Neurons and Neuronal Activity Control Gene Expression in Astrocytes to Regulate Their Development and Metabolism

Total Page:16

File Type:pdf, Size:1020Kb

Neurons and Neuronal Activity Control Gene Expression in Astrocytes to Regulate Their Development and Metabolism ARTICLE Received 29 Jul 2016 | Accepted 2 Mar 2017 | Published 2 May 2017 DOI: 10.1038/ncomms15132 OPEN Neurons and neuronal activity control gene expression in astrocytes to regulate their development and metabolism Philip Hasel1, Owen Dando1,2,3,*, Zoeb Jiwaji1,2,*, Paul Baxter1, Alison C. Todd1, Samuel Heron4,No´ra M. Ma´rkus1, Jamie McQueen1, David W. Hampton2, Megan Torvell2, Sachin S. Tiwari2, Sean McKay1, Abel Eraso-Pichot5, Antonio Zorzano6,7,8, Roser Masgrau5, Elena Galea5,9, Siddharthan Chandran2,3, David J.A. Wyllie1, T. Ian Simpson4 & Giles E. Hardingham1,10 The influence that neurons exert on astrocytic function is poorly understood. To investigate this, we first developed a system combining cortical neurons and astrocytes from closely related species, followed by RNA-seq and in silico species separation. This approach uncovers a wide programme of neuron-induced astrocytic gene expression, involving Notch signalling, which drives and maintains astrocytic maturity and neurotransmitter uptake function, is conserved in human development, and is disrupted by neurodegeneration. Separately, hundreds of astrocytic genes are acutely regulated by synaptic activity via mechanisms involving cAMP/PKA-dependent CREB activation. This includes the coordinated activity- dependent upregulation of major astrocytic components of the astrocyte–neuron lactate shuttle, leading to a CREB-dependent increase in astrocytic glucose metabolism and elevated lactate export. Moreover, the groups of astrocytic genes induced by neurons or neuronal activity both show age-dependent decline in humans. Thus, neurons and neuronal activity regulate the astrocytic transcriptome with the potential to shape astrocyte–neuron metabolic cooperation. 1 Deanery of Biomedical Sciences, Edinburgh Medical School, University of Edinburgh, Edinburgh EH8 9XD, UK. 2 MRC Centre for Regenerative Medicine, University of Edinburgh, Edinburgh EH16 4SB, UK. 3 Centre for Brain Development and Repair, Institute for Stem Cell Biology and Regenerative Medicine, National Centre for Biological Sciences, Bangalore 560065, India. 4 School of Informatics, University of Edinburgh, Edinburgh EH8 9AB, UK. 5 Institut de Neurocie`ncies and Departament de Bioquı´mica i Biologia Molecular, Unitat de Bioquı´mica de Medicina, Edifici M, Universitat Auto`noma de Barcelona, Bellaterra, Barcelona 08193, Spain. 6 Institute for Research in Biomedicine, Barcelona 08028, Spain. 7 Department of Biochemistry and Molecular Biology, University of Barcelona, Barcelona 08028, Spain. 8 Centro de Investigacio´n Biome´dica en Red de Diabetes y Enfermedades Metabo´licas Asociadas (CIBERDEM), Instituto de Salud Carlos III (ISCIII), Madrid 28029, Spain. 9 Institucio´ Catalana De Recerca I Estudis Avanc¸ats (ICREA), Passeig Lluı´s Companys 23, Barcelona, Catalonia, 08010, Spain. 10 UK Dementia Research Institute at The University of Edinburgh, Edinburgh Medical School, 47 Little France Crescent, Edinburgh EH16 4TJ, UK. * These authors contributed equally to this work. Correspondence and requests for materials should be addressed to G.E.H. (email: [email protected]). NATURE COMMUNICATIONS | 8:15132 | DOI: 10.1038/ncomms15132 | www.nature.com/naturecommunications 1 ARTICLE NATURE COMMUNICATIONS | DOI: 10.1038/ncomms15132 ignalling between neurons triggers programmes of gene another marker (Aldh1l1, 99.4±0.2% positive (n ¼ 4), Suppleme- expression that mediate specific functions in development ntary Fig. 2a). The astrocytes were o0.1% Neuro-Chrom þ Sand maturity. Signalling between neurons and glia is also neurons and o0.1% Iba þ microglia. implicated in diverse processes, however, insight into the scope Compared