SE-221 84, Sweden; Strategic Research Area MultiPark, Lund University, Lund SE-221 84, Sweden; Lund Stem Cell Center, Lund University, Lund SE-221 84, Sweden.
| |
Collapse
|
13
|
Steroids and Alzheimer's Disease: Changes Associated with Pathology and Therapeutic Potential. Int J Mol Sci 2020; 21:ijms21134812. [PMID: 32646017 PMCID: PMC7370115 DOI: 10.3390/ijms21134812] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2020] [Revised: 07/02/2020] [Accepted: 07/03/2020] [Indexed: 12/12/2022] Open
Abstract
Alzheimer's disease (AD) is a multifactorial age-related neurodegenerative disease that today has no effective treatment to prevent or slow its progression. Neuroactive steroids, including neurosteroids and sex steroids, have attracted attention as potential suitable candidates to alleviate AD pathology. Accumulating evidence shows that they exhibit pleiotropic neuroprotective properties that are relevant for AD. This review focuses on the relationship between selected neuroactive steroids and the main aspects of AD disease, pointing out contributions and gaps with reference to sex differences. We take into account the regulation of brain steroid concentrations associated with human AD pathology. Consideration is given to preclinical studies in AD models providing current knowledge on the neuroprotection offered by neuroactive (neuro)steroids on major AD pathogenic factors, such as amyloid-β (Aβ) and tau pathology, mitochondrial impairment, neuroinflammation, neurogenesis and memory loss. Stimulating endogenous steroid production opens a new steroid-based strategy to potentially overcome AD pathology. This article is part of a Special Issue entitled Steroids and the Nervous System.
Collapse
|
14
|
Vinklarova L, Schmidt M, Benek O, Kuca K, Gunn-Moore F, Musilek K. Friend or enemy? Review of 17β-HSD10 and its role in human health or disease. J Neurochem 2020; 155:231-249. [PMID: 32306391 DOI: 10.1111/jnc.15027] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2019] [Revised: 03/26/2020] [Accepted: 04/10/2020] [Indexed: 12/17/2022]
Abstract
17β-hydroxysteroid dehydrogenase (17β-HSD10) is a multifunctional human enzyme with important roles both as a structural component and also as a catalyst of many metabolic pathways. This mitochondrial enzyme has important functions in the metabolism, development and aging of the neural system, where it is involved in the homeostasis of neurosteroids, especially in regard to estradiol, changes in which make it an essential part of neurodegenerative pathology. These roles therefore, indicate that 17β-HSD10 may be a possible druggable target for neurodegenerative diseases including Alzheimer's disease (AD), and in hormone-dependent cancer. The objective of this review was to provide a summary about physiological functions and pathological roles of 17β-HSD10 and the modulators of its activity.
Collapse
Affiliation(s)
- Lucie Vinklarova
- Faculty of Science, Department of Chemistry, University of Hradec Kralove, Hradec Kralove, Czech Republic
| | - Monika Schmidt
- Faculty of Science, Department of Chemistry, University of Hradec Kralove, Hradec Kralove, Czech Republic
| | - Ondrej Benek
- Faculty of Science, Department of Chemistry, University of Hradec Kralove, Hradec Kralove, Czech Republic
| | - Kamil Kuca
- Faculty of Science, Department of Chemistry, University of Hradec Kralove, Hradec Kralove, Czech Republic
| | | | - Kamil Musilek
- Faculty of Science, Department of Chemistry, University of Hradec Kralove, Hradec Kralove, Czech Republic
| |
Collapse
|
15
|
Bai H, Sun K, Wu JH, Zhong ZH, Xu SL, Zhang HR, Gu YH, Lu SF. Proteomic and metabolomic characterization of cardiac tissue in acute myocardial ischemia injury rats. PLoS One 2020; 15:e0231797. [PMID: 32365112 PMCID: PMC7197859 DOI: 10.1371/journal.pone.0231797] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2020] [Accepted: 03/31/2020] [Indexed: 12/12/2022] Open
Abstract
