Influence of Prenatal Transportation Stress-Induced Differential DNA Methylation on the Physiological Control of Behavior and St

Total Page:16

File Type:pdf, Size:1020Kb

Influence of Prenatal Transportation Stress-Induced Differential DNA Methylation on the Physiological Control of Behavior and St Copyedited by: KD Journal of Animal Science, 2020, 1–18 doi:10.1093/jas/skz368 Advance Access publication December 6, 2019 Received: 31 August 2019 and Accepted: 5 December 2019 Fetal Programming Downloaded from https://academic.oup.com/jas/article-abstract/98/1/skz368/5661111 by Texas A&M College Station user on 30 April 2020 Fetal Programming Influence of prenatal transportation stress-induced differential DNA methylation on the physiological control of behavior and stress response in suckling Brahman bull calves Brittni P. Littlejohn,†,‡,1 Deborah M. Price,†,‡ Don A. Neuendorff,† Jeffery A. Carroll,|| Rhonda C. Vann,$ Penny K. Riggs,‡ David G. Riley,‡ Charles R. Long,†,‡ Ronald D. Randel,† and Thomas H. Welsh, Jr.‡,2 †Texas A&M AgriLife Research & Extension Center, Overton, TX 75684, ‡Department of Animal Science, Texas A&M University, and Texas A&M AgriLife Research, College Station, TX 77843, ||USDA-ARS, Livestock Issues Research Unit, Lubbock, TX 79403, $Mississippi Agricultural and Forestry Experiment Station, Mississippi State University, Raymond, MS 39154 1Current address: Department of Animal and Dairy Sciences, Mississippi State University, Mississippi State, MS 39762 2Corresponding author: [email protected] Abstract The objective of this experiment was to examine potential differential methylation of DNA as a mechanism for altered behavioral and stress responses in prenatally stressed (PNS) compared with nonprenatally stressed (Control) young bull calves. Mature Brahman cows (n = 48) were transported for 2-h periods at 60 ± 5, 80 ± 5, 100 ± 5, 120 ± 5, and 140 ± 5 d of gestation (Transported group) or maintained as nontransported Controls (n = 48). From the offspring born to Transported and Control cows, a subset of 28-d-old intact bulls (n = 7 PNS; n = 7 Control) were evaluated for methylation of DNA of behavior and stress response–associated genes. Methylation of DNA from white blood cells was assessed via reduced representation bisulfite sequencing methods. Because increased methylation of DNA within gene promoter regions has been associated with decreased transcriptional activity of the corresponding gene, differentially methylated (P ≤ 0.05) CG sites (cytosine followed by a guanine nucleotide) located within promoter regions (n = 1,205) were used to predict (using Ingenuity Pathway Analysis software) alterations to canonical pathways in PNS compared with Control bull calves. Among differentially methylated genes (P ≤ 0.05) related to behavior and the stress response were OPRK1, OPRM1, PENK, POMC, NR3C2, TH, DRD1, DRD5, COMT, HTR6, HTR5A, GABRA4, GABRQ, and GAD2. Among altered (P < 0.05) signaling pathways related to behavior and the stress response were Opioid Signaling, Corticotropin-Releasing Hormone Signaling, Dopamine Receptor Signaling, Dopamine-DARPP32 Feedback in cAMP Signaling, Serotonin Receptor Signaling, and GABA Receptor Signaling. Alterations to behavior and stress response–related genes and canonical pathways supported previously observed elevations in temperament score and serum cortisol through weaning in the larger population of PNS calves from which bulls in this study were derived. Differential methylation of DNA and predicted alterations to behavior and stress response–related pathways in PNS compared with Control bull calves suggest epigenetic programming of behavior and the stress response in utero. Key words: behavior, bovine, epigenetics, prenatal stress, stress response, temperament © The Author(s) 2019. Published by Oxford University Press on behalf of the American Society of Animal Science. All rights reserved. For permissions, please e-mail: [email protected]. 