to parallel astrocytic mono-cultures, co-cultured and mechanism by which different cell types in the neuro-glial astrocytes developed the expected stellate morphology over 9 days unit influence each other is lacking. A notable example is neuron- (Fig. 1a,b). At this point, RNA was extracted (3 biological to-astrocyte signalling: neurons induce the classic stellate replicates), RNA-seq performed and SSS employed to identify the morphology in astrocytes, resembling their appearance in vivo1. mouse (that is, astrocyte) reads. DGE analysis revealed wide- However, knowledge of the transcriptional changes that spread up- and down-regulation of astrocytic gene expression due accompany this transformation are restricted to three to the presence of neurons (Fig. 1c; Supplementary Data 5), astrocytic genes (Slc1a2, Slc1a3 and Gja1 (refs 2,3)) induced by including previously identified genes (Slc1a2, Slc1a3 and Gja1 neurons. Similarly, despite astrocytes playing a critical role in the (refs 2,3)). Four hundred and ten genes were induced more than uptake of synaptically released neurotransmitters, whether twofold and 353 genes repressed more than twofold (Adjusted P synaptic activity controls astrocytic gene expression is unclear. value o0.05, Supplementary Data 5). This contrasts with the wealth of knowledge concerning the A notable cluster of upregulated genes is involved in the mechanisms and the roles of activity-dependent gene expression uptake and metabolism of neurotransmitters, including in neurons4–7. GABA (Slc6a1, Slc6a11 and Abat), NAAG (Slc17a5), biogenic Understanding non-cell-autonomously regulated gene expres- amine neurotransmitters (Slc29a2 and Maob) and glutamate sion poses challenges associated with the physical separation of (Slc1a2, Slc1a3, Glul and Glud1; Supplementary Data 6). cell types before transcriptome analysis by methods including Focussing on glutamate uptake, we confirmed that these RNA-seq. Immunopanning or FACS are powerful approaches, transcriptional changes are reflected at the functional level, by but are subject to off-target cell type contamination, and cause measuring the electrogenic currents of the glutamate transporters aberrant induction of transcriptional stress responses, or the loss EAAT1/GLAST (Slc1a3), and EAAT2/GLT-1 (Slc1a2), using of material from delicate subcellular regions8–10. Theoretically, if whole-cell voltage clamp recordings. Astrocytic EAAT currents14 RNA-seq reads could be unambiguously attributed to one cell were increased around 10-fold by neuronal co-culture (Fig. 1d,e), type or another, physical sorting could be avoided. We while astrocytic resting membrane potential and passive memb- investigated the possibility of achieving this by deriving the rane conductance were unaffected (Supplementary Fig. 2b,c)15,16. distinct cell types from different species11. We chose rat and In addition, many cytoskeletal and extracellular matrix genes mouse, two species that we reasoned may be closely enough were altered (Supplementary Data 5), consistent with the related to enable questions of non-cell-autonomous signalling to profound changes in astrocytic morphology. Astrocyte-specific be answered. We used this system as a starting point to marker and endfoot component Aqp4 was also induced, but other investigate how and to what extent neurons and neuronal markers, S100b, Aldh1l1 and Gfap were unaffected (Fig. 1c). Thus, activity control astrocytic gene expression, as well as the neuron-induced changes to the morphology of astrocytes are underlying mechanisms and functional consequences. associated with marked changes to their transcriptome and functional properties. Results To assess the advantages of this approach over imperfect physical separation approaches, we firstly simulated a sorting Unsorting rat and mouse RNA-seq reads in silico. We first established the feasibility of our species-specific sorting (SSS) process that achieved 95% purity, by ‘contaminating’ the mouse astrocytic mRNA with mouse cortical neuronal mRNA at a ratio RNA-seq workflow. In the vast majority of genes, the majority of mouse RNA-seq paired-end reads can be unambiguously attributed of 95:5, by number of cells collected, and performed RNA-seq. The effects were substantial: 863 genes were expressed more than to mouse (as opposed to rat), using both a simulated set of reads (Supplementary Fig. 1a; Supplementary Data 1) as well as two-fold higher in the 95% sample, compared to the pure astrocytic sample (Supplementary Fig. 3a, Supplementary Data 7), a ‘real-world’ RNA-seq data set of mouse cortical neurons 4 (Supplementary Fig. 1b; Supplementary Data 2). Also, compared to of which 216 were expressed 10-fold higher. Second, we investigated the induction of transcriptional stress responses to normal read mapping, the SSS workflow does not substantially 9 affect differential gene expression analysis (DGE) when performed physical sorting methods . Both simple trypsinization, immunopanning and FACS protocols applied to a on a single-species (mouse) RNA-seq data-set taken from neurons treated±bicuculline þ 4-aminopyridine (BiC/4-AP) to induce homogeneous population of cultured astrocytes induced multiple immediate early genes (Supplementary Fig. 3b–d). synaptic activity via network disinhibition12:weobservedatight correlation in fold-induction, comparing normal and SSS These data show key advantages of in silico read separation in avoiding DGE artefacts introduced by physical sorting. approaches (Supplementary Fig. 1c; Supplementary Data 3). There was also little change in the adjusted P values: of the 4,632 genes altered by BiC/4-AP treatment, only 33 were rendered Neurons induce an in vivo-like transcriptome in astrocytes insignificant by the SSS approach (Supplementary Fig. 1d; . While neuronal co-culture promotes a more in vivo-like Supplementary Data 4). Large P value deviations were more
Recommended publications
  • Deep Learning–Based Multi-Omics Integration Robustly Predicts Survival in Liver Cancer Kumardeep Chaudhary1, Olivier B
    Published OnlineFirst October 5, 2017; DOI: 10.1158/1078-0432.CCR-17-0853 Statistics in CCR Clinical Cancer Research Deep Learning–Based Multi-Omics Integration Robustly Predicts Survival in Liver Cancer Kumardeep Chaudhary1, Olivier B. Poirion1, Liangqun Lu1,2, and Lana X. Garmire1,2 Abstract Identifying robust survival subgroups of hepatocellular car- index (C-index) ¼ 0.68]. More aggressive subtype is associated cinoma (HCC) will significantly improve patient care. Current- with frequent TP53 inactivation mutations, higher expression ly, endeavor of integrating multi-omicsdatatoexplicitlypredict of stemness markers (KRT19 and EPCAM)andtumormarker HCC survival from multiple patient cohorts is lacking. To fill BIRC5, and activated Wnt and Akt signaling pathways. We this gap, we present a deep learning (DL)–based model on HCC validated this multi-omics model on five external datasets of that robustly differentiates survival subpopulations of patients various omics types: LIRI-JP cohort (n ¼ 230, C-index ¼ 0.75), in six cohorts. We built the DL-based, survival-sensitive model NCI cohort (n ¼ 221, C-index ¼ 0.67), Chinese cohort (n ¼ on 360 HCC patients' data using RNA sequencing (RNA-Seq), 166, C-index ¼ 0.69), E-TABM-36 cohort (n ¼ 40, C-index ¼ miRNA sequencing (miRNA-Seq), and methylation data from 0.77), and Hawaiian cohort (n ¼ 27, C-index ¼ 0.82). This TheCancerGenomeAtlas(TCGA),whichpredictsprognosis is the first study to employ DL to identify multi-omics features as good as an alternative model where genomics and clinical linked to the differential survival of patients with HCC. Given data are both considered. This DL-based model provides two its robustness over multiple cohorts, we expect this workflow to optimal subgroups of patients with significant survival differ- be useful at predicting HCC prognosis prediction.