The pathological process and mechanism of myocardial ischemia (MI) is very complicated, and remains unclear. An integrated proteomic-metabolomics analysis was applied to comprehensively understand the pathological changes and mechanism of MI. Male Sprague-Dawley rats were randomly divided into a mock surgery (MS) group and an MI group. The MI model was made by ligating the left anterior descending coronary artery, twenty-four hours after which, echocardiography was employed to assess left ventricular (LV) function variables. Blood samples and left ventricular tissues were collected for ELISA, metabolomics and proteomics analysis. The results showed that LV function, including ejection fraction (EF) and fractional shortening (FS), was significantly reduced and the level of cTnT in the serum increased after MI. iTRAQ proteomics showed that a total of 169 proteins were altered including 52 and 117 proteins with increased and decreased expression, respectively, which were mainly involved in the following activities: complement and coagulation cascades, tight junction, regulation of actin cytoskeleton, MAPK signaling pathway, endocytosis, NOD-like receptor signaling pathway, as well as phagosome coupled with vitamin digestion and absorption. Altered metabolomic profiling of this transition was mostly enriched in pathways including ABC transporters, glycerophospholipid metabolism, protein digestion and absorption and aminoacyl-tRNA biosynthesis. The integrated metabolomics and proteomics analysis indicated that myocardial injury after MI is closely related to several metabolic pathways, especially energy metabolism, amino acid metabolism, vascular smooth muscle contraction, gap junction and neuroactive ligand-receptor interaction. These findings may contribute to understanding the mechanism of MI and have implication for new therapeutic targets.
Collapse
Affiliation(s)
- Hua Bai
- Acupuncture and Tuina college, Nanjing University of Chinese Medicine, Nanjing, China
| | - Ke Sun
- Acupuncture and Tuina college, Nanjing University of Chinese Medicine, Nanjing, China
| | - Jia-Hong Wu
- Acupuncture and Tuina college, Nanjing University of Chinese Medicine, Nanjing, China
| | - Ze-Hao Zhong
- Acupuncture and Tuina college, Nanjing University of Chinese Medicine, Nanjing, China
| | - Sen-Lei Xu
- Acupuncture and Tuina college, Nanjing University of Chinese Medicine, Nanjing, China
| | - Hong-Ru Zhang
- Acupuncture and Tuina college, Nanjing University of Chinese Medicine, Nanjing, China
| | - Yi-Huang Gu
- Acupuncture and Tuina college, Nanjing University of Chinese Medicine, Nanjing, China
- * E-mail: (SFL); (YHG)
| | - Sheng-Feng Lu
- Acupuncture and Tuina college, Nanjing University of Chinese Medicine, Nanjing, China
- Key Laboratory of Acupuncture and Medicine Research of Ministry of Education, Nanjing University of Chinese Medicine, Nanjing, China
- * E-mail: (SFL); (YHG)
| |
Collapse
|
16
|
Luo Y, Li D, Liao Y, Cai C, Wu Q, Ke H, Liu X, Li H, Hong H, Xu Y, Wang Q, Fang J, Fang S. Systems Pharmacology Approach to Investigate the Mechanism of Kai-Xin-San in Alzheimer's Disease. Front Pharmacol 2020; 11:381. [PMID: 32317964 PMCID: PMC7147119 DOI: 10.3389/fphar.2020.00381] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2019] [Accepted: 03/12/2020] [Indexed: 12/17/2022] Open
Abstract
Alzheimer's disease (AD) is a complex neurodegenerative disease characterized by cognitive dysfunction. Kai-Xin-San (KXS) is a traditional Chinese medicine (TCM) formula that has been used to treat AD patients for over a thousand years in China. However, the therapeutic mechanisms of KXS for treating AD have not been fully explored. Herein, we used a comprehensive network pharmacology approach to investigate the mechanism of action of KXS in the treatment of AD. This approach consists of construction of multiple networks and Gene Ontology enrichment and pathway analyses. Furthermore, animal experiments were performed to validate the predicted molecular mechanisms obtained from the systems pharmacology-based analysis. As a result, 50 chemicals in KXS and 39 AD-associated proteins were identified as major active compounds and targets, respectively. The therapeutic mechanisms of KXS in treating AD were primarily related to the regulation of four pathology modules, including amyloid beta metabolism, tau protein hyperphosphorylation process, cholinergic dysfunction, and inflammation. In scopolamine-induced cognitive dysfunction mice, we validated the anti-inflammatory effects of KXS on AD by determining the levels of inflammation cytokines including interleukin (IL)-6, IL-1β, and tumor necrosis factor (TNF)-α. We also found cholinergic system dysfunction amelioration of KXS is correlated with upregulation of the cholinergic receptor CHRNB2. In conclusion, our work proposes a comprehensive systems pharmacology approach to explore the underlying therapeutic mechanism of KXS for the treatment of AD.