1 Copyedited by: KD 2 | Journal of Animal Science, 2020, Vol. 98, No. 1 Introduction Animal Procedures Alteration of behavior and stress responsiveness is among the Mature Brahman cows were assigned to 1 of 2 treatment most noted outcomes of prenatal stress (PNS) in mammals groups (Transported: n = 48 and Control: n = 48), which were (Lindsay et al., 2019; Matthews and McGowan, 2019). The balanced by age, parity, and temperament (Littlejohn et al., mechanisms controlling these alterations are still largely 2016). Transported cows were hauled for 2 h at 60 ± 5, 80 ± 5, unclear; however, it is known that stress incurred by a gestating 100 ± 5, 120 ± 5, and 140 ± 5 d of gestation (Price et al., 2015). dam can result in suppression of the placental barrier enzyme Control cows were managed the same as Transported cows 11β-hydroxysteroid dehydrogenase type 2 (11-βHSD2). This with the exception of being transported. Both treatment groups Downloaded from https://academic.oup.com/jas/article-abstract/98/1/skz368/5661111 by Texas A&M College Station user on 30 April 2020 enzyme converts cortisol to an inactive form prior to entering were housed in the same pasture (at the Texas A&M AgriLife fetal circulation; therefore, decreased 11-βHSD2 can result Research and Extension Center at Overton, TX) and fed the in increased fetal exposure to corticosteroids (Benediktsson same diet (Littlejohn et al., 2016). From these groups, 26 male et al., 1997; Stirrat et al., 2018). Such alterations to the fetal and 18 female calves (Control group) were born to Control dams, environment may result in epigenetic modifications, such whereas 20 male and 21 female calves (Prenatally Stressed, as DNA methylation, which appear to be major regulators of PNS, group) were born to Transported dams. Male calves phenotypic changes due to prenatal stress (Szyf, 2012; Lester were maintained as bulls throughout the study. Each calf was and Marist, 2018). Deoxyribonucleic acid methylation is defined manually restrained for less than 5 min for sample collection as a covalent modification in which a methyl group is added at 28 d of age. One 10-mL vacuum tube (BD, Franklin Lakes, NJ) to the carbon 5 position of a cytosine nucleotide (Wyatt, 1950; containing EDTA was used to collect the blood sample from each Razin and Riggs, 1980). Increased methylation of DNA within calf via jugular venipuncture with a sterile 18-gauge needle. the promoter region of a gene has been reported to suppress Leukocytes were isolated and stored at −80 °C until DNA was transcriptional activity and gene expression (Levine et al., 1991; extracted. Samples from bull calves were stratified into groups Tate and Bird, 1993), whereas increased methylation of DNA in based on the following criteria: 1) adequate availability of DNA, the gene body region of a gene has been suggested to result in 2) calf treatment group, and 3) calf sire. From those stratified increased gene expression (Hellman and Chess, 2007). Therefore, groups, DNA samples from 7 PNS and 7 Control bull calves were the presence or absence of methyl groups at specific locations randomly selected for analysis in this study. within a gene can be utilized as a tool to better understand how prenatal environment shaped postnatal phenotype. Behavior Sample Analysis and stress response traits are multifaceted in nature and Processing of blood samples controlled by many components of neural, neuroendocrine, and Blood samples were centrifuged at 2,671 × g for 30 min at 6 °C. endocrine systems (Chan et al., 2018; Bale et al., 2019). Some Leukocytes were isolated and transferred into 2-mL nuclease- of these systems include the hypothalamic–pituitary–adrenal free microcentrifuge tubes. Samples were washed in red blood (HPA) axis and signaling by catecholamines, monoamines, and cell lysis buffer solution repeatedly until a clean cell pellet was opioids. Differential transcript abundance in the adrenal cortex produced. Leukocyte samples were stored at −80 °C until they (Friedrich et al., 2017) and differential methylation of DNA in were thawed for DNA isolation procedures. different brain regions (Cantrell et al., 2019) have been associated with temperament in cattle. Furthermore, emerging literature DNA extraction supports the ability of glucocorticoids to alter methylation of Phenol-chloroform extraction procedures were used to isolate DNA in developing neural cells; specifically, in vitro exposure DNA from each leukocyte pellet for DNA methylation analysis of early neural progenitor cells to dexamethasone resulted in by standard procedures as previously described (Littlejohn et al., genome-wide differences in methylation of DNA (Provençal 2018). Following DNA precipitation, purified DNA was stored at et al., 2019). Because behavior and stress response phenotypes −80 °C and submitted to Zymo Research Corp (Irvine, CA) for are of economic and biologic significance to beef cattle DNA methylation analysis. production, this study sought to understand the mechanisms by which a common stressor for beef cattle, transportation, Library construction for DNA methylation analysis incurred by a pregnant dam could