    [Show full text]
  • Upregulation of Peroxisome Proliferator-Activated Receptor-Α And
    Upregulation of peroxisome proliferator-activated receptor-α and the lipid metabolism pathway promotes carcinogenesis of ampullary cancer Chih-Yang Wang, Ying-Jui Chao, Yi-Ling Chen, Tzu-Wen Wang, Nam Nhut Phan, Hui-Ping Hsu, Yan-Shen Shan, Ming-Derg Lai 1 Supplementary Table 1. Demographics and clinical outcomes of five patients with ampullary cancer Time of Tumor Time to Age Differentia survival/ Sex Staging size Morphology Recurrence recurrence Condition (years) tion expired (cm) (months) (months) T2N0, 51 F 211 Polypoid Unknown No -- Survived 193 stage Ib T2N0, 2.41.5 58 F Mixed Good Yes 14 Expired 17 stage Ib 0.6 T3N0, 4.53.5 68 M Polypoid Good No -- Survived 162 stage IIA 1.2 T3N0, 66 M 110.8 Ulcerative Good Yes 64 Expired 227 stage IIA T3N0, 60 M 21.81 Mixed Moderate Yes 5.6 Expired 16.7 stage IIA 2 Supplementary Table 2. Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analysis of an ampullary cancer microarray using the Database for Annotation, Visualization and Integrated Discovery (DAVID). This table contains only pathways with p values that ranged 0.0001~0.05. KEGG Pathway p value Genes Pentose and 1.50E-04 UGT1A6, CRYL1, UGT1A8, AKR1B1, UGT2B11, UGT2A3, glucuronate UGT2B10, UGT2B7, XYLB interconversions Drug metabolism 1.63E-04 CYP3A4, XDH, UGT1A6, CYP3A5, CES2, CYP3A7, UGT1A8, NAT2, UGT2B11, DPYD, UGT2A3, UGT2B10, UGT2B7 Maturity-onset 2.43E-04 HNF1A, HNF4A, SLC2A2, PKLR, NEUROD1, HNF4G, diabetes of the PDX1, NR5A2, NKX2-2 young Starch and sucrose 6.03E-04 GBA3, UGT1A6, G6PC, UGT1A8, ENPP3, MGAM, SI, metabolism
    [Show full text]
  • Small-Molecule Inhibition of 6-Phosphofructo-2-Kinase Activity Suppresses Glycolytic Flux and Tumor Growth
    110 Small-molecule inhibition of 6-phosphofructo-2-kinase activity suppresses glycolytic flux and tumor growth Brian Clem,1,3 Sucheta Telang,1,3 Amy Clem,1,3 reduces the intracellular concentration of Fru-2,6-BP, Abdullah Yalcin,1,2,3 Jason Meier,2 glucose uptake, and growth of established tumors in vivo. Alan Simmons,1,3 Mary Ann Rasku,1,3 Taken together, these data support the clinical development Sengodagounder Arumugam,1,3 of 3PO and other PFKFB3 inhibitors as chemotherapeutic William L. Dean,2,3 John Eaton,1,3 Andrew Lane,1,3 agents. [Mol Cancer Ther 2008;7(1):110–20] John O. Trent,1,2,3 and Jason Chesney1,2,3 Departments of 1Medicine and 2Biochemistry and Molecular Introduction Biology and 3Molecular Targets Group, James Graham Brown Neoplastic transformation causes a marked increase in Cancer Center, University of Louisville, Louisville, Kentucky glucose uptake and catabolic conversion to lactate, which forms the basis for the most specific cancer diagnostic 18 Abstract examination—positron emission tomography of 2- F- fluoro-2-deoxyglucose (18F-2-DG) uptake (1). The protein 6-Phosphofructo-1-kinase, a rate-limiting enzyme of products of several oncogenes directly increase glycolytic glycolysis, is activated in neoplastic cells by fructose-2,6- flux even under normoxic conditions, a phenomenon bisphosphate (Fru-2,6-BP), a product of four 6-phospho- originally termed the Warburg effect (2, 3). For example, fructo-2-kinase/fructose-2,6-bisphosphatase isozymes c-myc is a transcription factor that promotes the expression (PFKFB1-4). The inducible PFKFB3 isozyme is constitu- of glycolytic enzyme mRNAs, and its expression is increased tively expressed by neoplastic cells and required for the in several human cancers regardless of the oxygen pressure high glycolytic rate and anchorage-independent growth of (4, 5).
