Mutation Screen of the SIM1 Gene in Pediatric Patients with Early-Onset Obesity

Total Page:16

File Type:pdf, Size:1020Kb

Mutation Screen of the SIM1 Gene in Pediatric Patients with Early-Onset Obesity International Journal of Obesity (2014) 38, 1000–1004 & 2014 Macmillan Publishers Limited All rights reserved 0307-0565/14 www.nature.com/ijo ORIGINAL ARTICLE Mutation screen of the SIM1 gene in pediatric patients with early-onset obesity D Zegers1, S Beckers1, R Hendrickx1, JK Van Camp1, V de Craemer1, A Verrijken2, K Van Hoorenbeeck3, SL Verhulst3, RP Rooman3, KN Desager3, G Massa4, LF Van Gaal2 and W Van Hul1 BACKGROUND: The transcription factor SIM1 (Single-minded 1) is involved in the control of food intake and in the pathogenesis of obesity. In mice, Sim1 is involved in the development of the paraventricular nucleus, and Sim1 deficiency leads to severe obesity and hyperphagia. In humans, chromosomal abnormalities in the SIM1 gene region have been reported in obese individuals. Furthermore, recent data also suggest that loss-of-function point mutations in SIM1 are responsible for SIM1 haplo-insufficiency that is involved in causing human obesity. In this study, we therefore wanted to expand the evidence regarding the involvement of SIM1 mutations in the pathogenesis of severe early-onset obesity. METHODS: We screened 561 severely overweight and obese children and adolescents and 453 lean adults for mutations in the coding region of the SIM1 gene. Mutation screening in all patients and lean individuals was performed by high-resolution melting curve analysis combined with direct sequencing. To evaluate the effect of the mutations on SIM1 transcriptional activity, luciferase reporter assays were performed. RESULTS: Mutation analysis identified four novel nonsynonymous coding variants in SIM1 in four unrelated obese individuals: p.L242V, p.T481K, p.A517V and p.D590E. Five synonymous variants, p.P57P, p.F93F, p.I183I, p.V208V and p.T653T, were also identified. Screening of the lean control population revealed the occurrence of four other rare SIM1 variants: p.G408R, p.R471P, p.S492P and p.S622F. For variants p.T481K and p.A517V, which were found in obese individuals, a decrease in SIM1 transcriptional activity was observed, whereas the transcriptional activity of all variants found in lean individuals resembled wild type. CONCLUSIONS: In this study, we have demonstrated the presence of rare SIM1 variants in both an obese pediatric population and a population of lean adult controls. Further, we have shown that functional in vitro analysis of SIM1 variants may help in distinguishing benign variants of no pathogenic significance from variants which contribute to the obesity phenotype. International Journal of Obesity (2014) 38, 1000–1004; doi:10.1038/ijo.2013.188 Keywords: mutation analysis; SIM1; melanocortin signaling INTRODUCTION hypothalamic oxytocin and PVN melanocortin 4 receptor (Mc4r) SIM1 (Single-minded 1) is a member of the bHLH-PAS (basic helix- mRNA, suggesting that the hyperphagic obesity in Sim1-deficient loop-helix Per-Arnt-Sim) family of transcription factors. In mice, mice may be attributable to changes in the leptin-melanocortin- 6 Sim1 is essential for the formation of the paraventricular nucleus oxytocin pathway. The hypothesis that SIM1 might be a factor (PVN) in the hypothalamus,1 a region that is known to be within the famous leptin-melanocortin signaling pathway is implicated in the regulation of body weight, as neurons in the PVN further supported by the investigation of transgenic mice express murine Mc4r and are a target of a-MSH.2,3 Homozygous overexpressing human SIM1. Overexpression of SIM1 completely Sim1-knockout mice (Sim1 À / À ) lack a PVN and die perinatally. rescued the hyperphagia and partially rescued the obesity In contrast, heterozygous Sim1 þ / À mice are viable, but they show exhibited by agouti yellow mice, in which melanocortin early-onset obesity, increased linear growth and are hyperphagic, signaling is interrupted, suggesting that Sim1 acts downstream making their phenotype resemble mc4r-mutant mice. Partial Sim1 of Mc4r to control food intake.7 deficiency results in hypodevelopment of the PVN that contains SIM1 deficiency in humans has also been reported: a girl with on average 24% fewer cells.4 As lesions in the PVN also induce early-onset severe obesity and a de novo translocation between increased appetite,5 it was suggested that this hypocellularity chromosomes 1p22.1 and 6q16.2 has been described by Holder causes the hyperphagia and obesity of the Sim þ / À mice.4 More et al.8 In this case, the translocation separates the 50 promoter recently, Tolson