Collapse
Affiliation(s)
- Yunxia Luo
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, China
- Department of Endocrinology, Fourth Clinical Medical College, Guangzhou University of Chinese Medicine, Shenzhen, China
| | - Dongli Li
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Yanfang Liao
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Chuipu Cai
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Qihui Wu
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Hanzhong Ke
- Department of Cancer Immunology and Virology, Dana-Farber Cancer Institute, Boston, MA, United States
- Department of Medicine, Harvard Medical School, Boston, MA, United States
| | - Xinning Liu
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Huilin Li
- Department of Endocrinology, Fourth Clinical Medical College, Guangzhou University of Chinese Medicine, Shenzhen, China
| | - Honghai Hong
- Department of Clinical Laboratory, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Yumin Xu
- Department of Encephalopathy First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou, China
| | - Qi Wang
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, China
- Institute of Clinical Pharmacology, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Jiansong Fang
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, China
- Institute of Clinical Pharmacology, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Shuhuan Fang
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, China
- Institute of Clinical Pharmacology, Guangzhou University of Chinese Medicine, Guangzhou, China
| |
Collapse
|
17
|
He XY, Dobkin C, Yang SY. 17β-Hydroxysteroid dehydrogenases and neurosteroid metabolism in the central nervous system. Mol Cell Endocrinol 2019; 489:92-97. [PMID: 30321584 DOI: 10.1016/j.mce.2018.10.002] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/29/2017] [Revised: 08/21/2018] [Accepted: 10/04/2018] [Indexed: 12/21/2022]
Abstract
17β-Hydroxysteroid dehydrogenases are indispensable for downstream enzyme steps of the neurosteroidogenesis. Neurosteroids are synthesized de novo in neurons and glia from cholesterol transported into mitochondria, or by conversion from proneurosteroids, e. g. dehydroepiandrosterone (DHEA) and pregnenolone, through the same metabolic pathway as revealed in the de novo neurosteroidogenesis. Hormonal steroids generated from endocrine glands are transported into brain from the circulation to exert neuronal activity via genomic pathway, whereas neurosteroids produced in brain cells without genomic targets identified could bind to cell surface targets, e.g., GABAA or NMDA receptors and elicit antidepressant, anxiolytic, anticonvulsant and anesthetic effects by regulating neuroexcitability. In a broad sense, neurosteroids include hormonal steroids in brain, and they are irrespective of origin playing important roles in brain development including neuroprotection, neurogenesis and neural plasticity. They are also a critical element in cognitive and memory functions. Mitochondrial 17β-HSD10, encoded by the HSD17B10 gene mapping to Xp11.2, is found in various brain regions, essential for the maintenance of neurosteroid homeostasis. Mutations identified in this gene resulted in two distinct brain diseases, namely HSD10 deficiency and MRXS10, of which clinical presentations and pathogenetic mechanisms are quite different. Since elevated levels of 17β-HSD10 was found in brains of Alzheimer's disease patients and AD mouse model, it may also act as an adverse factor in the AD pathogenesis due to an imbalance of neurosteroid metabolism.
Collapse
Affiliation(s)
- Xue-Ying He
- Laboratory of Medical Biochemistry, NYS Institute for Basic Research in Developmental Disabilities, Staten Island, NY, 10314, USA
| | - Carl Dobkin
- Department of Molecular Genetics, NYS Institute for Basic Research in Developmental Disabilities, Staten Island, NY, 10314, USA
| | - Song-Yu Yang
- Laboratory of Medical Biochemistry, NYS Institute for Basic Research in Developmental Disabilities, Staten Island, NY, 10314, USA; Ph.D. Program in Biology-Neuroscience, Graduate Center of the City University of New York, New York, NY, 10016, USA.