    [Show full text]
  • Open Full Page
    Published OnlineFirst February 12, 2018; DOI: 10.1158/0008-5472.CAN-17-2215 Cancer Metabolism and Chemical Biology Research RSK Regulates PFK-2 Activity to Promote Metabolic Rewiring in Melanoma Thibault Houles1, Simon-Pierre Gravel2,Genevieve Lavoie1, Sejeong Shin3, Mathilde Savall1, Antoine Meant 1, Benoit Grondin1, Louis Gaboury1,4, Sang-Oh Yoon3, Julie St-Pierre2, and Philippe P. Roux1,4 Abstract Metabolic reprogramming is a hallmark of cancer that includes glycolytic flux in melanoma cells, suggesting an important role for increased glucose uptake and accelerated aerobic glycolysis. This RSK in BRAF-mediated metabolic rewiring. Consistent with this, phenotypeisrequiredtofulfill anabolic demands associated with expression of a phosphorylation-deficient mutant of PFKFB2 aberrant cell proliferation and is often mediated by oncogenic decreased aerobic glycolysis and reduced the growth of melanoma drivers such as activated BRAF. In this study, we show that the in mice. Together, these results indicate that RSK-mediated phos- MAPK-activated p90 ribosomal S6 kinase (RSK) is necessary to phorylation of PFKFB2 plays a key role in the metabolism and maintain glycolytic metabolism in BRAF-mutated melanoma growth of BRAF-mutated melanomas. cells. RSK directly phosphorylated the regulatory domain of Significance: RSK promotes glycolytic metabolism and the 6-phosphofructo-2-kinase/fructose-2,6-bisphosphatase 2 (PFKFB2), growth of BRAF-mutated melanoma by driving phosphory- an enzyme that catalyzes the synthesis of fructose-2,6-bisphosphate lation of an important glycolytic enzyme. Cancer Res; 78(9); during glycolysis. Inhibition of RSK reduced PFKFB2 activity and 2191–204. Ó2018 AACR. Introduction but recently developed therapies that target components of the MAPK pathway have demonstrated survival advantage in pati- Melanoma is the most aggressive form of skin cancer and arises ents with BRAF-mutated tumors (7).
    [Show full text]
  • Volatile and Intravenous Anesthesia Alter Rat Liver Proteins: Proteomic Time Course Analysis of Rat Liver Proteins
    8 The Open Proteomics Journal, 2012, 5, 8-16 Open Access Volatile and Intravenous Anesthesia Alter Rat Liver Proteins: Proteomic Time Course Analysis of Rat Liver Proteins Hisashi Watanabe1,*, Chihiro Kamagata2, Yoshiaki Tsuboko2 and Atsuhiro Sakamoto3 1Graduate Student, Department of Anesthesiology and Pain Medicine, Graduate School of Medicine, Nippon Medical School, Tokyo, Japan 2Staff Member, Department of Anesthesiology and Pain Medicine, Graduate School of Medicine, Nippon Medical School, Tokyo, Japan 3Professor and Chair, Department of Anesthesiology and Pain Medicine, Graduate School of Medicine, Nippon Medical School, Tokyo, Japan Abstract: Background: Our previous microarray study showed that sevoflurane anesthesia affects the expression of rat genes in multiple organs including the liver. In this study, we investigated whether liver protein expression was altered after propofol, sevoflurane, or isoflurane anesthesia. We also investigated differences in the time course of each drug 24 and 72 h after anesthesia. Methods: Rats were randomly assigned to four groups (non-anesthetized group and three groups anesthetized at each time point, n = 6 per group). A venous catheter was inserted into the caudal vein of all rats. Rats were anesthetized with each agent for 6 h, and the liver was obtained immediately after anesthesia. Proteomic analysis was performed. Results: About 4200 spots in each gel were discriminated, and at least 2619 spots were matched. Using LC-MS/MS, we identified 47 spots for propofol, 45 spots for sevoflurane, and 21 spots for isoflurane that were differentially expressed (p < 0.05) 0 h after anesthesia. The numbers of altered proteins were 14 and 19 in the isoflurane and sevoflurane groups, respectively, 72 h after anesthesia, but alterations in 40 proteins were seen in the propofol group 72 h after anesthesia.