et al.6 demonstrated that the role of Sim1 in region and bHLH domain from the rest of the gene. It was feeding regulation is not limited to formation of the PVN. They hypothesized that haplo-insufficiency of SIM1 was responsible for conditionally deleted Sim1 in mice postnatally, thereby generating the severe obesity in the subject.8 Further, in multiple obese conditional Sim1 heterozygous mice and conditional Sim1 patients with a Prader-Willi-like phenotype, deletions of the homozygote mice and revealing a dosage-dependent effect of 6q16.2-6q16.3 region have been reported. These deletions on 6q Sim1 on obesity. These mice exhibited a remarkable decrease in disrupt or delete the SIM1 gene, possibly giving rise to obesity.9–11 1Department of Medical Genetics, University of Antwerp and Antwerp University Hospital, Antwerp, Belgium; 2Department of Endocrinology, Diabetology and Metabolic Diseases, Antwerp University Hospital, Antwerp, Belgium; 3Department of Paediatrics, Antwerp University Hospital, Antwerp, Belgium and 4Department of Paediatrics, Jessa Hospital, Hasselt, Belgium. Correspondence: Professor Dr W Van Hul, Department of Medical Genetics, University of Antwerp, Universiteitsplein 1, Antwerp B-2610, Belgium. E-mail: [email protected] Received 11 April 2013; revised 12 September 2013; accepted 24 September 2013; accepted article preview online 7 October 2013; advance online publication, 29 October 2013 Involvement of SIM1 mutations in childhood obesity D Zegers et al 1001 More recent data also suggest that loss-of-function point standard PCR protocol. Real-time PCR was performed as previously mutations in SIM1 are responsible for SIM1 haplo-insufficiency described.19 After amplification, samples were first denaturated at 95 1C that is involved in causing human obesity12–14 and Prader- and subsequently renaturated at 40 1C and then heated during Willi-like syndrome.12,13 Because of the alleged role of SIM1 fluorescence acquisition between 70 and 95 1C. When melting curves downstream of the leptin-melanocortin pathway and the reports deviated from WT, direct sequencing using an ABI Prism 3130xl Genetic Analyzer with ABI BigDye Terminator v1.1 Cycle Sequencing kits (Applied on SIM1 haplo-insufficiency in obese humans, we hypothesized Biosystems, Foster City, CA, USA) was performed in order to characterize that heterozygous point mutations in the SIM1 gene might be the putative sequence variations. Primer sequences for high-resolution melting cause of severe early-onset obesity, and we set out to screen the curve analysis and direct sequencing are available on request. SIM1 gene region in an extensive cohort of obese children. Constructs MATERIALS AND METHODS Wild-type (WT) human SIM1 subcloned into a pDR2 vector was kindly provided by Professor Chrast (University of Lausanne, Lausanne, Switzer- Study population land). The entire coding region of the hSIM1 gene was amplified using Mutation screening of the SIM1 gene was performed in a cohort of 561 specific primers introducing a HindIII and an EcoRI restriction site 50 and 30 severely overweight and obese children and adolescents and 453 healthy, of the SIM1 sequence, respectively. The PCR product was subsequently lean adults. Children and adolescents (243 boys, 318 girls) were recruited cloned into a pcDNA3 expression vector (Invitrogen, Carlsbad, CA, USA). from the pediatrics departments of the Antwerp University Hospital and Mutations were introduced into WT SIM1, subcloned into the pcDNA3 the Jessa Hospital, Hasselt, both in Belgium. This group (age range 0–12) expression vector, using the QuikChange II Site-Directed Mutagenesis Kit included 257 children aged 8.7±2.5 years (mean±s.d.) and 304 (Stratagene, La Jolla, CA, USA). The WT sequence and the presence of the adolescents aged 15.2±2.3 years, (age range 12–21; summary of all mutations were checked and confirmed by direct sequencing using an ABI characteristics: Table 1). The cohort of healthy lean adults (18.5 kg m À 2 3130xl Genetic Analyzer (Applied Biosystems) according to the protocol Xbody mass index (BMI)X24.9 kg m À 2) was recruited among employees described above to ensure that no errors had been induced. Luciferase from the university and the university hospital and among couples seeking reporter plasmid pGL3-HRE-Luc was kindly provided by professor Whitelaw prenatal counseling at the Department of Medical Genetics. This control (University of Adelaide, Adelaide, Australia). Wild-type human ARNT2 cDNA group included 160 men and 293 women, aged 35.2±7.2 years, with a cloned in a pCMV6-XL4 expression vector was obtained from Origene mean BMI of 22.1±1.7 kg m À 2. The