| |
Collapse
|
18
|
Hiltunen JK, Kastaniotis AJ, Autio KJ, Jiang G, Chen Z, Glumoff T. 17B-hydroxysteroid dehydrogenases as acyl thioester metabolizing enzymes. Mol Cell Endocrinol 2019; 489:107-118. [PMID: 30508570 DOI: 10.1016/j.mce.2018.11.012] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/30/2018] [Revised: 11/23/2018] [Accepted: 11/23/2018] [Indexed: 01/10/2023]
Abstract
17β-Hydroxysteroid dehydrogenases (HSD17B) catalyze the oxidation/reduction of 17β-hydroxy/keto group in position C17 in C18- and C19 steroids. Most HSD17Bs are also catalytically active with substrates other than steroids. A subset of these enzymes is able to process thioesters of carboxylic acids. This group of enzymes includes HSD17B4, HSD17B8, HSD17B10 and HSD17B12, which execute reactions in intermediary metabolism, participating in peroxisomal β-oxidation of fatty acids, mitochondrial oxidation of 3R-hydroxyacyl-groups, breakdown of isoleucine and fatty acid chain elongation in endoplasmic reticulum. Divergent substrate acceptance capabilities exemplify acquirement of catalytic site adaptiveness during evolution. As an additional common feature these HSD17Bs are multifunctional enzymes that arose either via gene fusions (HSD17B4) or are incorporated as subunits into multifunctional protein complexes (HSD17B8 and HSD17B10). Crystal structures of HSD17B4, HSD17B8 and HSD17B10 give insight into their structure-function relationships. Thus far, deficiencies of HSD17B4 and HSD17B10 have been assigned to inborn errors in humans, underlining their significance as enzymes of metabolism.
Collapse
Affiliation(s)
- J Kalervo Hiltunen
- Faculty of Biochemistry and Molecular Medicine, University of Oulu, Oulu, Finland; State Key Laboratory of Supramolecular Structure and Materials and Institute of Theoretical Chemistry, Jilin University, 2699 Qianjin Street, Changchun, 130012, PR China.
| | | | - Kaija J Autio
- Faculty of Biochemistry and Molecular Medicine, University of Oulu, Oulu, Finland
| | - Guangyu Jiang
- Faculty of Biochemistry and Molecular Medicine, University of Oulu, Oulu, Finland
| | - Zhijun Chen
- Faculty of Biochemistry and Molecular Medicine, University of Oulu, Oulu, Finland; State Key Laboratory of Supramolecular Structure and Materials and Institute of Theoretical Chemistry, Jilin University, 2699 Qianjin Street, Changchun, 130012, PR China
| | - Tuomo Glumoff
- Faculty of Biochemistry and Molecular Medicine, University of Oulu, Oulu, Finland
| |
Collapse
|
19
|
de Crécy-Lagard V, Boccaletto P, Mangleburg CG, Sharma P, Lowe TM, Leidel SA, Bujnicki JM. Matching tRNA modifications in humans to their known and predicted enzymes. Nucleic Acids Res 2019; 47:2143-2159. [PMID: 30698754 PMCID: PMC6412123 DOI: 10.1093/nar/gkz011] [Citation(s) in RCA: 111] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2018] [Revised: 12/28/2018] [Accepted: 01/10/2019] [Indexed: 12/25/2022] Open
Abstract
tRNA are post-transcriptionally modified by chemical modifications that affect all aspects of tRNA biology. An increasing number of mutations underlying human genetic diseases map to genes encoding for tRNA modification enzymes. However, our knowledge on human tRNA-modification genes remains fragmentary and the most comprehensive RNA modification database currently contains information on approximately 20% of human cytosolic tRNAs, primarily based on biochemical studies. Recent high-throughput methods such as DM-tRNA-seq now allow annotation of a majority of tRNAs for six specific base modifications. Furthermore, we identified large gaps in knowledge when we predicted all cytosolic and mitochondrial human tRNA modification genes. Only 48% of the candidate cytosolic tRNA modification enzymes have been experimentally validated in mammals (either directly or in a heterologous system). Approximately 23% of the modification genes (cytosolic and mitochondrial combined) remain unknown. We discuss these 'unidentified enzymes' cases in detail and propose candidates whenever possible. Finally, tissue-specific expression analysis shows that modification genes are highly expressed in proliferative tissues like testis and transformed cells, but scarcely in differentiated tissues, with the exception of the cerebellum. Our work provides a comprehensive up to date compilation of human tRNA modifications and their enzymes that can be used as a resource for further studies.