affect behavior and the stress A method of reduced representation bisulfite sequencing, response of offspring. The objective of this study was to identify Methyl-MiniSeq (Zymo Research; Irvine, CA), was utilized to differentially methylated genes from a previously reported evaluate differential methylation of DNA in samples from PNS genome-wide assessment of DNA methylation in prenatally compared with Control bull calves. Libraries were prepared from stressed bull calves that were related to behavior and stress 200 to 500 ng of genomic DNA digested with 60 units of TaqαI responses (Littlejohn et al., 2018). Because behavior and stress and 30 units of MspI (New England Biolabs, Inc., Ipswich, MA) response phenotypes were altered in the
Recommended publications
  • Original Article Upregulation of HOXA13 As a Potential Tumorigenesis and Progression Promoter of LUSC Based on Qrt-PCR and Bioinformatics
    Int J Clin Exp Pathol 2017;10(10):10650-10665 www.ijcep.com /ISSN:1936-2625/IJCEP0065149 Original Article Upregulation of HOXA13 as a potential tumorigenesis and progression promoter of LUSC based on qRT-PCR and bioinformatics Rui Zhang1*, Yun Deng1*, Yu Zhang1, Gao-Qiang Zhai1, Rong-Quan He2, Xiao-Hua Hu2, Dan-Ming Wei1, Zhen-Bo Feng1, Gang Chen1 Departments of 1Pathology, 2Medical Oncology, First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region, China. *Equal contributors. Received September 7, 2017; Accepted September 29, 2017; Epub October 1, 2017; Published October 15, 2017 Abstract: In this study, we investigated the levels of homeobox A13 (HOXA13) and the mechanisms underlying the co-expressed genes of HOXA13 in lung squamous cancer (LUSC), the signaling pathways in which the co-ex- pressed genes of HOXA13 are involved and their functional roles in LUSC. The clinical significance of 23 paired LUSC tissues and adjacent non-tumor tissues were gathered. HOXA13 levels in LUSC were detected by quantita- tive real-time polymerase chain reaction (qRT-PCR). HOXA13 levels in LUSC from The Cancer Genome Atlas (TCGA) and Oncomine were analyzed. We performed receiver operator characteristic (ROC) curves of various clinicopath- ological features of LUSC. Co-expressed of HOXA13 were collected from MEM, cBioPortal and GEPIA. The func- tions and pathways of the most reliable overlapped genes were achieved from the Gene Otology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) databases, respectively. The protein-protein interaction (PPI) net- works were mapped using STRING. HOXA13 in LUSC were markedly upregulated compared with those in the non- cancerous controls as demonstrated by qRT-PCR (LUSC: 0.330±0.360; CONTROLS: 0.155±0.142; P=0.021).
    [Show full text]
  • Genome-Wide Association and Transcriptome Studies Identify Candidate Genes and Pathways for Feed Conversion Ratio in Pigs
    Miao et al. BMC Genomics (2021) 22:294 https://doi.org/10.1186/s12864-021-07570-w RESEARCH ARTICLE Open Access Genome-wide association and transcriptome studies identify candidate genes and pathways for feed conversion ratio in pigs Yuanxin Miao1,2,3, Quanshun Mei1,2, Chuanke Fu1,2, Mingxing Liao1,2,4, Yan Liu1,2, Xuewen Xu1,2, Xinyun Li1,2, Shuhong Zhao1,2 and Tao Xiang1,2* Abstract Background: The feed conversion ratio (FCR) is an important productive trait that greatly affects profits in the pig industry. Elucidating the genetic mechanisms underpinning FCR may promote more efficient improvement of FCR through artificial selection. In this study, we integrated a genome-wide association study (GWAS) with transcriptome analyses of different tissues in Yorkshire pigs (YY) with the aim of identifying key genes and signalling pathways associated with FCR. Results: A total of 61 significant single nucleotide polymorphisms (SNPs) were detected by GWAS in YY. All of these SNPs were located on porcine chromosome (SSC) 5, and the covered region was considered a quantitative trait locus (QTL) region for FCR. Some genes distributed around these significant SNPs were considered as candidates for regulating FCR, including TPH2, FAR2, IRAK3, YARS2, GRIP1, FRS2, CNOT2 and TRHDE. According to transcriptome analyses in the hypothalamus, TPH2 exhibits the potential to regulate intestinal motility through serotonergic synapse and oxytocin signalling pathways. In addition, GRIP1 may be involved in glutamatergic and GABAergic signalling pathways, which regulate FCR by affecting appetite in pigs. Moreover, GRIP1, FRS2, CNOT2,andTRHDE may regulate metabolism in various tissues through a thyroid hormone signalling pathway.