study was approved by the local ethics Technologies (Rockville, MD, USA). committee, and all participants gave their written informed consent once the aim and design of the study had been explained. Reporter assay SIM1 For monitoring SIM1 transcriptional activity in cells transiently expressing Anthropometry WT or mutant SIM1, a luciferase assay was performed. HEK293 cells were Height was measured to the nearest 0.5 cm, and body weight was maintained in DMEM supplemented with 10% fetal bovine serum, measured on a digital scale to the nearest 0.1 kg. BMI was calculated for all 100 units ml À 1 penicillin and 100 mgmlÀ 1 streptomycin (Invitrogen). Cells patients and BMI-for-age percentile (490th percentile for all patients were incubated at 37 1C in humidified air containing 5% CO2. Cells were included) and Z-scores were based on the ‘Flemish Growth Charts 2004’.15 plated in 24-well plates at a density of 50 000 cells per well.
Recommended publications
  • 1 Evidence for Gliadin Antibodies As Causative Agents in Schizophrenia
    1 Evidence for gliadin antibodies as causative agents in schizophrenia. C.J.Carter PolygenicPathways, 20 Upper Maze Hill, Saint-Leonard’s on Sea, East Sussex, TN37 0LG [email protected] Tel: 0044 (0)1424 422201 I have no fax Abstract Antibodies to gliadin, a component of gluten, have frequently been reported in schizophrenia patients, and in some cases remission has been noted following the instigation of a gluten free diet. Gliadin is a highly immunogenic protein, and B cell epitopes along its entire immunogenic length are homologous to the products of numerous proteins relevant to schizophrenia (p = 0.012 to 3e-25). These include members of the DISC1 interactome, of glutamate, dopamine and neuregulin signalling networks, and of pathways involved in plasticity, dendritic growth or myelination. Antibodies to gliadin are likely to cross react with these key proteins, as has already been observed with synapsin 1 and calreticulin. Gliadin may thus be a causative agent in schizophrenia, under certain genetic and immunological conditions, producing its effects via antibody mediated knockdown of multiple proteins relevant to the disease process. Because of such homology, an autoimmune response may be sustained by the human antigens that resemble gliadin itself, a scenario supported by many reports of immune activation both in the brain and in lymphocytes in schizophrenia. Gluten free diets and removal of such antibodies may be of therapeutic benefit in certain cases of schizophrenia. 2 Introduction A number of studies from China, Norway, and the USA have reported the presence of gliadin antibodies in schizophrenia 1-5. Gliadin is a component of gluten, intolerance to which is implicated in coeliac disease 6.
    [Show full text]
  • A Computational Approach for Defining a Signature of Β-Cell Golgi Stress in Diabetes Mellitus
    Page 1 of 781 Diabetes A Computational Approach for Defining a Signature of β-Cell Golgi Stress in Diabetes Mellitus Robert N. Bone1,6,7, Olufunmilola Oyebamiji2, Sayali Talware2, Sharmila Selvaraj2, Preethi Krishnan3,6, Farooq Syed1,6,7, Huanmei Wu2, Carmella Evans-Molina 1,3,4,5,6,7,8* Departments of 1Pediatrics, 3Medicine, 4Anatomy, Cell Biology & Physiology, 5Biochemistry & Molecular Biology, the 6Center for Diabetes & Metabolic Diseases, and the 7Herman B. Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN 46202; 2Department of BioHealth Informatics, Indiana University-Purdue University Indianapolis, Indianapolis, IN, 46202; 8Roudebush VA Medical Center, Indianapolis, IN 46202. *Corresponding Author(s): Carmella Evans-Molina, MD, PhD ([email protected]) Indiana University School of Medicine, 635 Barnhill Drive, MS 2031A, Indianapolis, IN 46202, Telephone: (317) 274-4145, Fax (317) 274-4107 Running Title: Golgi Stress Response in Diabetes Word Count: 4358 Number of Figures: 6 Keywords: Golgi apparatus stress, Islets, β cell, Type 1 diabetes, Type 2 diabetes 1 Diabetes Publish Ahead of Print, published online August 20, 2020 Diabetes Page 2 of 781 ABSTRACT The Golgi apparatus (GA) is an important site of insulin processing and granule maturation, but whether GA organelle dysfunction and GA stress are present in the diabetic β-cell has not been tested. We utilized an informatics-based approach to develop a transcriptional signature of β-cell GA stress using existing RNA sequencing and microarray datasets generated using human islets from donors with diabetes and islets where type 1(T1D) and type 2 diabetes (T2D) had been modeled ex vivo. To narrow our results to GA-specific genes, we applied a filter set of 1,030 genes accepted as GA associated.