    [Show full text]
  • Identify Distinct Prognostic Impact of ALDH1 Family Members by TCGA Database in Acute Myeloid Leukemia
    Open Access Annals of Hematology & Oncology Research Article Identify Distinct Prognostic Impact of ALDH1 Family Members by TCGA Database in Acute Myeloid Leukemia Yi H, Deng R, Fan F, Sun H, He G, Lai S and Su Y* Department of Hematology, General Hospital of Chengdu Abstract Military Region, China Background: Acute myeloid leukemia is a heterogeneous disease. Identify *Corresponding author: Su Y, Department of the prognostic biomarker is important to guide stratification and therapeutic Hematology, General Hospital of Chengdu Military strategies. Region, Chengdu, 610083, China Method: We detected the expression level and the prognostic impact of Received: November 25, 2017; Accepted: January 18, each ALDH1 family members in AML by The Cancer Genome Atlas (TCGA) 2018; Published: February 06, 2018 database. Results: Upon 168 patients whose expression level of ALDH1 family members were available. We found that the level of ALDH1A1correlated to the prognosis of AML by the National Comprehensive Cancer Network (NCCN) stratification but not in other ALDH1 members. Moreover, we got survival data from 160 AML patients in TCGA database. We found that high ALDH1A1 expression correlated to poor Overall Survival (OS), mostly in Fms-like Tyrosine Kinase-3 (FLT3) mutated group. HighALDH1A2 expression significantly correlated to poor OS in FLT3 wild type population but not in FLT3 mutated group. High ALDH1A3 expression significantly correlated to poor OS in FLT3 mutated group but not in FLT3 wild type group. There was no relationship between the OS of AML with the level of ALDH1B1, ALDH1L1 and ALDH1L2. Conclusion: The prognostic impacts were different in each ALDH1 family members, which needs further investigation.
    [Show full text]
  • Overall Survival of Pancreatic Ductal Adenocarcinoma Is Doubled by Aldh7a1 Deletion in the KPC Mouse
    Overall survival of pancreatic ductal adenocarcinoma is doubled by Aldh7a1 deletion in the KPC mouse Jae-Seon Lee1,2*, Ho Lee3*, Sang Myung Woo4, Hyonchol Jang1, Yoon Jeon1, Hee Yeon Kim1, Jaewhan Song2, Woo Jin Lee4, Eun Kyung Hong5, Sang-Jae Park4, Sung- Sik Han4§§ and Soo-Youl Kim1§ 1Division of Cancer Biology, Research Institute, National Cancer Center, Goyang, Republic of Korea. 2Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul, Republic of Korea. 3Graduate School of Cancer Science and Policy, 4Department of Surgery, Center for Liver and Pancreatobiliary Cancer and 5Department of Pathology, National Cancer Center, Goyang, Republic of Korea. Correspondence §Corresponding author: [email protected] (S.-Y.K.) §§Co-corresponding author: [email protected] (S.-S.H.) *These authors contributed equally to this work 1 Abstract Rationale: The activity of aldehyde dehydrogenase 7A1 (ALDH7A1), an enzyme that catalyzes the lipid peroxidation of fatty aldehydes was found to be upregulated in pancreatic ductal adenocarcinoma (PDAC). ALDH7A1 knockdown significantly reduced tumor formation in PDAC. We raised a question how ALDH7A1 contributes to cancer progression. Methods: To answer the question, the role of ALDH7A1 in energy metabolism was investigated by knocking down and knockdown gene in mouse model, because the role of ALDH7A1 has been reported as a catabolic enzyme catalyzing fatty aldehyde from lipid peroxidation to fatty acid. Oxygen consumption rate (OCR), ATP production, mitochondrial membrane potential, proliferation assay and immunoblotting were performed. In in vivo study, two human PDAC cell lines were used for pre-clinical xenograft model as well as spontaneous PDAC model of KPC mice was also employed for anti-cancer therapeutic effect.
    [Show full text]
  • NIH Public Access Author Manuscript Expert Opin Drug Metab Toxicol
    NIH Public Access Author Manuscript Expert Opin Drug Metab Toxicol. Author manuscript; available in PMC 2009 March 19. NIH-PA Author ManuscriptPublished NIH-PA Author Manuscript in final edited NIH-PA Author Manuscript form as: Expert Opin Drug Metab Toxicol. 2008 June ; 4(6): 697±720. doi:10.1517/17425250802102627. Non-P450 aldehyde oxidizing enzymes: the aldehyde dehydrogenase superfamily Satori A Marchitti, Chad Brocker*, Dimitrios Stagos*, and Vasilis Vasiliou† University of Colorado Health Sciences Center, Molecular Toxicology & Environmental Health Sciences Program, Department of Pharmaceutical Sciences, Denver, Colorado 80262, USA Abstract Background—Aldehydes are highly reactive molecules. While several non-P450 enzyme systems participate in their metabolism, one of the most important is the aldehyde dehydrogenase (ALDH) superfamily, composed of NAD(P)+-dependent enzymes that catalyze aldehyde oxidation. Objective—This article presents a review of what is currently known about each member of the human ALDH superfamily including the pathophysiological significance of these enzymes. Methods—Relevant literature involving all members of the human ALDH family was extensively reviewed, with the primary focus on recent and novel findings. Conclusion—To date, 19 ALDH genes have been identified in the human genome and mutations in these genes and subsequent inborn errors in aldehyde metabolism are the molecular basis of several diseases, including Sjögren-Larsson syndrome, type II hyperprolinemia, γ-hydroxybutyric aciduria and pyridoxine-dependent seizures. ALDH enzymes also play important roles in embryogenesis and development, neurotransmission, oxidative stress and cancer. Finally, ALDH enzymes display multiple catalytic and non-catalytic functions including ester hydrolysis, antioxidant properties, xenobiotic bioactivation and UV light absorption. Keywords aldehyde dehydrogenase; aldehyde metabolism; ALDH 1.