    [Show full text]
  • Supplementary Table S4. FGA Co-Expressed Gene List in LUAD
    Supplementary Table S4. FGA co-expressed gene list in LUAD tumors Symbol R Locus Description FGG 0.919 4q28 fibrinogen gamma chain FGL1 0.635 8p22 fibrinogen-like 1 SLC7A2 0.536 8p22 solute carrier family 7 (cationic amino acid transporter, y+ system), member 2 DUSP4 0.521 8p12-p11 dual specificity phosphatase 4 HAL 0.51 12q22-q24.1histidine ammonia-lyase PDE4D 0.499 5q12 phosphodiesterase 4D, cAMP-specific FURIN 0.497 15q26.1 furin (paired basic amino acid cleaving enzyme) CPS1 0.49 2q35 carbamoyl-phosphate synthase 1, mitochondrial TESC 0.478 12q24.22 tescalcin INHA 0.465 2q35 inhibin, alpha S100P 0.461 4p16 S100 calcium binding protein P VPS37A 0.447 8p22 vacuolar protein sorting 37 homolog A (S. cerevisiae) SLC16A14 0.447 2q36.3 solute carrier family 16, member 14 PPARGC1A 0.443 4p15.1 peroxisome proliferator-activated receptor gamma, coactivator 1 alpha SIK1 0.435 21q22.3 salt-inducible kinase 1 IRS2 0.434 13q34 insulin receptor substrate 2 RND1 0.433 12q12 Rho family GTPase 1 HGD 0.433 3q13.33 homogentisate 1,2-dioxygenase PTP4A1 0.432 6q12 protein tyrosine phosphatase type IVA, member 1 C8orf4 0.428 8p11.2 chromosome 8 open reading frame 4 DDC 0.427 7p12.2 dopa decarboxylase (aromatic L-amino acid decarboxylase) TACC2 0.427 10q26 transforming, acidic coiled-coil containing protein 2 MUC13 0.422 3q21.2 mucin 13, cell surface associated C5 0.412 9q33-q34 complement component 5 NR4A2 0.412 2q22-q23 nuclear receptor subfamily 4, group A, member 2 EYS 0.411 6q12 eyes shut homolog (Drosophila) GPX2 0.406 14q24.1 glutathione peroxidase
    [Show full text]
  • Table S1. Detailed Clinical Features of Individuals with IDDCA and LADCI Syndromes
    BMJ Publishing Group Limited (BMJ) disclaims all liability and responsibility arising from any reliance Supplemental material placed on this supplemental material which has been supplied by the author(s) J Med Genet Table S1. Detailed clinical features of individuals with IDDCA and LADCI syndromes Lodder E., De Nittis P., Koopman C. et al., 2016 Family A Family B Family C Family D Family E Family F Individual 1 2 3 4 5 6 7 8 9 Gender, Age (years) F, 22 F, 20 F, 6 F, 11 M, 9 F, 12 F, 13 M, 8 M, 23 c.249G>A,r.249_250 c.249G>A,r.249_250 Nucleotide change c.249+1G>T/ c.249+3G>T/ c.249+3G>T/ c.906C>G/ c.242C>T/ c.242C>T/ c.242C>T/ ins249+1_249+25/ ins249+1_249+25/ (NM_006578.3) c.249+1G>T c.249+3G>T c.249+3G>T c.906C>G c.242C>T c.242C>T c.242C>T c.994C>T c.994C>T Amino acid change p.Asp84Valfs*52/ p.Asp84Valfs*52/ p.Asp84Leufs*31/ p.Asp84Valfs*31/ p.Asp84Valfs*31/ p.Tyr302*/ p.(Ser81Leu)/ p.(Ser81Leu)/ p.(Ser81Leu)/ (NP_006569.1) p.(Arg332*) p.(Arg332*) p.Asp84Leufs*31 p.Asp84Valfs*31 p.Asp84Valfs*31 p.Tyr302* p.(Ser81Leu) p.(Ser81Leu) p.(Ser81Leu) 3580 g (50th 2751 g (15 th Birth weight NA NA NA 2845 g (15th NA NA NA percentile) percentile) percentile) Ethnicity Italy Italy Jordan Puerto Rico Puerto Rico India Morocco Morocco Brazil Consanguinity − − + + + − − − + Altered speech + + NR + + + + + NA development - Verbal NA NA nonverbal unremarkable unremarkable NA NA NA NA understanding - Lexical production NA NA nonverbal delayed delayed nonverbal delayed delayed NA Intellectual + + + + + + mild mild mild disability (ID) Epilepsy + + + -
    [Show full text]
  • ADCY1 (N) Antibody, Rabbit Polyclonal