    [Show full text]
  • User's Manual Mobile Phone
    User’s Manual Mobile Phone Model: AM206 FCC ID: UOSAM206 1 TABLE OF CONTENT 1. BASIC INFORMATION .................... 6 1.1. BRIEF INTRODUCTION ........................................... 6 2. BEFORE USE ..................................... 7 2.1. NAME AND EXPLANATION OF EACH PART ............. 7 2.1.1. Appearance Sketch Map ........................... 7 2.1.2. Description of the Standby Pictures .......... 7 2.1.3. Description of Keys .................................. 8 2.2. INSTALL SIM CARD AND MEMORY CARD ........... 10 2.2.1. SIM Card ................................................. 10 2.2.2. Inserting and Taking out SIM Card ........ 11 2.2.3. Memory Card .......................................... 12 2.3. BATTERY ............................................................ 12 2.3.1. Install the Battery .................................... 12 2.3.2. Charging .................................................. 13 2.4. SECURITY PASSWORD ......................................... 14 2 3. QUICK USER GUIDE ..................... 15 3.1. DIAL A CALL ....................................................... 15 3.2. REJECT A CALL ................................................... 15 3.3. RECEIVE A CALL ................................................. 15 3.4. END A CALL ........................................................ 16 3.5. EMERGENCY SERVICES ....................................... 16 3.6. EXTENSION SPEED DIAL ..................................... 16 3.7. DIALED CALLS/MISSED CALLS/RECEIVED CALLS/REJECTED CALLS .............................................
    [Show full text]
  • Supplementary Materials
    Supplementary materials Supplementary Table S1: MGNC compound library Ingredien Molecule Caco- Mol ID MW AlogP OB (%) BBB DL FASA- HL t Name Name 2 shengdi MOL012254 campesterol 400.8 7.63 37.58 1.34 0.98 0.7 0.21 20.2 shengdi MOL000519 coniferin 314.4 3.16 31.11 0.42 -0.2 0.3 0.27 74.6 beta- shengdi MOL000359 414.8 8.08 36.91 1.32 0.99 0.8 0.23 20.2 sitosterol pachymic shengdi MOL000289 528.9 6.54 33.63 0.1 -0.6 0.8 0 9.27 acid Poricoic acid shengdi MOL000291 484.7 5.64 30.52 -0.08 -0.9 0.8 0 8.67 B Chrysanthem shengdi MOL004492 585 8.24 38.72 0.51 -1 0.6 0.3 17.5 axanthin 20- shengdi MOL011455 Hexadecano 418.6 1.91 32.7 -0.24 -0.4 0.7 0.29 104 ylingenol huanglian MOL001454 berberine 336.4 3.45 36.86 1.24 0.57 0.8 0.19 6.57 huanglian MOL013352 Obacunone 454.6 2.68 43.29 0.01 -0.4 0.8 0.31 -13 huanglian MOL002894 berberrubine 322.4 3.2 35.74 1.07 0.17 0.7 0.24 6.46 huanglian MOL002897 epiberberine 336.4 3.45 43.09 1.17 0.4 0.8 0.19 6.1 huanglian MOL002903 (R)-Canadine 339.4 3.4 55.37 1.04 0.57 0.8 0.2 6.41 huanglian MOL002904 Berlambine 351.4 2.49 36.68 0.97 0.17 0.8 0.28 7.33 Corchorosid huanglian MOL002907 404.6 1.34 105 -0.91 -1.3 0.8 0.29 6.68 e A_qt Magnogrand huanglian MOL000622 266.4 1.18 63.71 0.02 -0.2 0.2 0.3 3.17 iolide huanglian MOL000762 Palmidin A 510.5 4.52 35.36 -0.38 -1.5 0.7 0.39 33.2 huanglian MOL000785 palmatine 352.4 3.65 64.6 1.33 0.37 0.7 0.13 2.25 huanglian MOL000098 quercetin 302.3 1.5 46.43 0.05 -0.8 0.3 0.38 14.4 huanglian MOL001458 coptisine 320.3 3.25 30.67 1.21 0.32 0.9 0.26 9.33 huanglian MOL002668 Worenine
    [Show full text]
  • Brain Nuclear Receptors and Body Weight Regulation