    [Show full text]
  • Chuanxiong Rhizoma Compound on HIF-VEGF Pathway and Cerebral Ischemia-Reperfusion Injury’S Biological Network Based on Systematic Pharmacology
    ORIGINAL RESEARCH published: 25 June 2021 doi: 10.3389/fphar.2021.601846 Exploring the Regulatory Mechanism of Hedysarum Multijugum Maxim.-Chuanxiong Rhizoma Compound on HIF-VEGF Pathway and Cerebral Ischemia-Reperfusion Injury’s Biological Network Based on Systematic Pharmacology Kailin Yang 1†, Liuting Zeng 1†, Anqi Ge 2†, Yi Chen 1†, Shanshan Wang 1†, Xiaofei Zhu 1,3† and Jinwen Ge 1,4* Edited by: 1 Takashi Sato, Key Laboratory of Hunan Province for Integrated Traditional Chinese and Western Medicine on Prevention and Treatment of 2 Tokyo University of Pharmacy and Life Cardio-Cerebral Diseases, Hunan University of Chinese Medicine, Changsha, China, Galactophore Department, The First 3 Sciences, Japan Hospital of Hunan University of Chinese Medicine, Changsha, China, School of Graduate, Central South University, Changsha, China, 4Shaoyang University, Shaoyang, China Reviewed by: Hui Zhao, Capital Medical University, China Background: Clinical research found that Hedysarum Multijugum Maxim.-Chuanxiong Maria Luisa Del Moral, fi University of Jaén, Spain Rhizoma Compound (HCC) has de nite curative effect on cerebral ischemic diseases, *Correspondence: such as ischemic stroke and cerebral ischemia-reperfusion injury (CIR). However, its Jinwen Ge mechanism for treating cerebral ischemia is still not fully explained. [email protected] †These authors share first authorship Methods: The traditional Chinese medicine related database were utilized to obtain the components of HCC. The Pharmmapper were used to predict HCC’s potential targets. Specialty section: The CIR genes were obtained from Genecards and OMIM and the protein-protein This article was submitted to interaction (PPI) data of HCC’s targets and IS genes were obtained from String Ethnopharmacology, a section of the journal database.
    [Show full text]
  • TPI1 Gene Triosephosphate Isomerase 1
    TPI1 gene triosephosphate isomerase 1 Normal Function The TPI1 gene provides instructions for making an enzyme called triosephosphate isomerase 1. This enzyme is involved in a critical energy-producing process known as glycolysis. During glycolysis, the simple sugar glucose is broken down to produce energy for cells. The triosephosphate isomerase 1 enzyme carries out a specific reaction during glycolysis: the conversion of a molecule called dihydroxyacetone phosphate (DHAP) to glyceraldehyde 3-phosphate. This conversion can go both ways, meaning that the triosephosphate isomerase 1 enzyme can also convert glyceraldehyde 3-phosphate back into DHAP. Additional steps convert glyceraldehyde 3-phosphate into other molecules that ultimately produce energy in the form of a molecule called ATP. For the triosephosphate isomerase 1 enzyme to be turned on (active), it has to attach ( bind) to another triosephosphate isomerase 1 enzyme, forming a two-enzyme complex called a dimer. Health Conditions Related to Genetic Changes Triosephosphate isomerase deficiency At least 12 mutations in the TPI1 gene have been found to cause triosephosphate isomerase deficiency. This condition is characterized by a shortage of red blood cells ( anemia), movement problems, increased susceptibility to infection, and muscle weakness that can affect breathing and heart function. TPI1 gene mutations can lead to the production of an enzyme with decreased activity. As a result, glycolysis is impaired and cells have a decreased supply of energy. One TPI1 gene mutation accounts for approximately 80 percent of triosephosphate isomerase deficiency cases. This change replaces the protein building block (amino acid) glutamic acid with the amino acid aspartic acid at position 104 in the triosephosphate isomerase 1 enzyme (written as Glu104Asp or E104D).