    Order: (888)-282-5810 (Phone) (818)-707-0392 (Fax) [email protected] Web: www.Abiocode.com ADCY1 (N) Antibody, Rabbit Polyclonal Cat#: R0758-1 Lot#: Refer to vial Quantity: 100 ul Application: WB, IP Predicted | Observed MW: 123 | 130-180 kDa Uniprot ID: Q08828 Background: Adenylate cyclase type 1 (ADCY1) is a multi-pass membrane protein that belongs to the adenylyl cyclase class-4/guanylyl cyclase family. ADCY1 is the neural-specific calmodulin-sensitive adenylyl cyclase. Adenylyl cyclases are a group of enzymes that convert adenosine-5'-triphosphate (ATP) into 3',5'-adenosine monophosphate (cAMP) and pyrophosphate. ADCY1 may be involved in regulatory processes in the central nervous system. It may play a role in memory acquisition and learning. Other Names: Adenylate cyclase type 1, ATP pyrophosphate-lyase 1, Adenylate cyclase type I, Adenylyl cyclase 1, Ca(2+)/calmodulin-activated adenylyl cyclase Source and Purity: Rabbit polyclonal antibodies were produced by immunizing animals with a GST-fusion protein containing the N-terminal region of human ADCY1. Antibodies were purified by affinity purification using immunogen. Storage Buffer and Condition: Supplied in 1 x PBS (pH 7.4), 100 ug/ml BSA, 40% Glycerol, 0.01% NaN3. Store at -20 °C. Stable for 6 months from date of receipt. Species Specificity: Human, Mouse Tested Applications: WB: 1:500-1:2,000 (detect endogenous protein*) IP: 1:100-1:200 *: The apparent protein size on WB may be different from the calculated M.W. due to modifications. For research use only. Not for therapeutic or diagnostic purposes. Abiocode, Inc., 29397 Agoura Rd., Ste 106, Agoura Hills, CA 91301 Order: (888)-282-5810 (Phone) (818)-707-0392 (Fax) [email protected] Web: www.Abiocode.com Product Data: IP A B -1 kDa IgG R0758 175 ADCY1 kDa 175 ADCY1 80 80 58 58 46 46 WB: R0758-1 WB: R0758-1 Fig 1.
    [Show full text]
  • Increased Plasma Levels of Adenylate Cyclase 8 and Camp Are Associated with Obesity and Type 2 Diabetes: Results from a Cross-Sectional Study
    biology Article Increased Plasma Levels of Adenylate Cyclase 8 and cAMP Are Associated with Obesity and Type 2 Diabetes: Results from a Cross-Sectional Study 1, 2, , 2, 2 Samy M. Abdel-Halim y, Ashraf Al Madhoun * y , Rasheeba Nizam y, Motasem Melhem , Preethi Cherian 3, Irina Al-Khairi 3, Dania Haddad 2, Mohamed Abu-Farha 3 , Jehad Abubaker 3, Milad S. Bitar 4 and Fahd Al-Mulla 2,* 1 Department of Oncology, Karolinska University Hospital, 17177 Stockholm, Sweden; [email protected] 2 Department of Genetics and Bioinformatics, Dasman Diabetes Institute, Dasman 15462, Kuwait; [email protected] (R.N.); [email protected] (M.M.); [email protected] (D.H.) 3 Department of Biochemistry and Molecular Biology, Dasman Diabetes Institute, Dasman 15462, Kuwait; [email protected] (P.C.); [email protected] (I.A.-K.); [email protected] (M.A.-F.); [email protected] (J.A.) 4 Department of Pharmacology and Toxicology, Faculty of Medicine, Kuwait University, Jabriya 046302, Kuwait; [email protected] * Correspondence: [email protected] (A.A.M.); [email protected] (F.A.-M.); Tel.: +965-2224-2999 (ext. 2805) (A.A.M.); +965-6777-1040 (F.A.-M.) These authors equally contributed to this work. y Received: 5 July 2020; Accepted: 20 August 2020; Published: 24 August 2020 Abstract: Adenylate cyclases (ADCYs) catalyze the conversion of ATP to cAMP,an important co-factor in energy homeostasis. Giving ADCYs role in obesity, diabetes and inflammation, we questioned whether calcium-stimulated ADCY isoforms may be variably detectable in human plasma.