    Brain nuclear receptors and body weight regulation Yong Xu, … , Bert W. O’Malley, Joel K. Elmquist J Clin Invest. 2017;127(4):1172-1180. https://doi.org/10.1172/JCI88891. Review Series Neural pathways, especially those in the hypothalamus, integrate multiple nutritional, hormonal, and neural signals, resulting in the coordinated control of body weight balance and glucose homeostasis. Nuclear receptors (NRs) sense changing levels of nutrients and hormones, and therefore play essential roles in the regulation of energy homeostasis. Understanding the role and the underlying mechanisms of NRs in the context of energy balance control may facilitate the identification of novel targets to treat obesity. Notably, NRs are abundantly expressed in the brain, and emerging evidence indicates that a number of these brain NRs regulate multiple aspects of energy balance, including feeding, energy expenditure and physical activity. In this Review we summarize some of the recent literature regarding effects of brain NRs on body weight regulation and discuss mechanisms underlying these effects. Find the latest version: http://jci.me/88891/pdf REVIEW SERIES: NUCLEAR RECEPTORS The Journal of Clinical Investigation Series Editor: Mitchell A. Lazar Brain nuclear receptors and body weight regulation Yong Xu,1,2 Bert W. O’Malley,2 and Joel K. Elmquist3 1Children’s Nutrition Research Center, Department of Pediatrics, and 2Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas, USA. 3Division of Hypothalamic Research, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas, USA. Neural pathways, especially those in the hypothalamus, integrate multiple nutritional, hormonal, and neural signals, resulting in the coordinated control of body weight balance and glucose homeostasis.
    [Show full text]
  • Supporting Information Supplementary Figure S1
    Supporting Information Supplementary Figure S1. Expression of Mir96, Mir182 and Mir183 in Mir183/96dko mutant mice (top) and Mir182ko mutant mice (bottom), relative to Mir99a, which is expressed in cochlear sensory epithelium. Homozygote (red; right bars) and heterozygote (blue; middle bars) expression levels have been normalised to expression in the wildtype (green; left bars). Mir183/96dko: wildtype n=7, heterozygote n=5, homozygote n=6. One way ANOVA: Mir96 p<0.001 (wildtype vs. heterozygote Bonferroni‐corrected p<0.001; wildtype vs. homozygote Bonferroni‐corrected p<0.001; heterozygote vs. homozygote Bonferroni‐corrected p=0.001) ; Mir182 p=0.37; Mir183 p<0.001 (wildtype vs. heterozygote Bonferroni‐corrected p=0.001; wildtype vs. homozygote Bonferroni‐corrected p<0.001; heterozygote vs. homozygote Bonferroni‐corrected p<0.001). Mir182ko: wildtype n=4, heterozygote n=4, homozygote n=4. One way ANOVA: Mir96 p=0.685; Mir182 p=0.003 (wildtype vs. heterozygote Bonferroni‐corrected p=0.397; wildtype vs. homozygote Bonferroni‐corrected p=0.003; heterozygote vs. homozygote Bonferroni‐corrected p=0.032); Mir183 p=0.04 (wildtype vs. heterozygote Bonferroni‐corrected p=1.0; wildtype vs. homozygote Bonferroni‐corrected p=0.068; heterozygote vs. homozygote Bonferroni‐corrected p=0.094), Error bars are standard deviation (* = P < 0.05, ** = P ≤ 0.01). Supplementary Figure S2. Individual ABR thresholds of wildtype, heterozygous and homozygous Mir183/96dko mice at all ages tested. Number of mice of each genotype tested at each age is shown on the threshold plot. Supplementary Figure S3. Individual ABR thresholds of wildtype, heterozygous and homozygous Mir182ko mice at all ages tested.