Collapse
Affiliation(s)
- Valérie de Crécy-Lagard
- Department of Microbiology and Cell Sciences, University of Florida, Gainesville, FL 32611, USA
- Cancer and Genetic Institute, University of Florida, Gainesville, FL 32611, USA
| | - Pietro Boccaletto
- Laboratory of Bioinformatics and Protein Engineering, International Institute of Molecular and Cell Biology, ul. Trojdena 4, 02-109 Warsaw, Poland
| | - Carl G Mangleburg
- Department of Microbiology and Cell Sciences, University of Florida, Gainesville, FL 32611, USA
| | - Puneet Sharma
- Max Planck Research Group for RNA Biology, Max Planck Institute for Molecular Biomedicine, 48149 Muenster, Germany
- Cells-in-Motion Cluster of Excellence, University of Muenster, 48149 Muenster, Germany
| | - Todd M Lowe
- Department of Biomolecular Engineering, University of California, Santa Cruz, Santa Cruz, CA 95064, USA
| | - Sebastian A Leidel
- Max Planck Research Group for RNA Biology, Max Planck Institute for Molecular Biomedicine, 48149 Muenster, Germany
- Cells-in-Motion Cluster of Excellence, University of Muenster, 48149 Muenster, Germany
- Research Group for RNA Biochemistry, Institute of Chemistry and Biochemistry, University of Bern, 3012 Bern, Switzerland
| | - Janusz M Bujnicki
- Laboratory of Bioinformatics and Protein Engineering, International Institute of Molecular and Cell Biology, ul. Trojdena 4, 02-109 Warsaw, Poland
- Institute of Molecular Biology and Biotechnology, Faculty of Biology, Adam Mickiewicz University, ul. Umultowska 89, 61-614 Poznań, Poland
| |
Collapse
|
20
|
Boutin S, Roy J, Maltais R, Alata W, Calon F, Poirier D. Identification of steroidal derivatives inhibiting the transformations of allopregnanolone and estradiol by 17β-hydroxysteroid dehydrogenase type 10. Bioorg Med Chem Lett 2018; 28:3554-3559. [DOI: 10.1016/j.bmcl.2018.09.031] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2018] [Revised: 09/20/2018] [Accepted: 09/24/2018] [Indexed: 12/17/2022]
|
21
|
Morsy A, Trippier PC. Amyloid-Binding Alcohol Dehydrogenase (ABAD) Inhibitors for the Treatment of Alzheimer’s Disease. J Med Chem 2018; 62:4252-4264. [DOI: 10.1021/acs.jmedchem.8b01530] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Affiliation(s)
- Ahmed Morsy
- Department of Pharmaceutical Sciences, School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, Texas 79106, United States
| | - Paul C. Trippier
- Department of Pharmaceutical Sciences, School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, Texas 79106, United States
- Center for Chemical Biology, Department of Chemistry and Biochemistry, Texas Tech University, Lubbock, Texas 79409, United States
| |
Collapse
|
22
|
Structure Confirmation and Evaluation of a Nonsteroidal Inhibitor of 17β-Hydroxysteroid Dehydrogenase Type 10. MAGNETOCHEMISTRY 2018. [DOI: 10.3390/magnetochemistry4030032] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
17β-Hydroxysteroid dehydrogenase type 10 (17β-HSD10) is a steroidogenesis enzyme known for its potential role in Alzheimer’s disease. For comparison purposes between steroidal and nonsteroidal 17β-HSD10 inhibitors 1 and 2, respectively, we attempted the chemical synthesis of benzothiazole phosphonate derivative 2. Instead of a one-pot synthesis, we report a two-step synthesis with characterization of both imine intermediate 5 and final compound 2. Furthermore, complete assignation of 1H and 13C nuclear magnetic resonance (NMR) signals of 2 is provided, as we observed a divergence of NMR data with those published previously. Finally, biological assays showed that 1 and 2 inhibited the oxidation of estradiol (E2) into estrone (E1) by the 17β-HSD10 recombinant protein. However, in human embryonic kidney (HEK)-293 intact cells transfected with 17β-HSD10, only the steroidal inhibitor 1 induced a dose-dependent inhibition of E2 to E1 transformation.
Collapse
|