    [Show full text]
  • Supplementary Table 1
    Supplementary Table 1. 492 genes are unique to 0 h post-heat timepoint. The name, p-value, fold change, location and family of each gene are indicated. Genes were filtered for an absolute value log2 ration 1.5 and a significance value of p ≤ 0.05. Symbol p-value Log Gene Name Location Family Ratio ABCA13 1.87E-02 3.292 ATP-binding cassette, sub-family unknown transporter A (ABC1), member 13 ABCB1 1.93E-02 −1.819 ATP-binding cassette, sub-family Plasma transporter B (MDR/TAP), member 1 Membrane ABCC3 2.83E-02 2.016 ATP-binding cassette, sub-family Plasma transporter C (CFTR/MRP), member 3 Membrane ABHD6 7.79E-03 −2.717 abhydrolase domain containing 6 Cytoplasm enzyme ACAT1 4.10E-02 3.009 acetyl-CoA acetyltransferase 1 Cytoplasm enzyme ACBD4 2.66E-03 1.722 acyl-CoA binding domain unknown other containing 4 ACSL5 1.86E-02 −2.876 acyl-CoA synthetase long-chain Cytoplasm enzyme family member 5 ADAM23 3.33E-02 −3.008 ADAM metallopeptidase domain Plasma peptidase 23 Membrane ADAM29 5.58E-03 3.463 ADAM metallopeptidase domain Plasma peptidase 29 Membrane ADAMTS17 2.67E-04 3.051 ADAM metallopeptidase with Extracellular other thrombospondin type 1 motif, 17 Space ADCYAP1R1 1.20E-02 1.848 adenylate cyclase activating Plasma G-protein polypeptide 1 (pituitary) receptor Membrane coupled type I receptor ADH6 (includes 4.02E-02 −1.845 alcohol dehydrogenase 6 (class Cytoplasm enzyme EG:130) V) AHSA2 1.54E-04 −1.6 AHA1, activator of heat shock unknown other 90kDa protein ATPase homolog 2 (yeast) AK5 3.32E-02 1.658 adenylate kinase 5 Cytoplasm kinase AK7
    [Show full text]
  • Enhanced Expression of ADCY1 Underlies Aberrant Neuronal Signalling and Behaviour in a Syndromic Autism Model
    ARTICLE Received 16 Jun 2016 | Accepted 20 Dec 2016 | Published 20 Feb 2017 DOI: 10.1038/ncomms14359 OPEN Enhanced expression of ADCY1 underlies aberrant neuronal signalling and behaviour in a syndromic autism model Ferzin Sethna1, Wei Feng2, Qi Ding3, Alfred J. Robison3,4, Yue Feng2 & Hongbing Wang3,4 Fragile X syndrome (FXS), caused by the loss of functional FMRP, is a leading cause of autism. Neurons lacking FMRP show aberrant mRNA translation and intracellular signalling. Here, we identify that, in Fmr1 knockout neurons, type 1 adenylyl cyclase (Adcy1) mRNA translation is enhanced, leading to excessive production of ADCY1 protein and insensitivity to neuronal stimulation. Genetic reduction of Adcy1 normalizes the aberrant ERK1/2- and PI3K-mediated signalling, attenuates excessive protein synthesis and corrects dendritic spine abnormality in Fmr1 knockout mice. Genetic reduction of Adcy1 also ameliorates autism-related symptoms including repetitive behaviour, defective social interaction and audiogenic seizures. Moreover, peripheral administration of NB001, an experimental compound that preferentially suppresses ADCY1 activity over other ADCY subtypes, attenuates the behavioural abnorm- alities in Fmr1 knockout mice. These results demonstrate a connection between the elevated Adcy1 translation and abnormal ERK1/2 signalling and behavioural symptoms in FXS. 1 Genetics Program, Michigan State University, East Lansing, Michigan 48824, USA. 2 Department of Pharmacology, Emory University School of Medicine, Atlanta, Georgia 30322, USA. 3
    [Show full text]
  • Therapeutic Opportunities in Colon Cancer Focus on Phosphodiesterase Inhibitors