    [Show full text]
  • Exploring the Non-Canonical Functions of Metabolic Enzymes Peiwei Huangyang1,2 and M
    © 2018. Published by The Company of Biologists Ltd | Disease Models & Mechanisms (2018) 11, dmm033365. doi:10.1242/dmm.033365 REVIEW SPECIAL COLLECTION: CANCER METABOLISM Hidden features: exploring the non-canonical functions of metabolic enzymes Peiwei Huangyang1,2 and M. Celeste Simon1,3,* ABSTRACT A key finding from studies of metabolic enzymes is the existence The study of cellular metabolism has been rigorously revisited over the of mechanistic links between their nuclear localization and the past decade, especially in the field of cancer research, revealing new regulation of transcription. By modulating gene expression, insights that expand our understanding of malignancy. Among these metabolic enzymes themselves facilitate adaptation to rapidly insights isthe discovery that various metabolic enzymes have surprising changing environments. Furthermore, they can directly shape a ’ activities outside of their established metabolic roles, including in cell s epigenetic landscape (Kaelin and McKnight, 2013). the regulation of gene expression, DNA damage repair, cell cycle Strikingly, several metabolic enzymes exert completely distinct progression and apoptosis. Many of these newly identified functions are functions in different cellular compartments. Nuclear fructose activated in response to growth factor signaling, nutrient and oxygen bisphosphate aldolase, for example, directly interacts with RNA ́ availability, and external stress. As such, multifaceted enzymes directly polymerase III to control transcription (Ciesla et al., 2014),
    [Show full text]
  • Oxidative Stress Activates SIRT2 to Deacetylate and Stimulate Phosphoglycerate Mutase
    Published OnlineFirst May 1, 2014; DOI: 10.1158/0008-5472.CAN-13-3615 Cancer Tumor and Stem Cell Biology Research Oxidative Stress Activates SIRT2 to Deacetylate and Stimulate Phosphoglycerate Mutase Yanping Xu2,3, Fulong Li2,3, Lei Lv1,2, Tingting Li2,3, Xin Zhou2,3, Chu-Xia Deng4, Kun-Liang Guan1,2,5, Qun-Ying Lei1,2, and Yue Xiong1,2,6 Abstract Glycolytic enzyme phosphoglycerate mutase (PGAM) plays an important role in coordinating energy production with generation of reducing power and the biosynthesis of nucleotide precursors and amino acids. Inhibition of PGAM by small RNAi or small molecule attenuates cell proliferation and tumor growth. PGAM activity is commonly upregulated in tumor cells, but how PGAM activity is regulated in vivo remains poorly understood. Here we report that PGAM is acetylated at lysine 100 (K100), an active site residue that is invariably conserved from bacteria, to yeast, plant, and mammals. K100 acetylation is detected in fly, mouse, and human cells and in multiple tissues and decreases PGAM2 activity. The cytosolic protein deacetylase sirtuin 2 (SIRT2) deacetylates and activates PGAM2. Increased levels of reactive oxygen species stimulate PGAM2 deacetylation and activity by promoting its interaction with SIRT2. Substitution of endogenous PGAM2 with an acetylation mimetic mutant K100Q reduces cellular NADPH production and inhibits cell proliferation and tumor growth. These results reveal a mechanism of PGAM2 regulation and NADPH homeostasis in response to oxidative stress that impacts cell proliferation and tumor growth. Cancer Res; 74(13); 1–13. Ó2014 AACR. Introduction contains two PGAM genes, PGAM1 (also known as PGAM-B), Enhanced glycolysis, commonly referred to as the "Warburg which is expressed in brain and most other tissues, and PGAM2 effect" (1), is a distinctive and prominent feature of cancer cells.
    [Show full text]