    Life Sciences 230 (2019) 150–161 Contents lists available at ScienceDirect Life Sciences journal homepage: www.elsevier.com/locate/lifescie Review article Therapeutic opportunities in colon cancer: Focus on phosphodiesterase inhibitors T ⁎ Ankita Mehta, Bhoomika M. Patel Institute of Pharmacy, Nirma University, Ahmedabad, India ARTICLE INFO ABSTRACT Keywords: Despite novel technologies, colon cancer remains undiagnosed and 25% of patients are diagnosed with meta- Phosphodiesterases static colon cancer. Resistant to chemotherapeutic agents is one of the major problems associated with treating cAMP colon cancer which creates the need to develop novel agents targeting towards newer targets. A phosphodies- cGMP terase is a group of isoenzyme, which, hydrolyze cyclic nucleotides and thereby lowers intracellular levels of Adenylate cyclase cAMP and cGMP leading to tumorigenic effects. Many in vitro and in vivo studies have confirmed increased PDE Guanylate cyclase expression in different types of cancers including colon cancer. cAMP-specific PDE inhibitors increase in- Colon cancer tracellular cAMP that leads to activation of effector molecules-cAMP-dependent protein kinase A, exchange protein activated by cAMP and cAMP gated ion channels. These molecules regulate cellular responses and exert its anticancer role through different mechanisms including apoptosis, inhibition of angiogenesis, upregulating tumor suppressor genes and suppressing oncogenes. On the other hand, cGMP specific PDE inhibitors exhibit anticancer effects through cGMP dependent protein kinase and cGMP dependent cation channels. Elevation in cGMP works through activation of caspases, suppression of Wnt/b-catenin pathway and TCF transcription leading to inhibition of CDK and survivin. These studies point out towards the fact that PDE inhibition is as- sociated with anti-proliferative, anti-apoptotic and anti-angiogenic pathways involved in its anticancer effects in colon cancer.
    [Show full text]
  • Antagonism of Forkhead Box Subclass O Transcription Factors Elicits Loss of Soluble Guanylyl Cyclase Expression S
    Supplemental material to this article can be found at: http://molpharm.aspetjournals.org/content/suppl/2019/04/15/mol.118.115386.DC1 1521-0111/95/6/629–637$35.00 https://doi.org/10.1124/mol.118.115386 MOLECULAR PHARMACOLOGY Mol Pharmacol 95:629–637, June 2019 Copyright ª 2019 by The Author(s) This is an open access article distributed under the CC BY-NC Attribution 4.0 International license. Antagonism of Forkhead Box Subclass O Transcription Factors Elicits Loss of Soluble Guanylyl Cyclase Expression s Joseph C. Galley, Brittany G. Durgin, Megan P. Miller, Scott A. Hahn, Shuai Yuan, Katherine C. Wood, and Adam C. Straub Heart, Lung, Blood and Vascular Medicine Institute (J.C.G., B.G.D., M.P.M., S.A.H., S.Y., K.C.W., A.C.S.) and Department of Pharmacology and Chemical Biology (J.C.G., A.C.S.), University of Pittsburgh, Pittsburgh, Pennsylvania Received November 29, 2018; accepted March 31, 2019 Downloaded from ABSTRACT Nitric oxide (NO) stimulates soluble guanylyl cyclase (sGC) protein expression showed a concentration-dependent down- activity, leading to elevated intracellular cyclic guano- regulation. Consistent with the loss of sGC a and b mRNA and sine 39,59-monophosphate (cGMP) and subsequent vascular protein expression, pretreatment of vascular smooth muscle smooth muscle relaxation. It is known that downregulation of cells with the FoxO inhibitor decreased sGC activity mea- sGC expression attenuates vascular dilation and contributes to sured by cGMP production following stimulation with an NO molpharm.aspetjournals.org the pathogenesis of cardiovascular disease. However, it is not donor.
    [Show full text]
  • Neomorphic Effects of Recurrent Somatic Mutations in Yin Yang 1 in Insulin-Producing Adenomas
    Neomorphic effects of recurrent somatic mutations in Yin Yang 1 in insulin-producing adenomas M. Kyle Cromera,b, Murim Choia,b, Carol Nelson-Williamsa,b, Annabelle L. Fonsecac,d,e, John W. Kunstmanc,d,e, Reju M. Korahc,d,e, John D. Overtona,f, Shrikant Manea,f, Barton Kenneyg, Carl D. Malchoffh,i, Peter Stalbergj, Göran Akerströmj, Gunnar Westinj, Per Hellmanj, Tobias Carlingc,d,e, Peyman Björklundj, and Richard P. Liftona,b,f,k,1 Departments of aGenetics, cSurgery, gPathology, and kInternal Medicine, bHoward Hughes Medical Institute, dYale Endocrine Neoplasia Laboratory, eYale Cancer Center, and fYale Center for Genome Analysis, Yale University School of Medicine, New Haven, CT 06510; hDivision of Endocrinology and iNeag Cancer Center, University of Connecticut Health Center, Farmington, CT 06030; and jDepartment of Surgical Sciences, Uppsala University, Uppsala, Sweden 751 05 Contributed by Richard P. Lifton, February 25, 2015 (sent for review January 15, 2015; reviewed by Mitchell A. Lazar and Vamsi K. Mootha) Insulinomas are pancreatic islet tumors that inappropriately secrete provides the basis for the clinical efficacy of GLP-1 and inhibitors insulin, producing hypoglycemia. Exome and targeted sequencing of dipeptidyl peptidase 4, which normally degrades GLP-1; these revealed that 14 of 43 insulinomas harbored the identical somatic drugs increase glucose-dependent insulin secretion and lower mutation in the DNA-binding zinc finger of the transcription fac- blood glucose (10). + tor Yin Yang 1 (YY1). Chromatin immunoprecipitation sequencing Sustained elevations in both intracellular Ca2 and GLP-1 sig- (ChIP-Seq) showed that this T372R substitution changes the DNA naling are known to also promote proliferation of β-cells (11, 12).
    [Show full text]
  • Supplemental Figures 04 12 2017
    Jung et al. 1 SUPPLEMENTAL FIGURES 2 3 Supplemental Figure 1. Clinical relevance of natural product methyltransferases (NPMTs) in brain disorders. (A) 4 Table summarizing characteristics of 11 NPMTs using data derived from the TCGA GBM and Rembrandt datasets for 5 relative expression levels and survival. In addition, published studies of the 11 NPMTs are summarized. (B) The 1 Jung et al. 6 expression levels of 10 NPMTs in glioblastoma versus non‐tumor brain are displayed in a heatmap, ranked by 7 significance and expression levels. *, p<0.05; **, p<0.01; ***, p<0.001. 8 2 Jung et al. 9 10 Supplemental Figure 2. Anatomical distribution of methyltransferase and metabolic signatures within 11 glioblastomas. The Ivy GAP dataset was downloaded and interrogated by histological structure for NNMT, NAMPT, 12 DNMT mRNA expression and selected gene expression signatures. The results are displayed on a heatmap. The 13 sample size of each histological region as indicated on the figure. 14 3 Jung et al. 15 16 Supplemental Figure 3. Altered expression of nicotinamide and nicotinate metabolism‐related enzymes in 17 glioblastoma. (A) Heatmap (fold change of expression) of whole 25 enzymes in the KEGG nicotinate and 18 nicotinamide metabolism gene set were analyzed in indicated glioblastoma expression datasets with Oncomine. 4 Jung et al. 19 Color bar intensity indicates percentile of fold change in glioblastoma relative to normal brain. (B) Nicotinamide and 20 nicotinate and methionine salvage pathways are displayed with the relative expression levels in glioblastoma 21 specimens in the TCGA GBM dataset indicated. 22 5 Jung et al. 23 24 Supplementary Figure 4.
    [Show full text]