    [Show full text]
  • Reprogramming of Lysosomal Gene Expression by Interleukin-4 and Stat6 Brignull Et Al
    Reprogramming of lysosomal gene expression by interleukin-4 and Stat6 Brignull et al. Brignull et al. BMC Genomics 2013, 14:853 http://www.biomedcentral.com/1471-2164/14/853 Brignull et al. BMC Genomics 2013, 14:853 http://www.biomedcentral.com/1471-2164/14/853 RESEARCH ARTICLE Open Access Reprogramming of lysosomal gene expression by interleukin-4 and Stat6 Louise M Brignull1†, Zsolt Czimmerer2†, Hafida Saidi1,3, Bence Daniel2, Izabel Villela4,5, Nathan W Bartlett6, Sebastian L Johnston6, Lisiane B Meira4, Laszlo Nagy2,7 and Axel Nohturfft1* Abstract Background: Lysosomes play important roles in multiple aspects of physiology, but the problem of how the transcription of lysosomal genes is coordinated remains incompletely understood. The goal of this study was to illuminate the physiological contexts in which lysosomal genes are coordinately regulated and to identify transcription factors involved in this control. Results: As transcription factors and their target genes are often co-regulated, we performed meta-analyses of array-based expression data to identify regulators whose mRNA profiles are highly correlated with those of a core set of lysosomal genes. Among the ~50 transcription factors that rank highest by this measure, 65% are involved in differentiation or development, and 22% have been implicated in interferon signaling. The most strongly correlated candidate was Stat6, a factor commonly activated by interleukin-4 (IL-4) or IL-13. Publicly available chromatin immunoprecipitation (ChIP) data from alternatively activated mouse macrophages show that lysosomal genes are overrepresented among Stat6-bound targets. Quantification of RNA from wild-type and Stat6-deficient cells indicates that Stat6 promotes the expression of over 100 lysosomal genes, including hydrolases, subunits of the vacuolar H+ ATPase and trafficking factors.
    [Show full text]
  • Transcription Factor -Catenin Plays a Key Role in Fluid Flow Shear Stress
    cells Article Transcription Factor β-Catenin Plays a Key Role in Fluid Flow Shear Stress-Mediated Glomerular Injury in Solitary Kidney Tarak Srivastava 1,2,3,*, Daniel P. Heruth 4 , R. Scott Duncan 5, Mohammad H. Rezaiekhaligh 1, Robert E. Garola 6, Lakshmi Priya 1, Jianping Zhou 2,7, Varun C. Boinpelly 2,7, Jan Novak 8, Mohammed Farhan Ali 1, Trupti Joshi 9,10,11,12, Uri S. Alon 1, Yuexu Jiang 10,11, Ellen T. McCarthy 13, Virginia J. Savin 7, Ram Sharma 7, Mark L. Johnson 3 and Mukut Sharma 2,7,13 1 Section of Nephrology, Children’s Mercy Hospital and University of Missouri at Kansas City, Kansas City, MO 64108, USA; [email protected] (M.H.R.); [email protected] (L.P.); [email protected] (M.F.A.); [email protected] (U.S.A.) 2 Midwest Veterans’ Biomedical Research Foundation (MVBRF), Kansas City, MO 64128, USA; [email protected] (J.Z.); [email protected] (V.C.B.); [email protected] (M.S.) 3 Department of Oral and Craniofacial Sciences, School of Dentistry, University of Missouri at Kansas City, Kansas City, MO 64108, USA; [email protected] 4 Children’s Mercy Research Institute, Children’s Mercy Hospital and University of Missouri at Kansas City, Kansas City, MO 64108, USA; [email protected] 5 School of Biological Sciences, University of Missouri at Kansas City, Kansas City, MO 64108, USA; [email protected] 6 Department of Pathology and Laboratory Medicine, Children’s Mercy Hospital and University of Missouri at Kansas City, Kansas City, MO 64108, USA; [email protected] 7 Kansas City VA Medical Center, Kansas City, MO 64128, USA; [email protected] (V.J.S.); Citation: Srivastava, T.; Heruth, D.P.; [email protected] (R.S.) 8 Duncan, R.S.; Rezaiekhaligh, M.H.; Department of Microbiology, University of Alabama at Birmingham, Birmingham, AL 35487, USA; Garola, R.E.; Priya, L.; Zhou, J.; [email protected] 9 Department of Health Management and Informatics, University of Missouri, Columbia, MO 65211, USA; Boinpelly, V.C.; Novak, J.; Ali, M.F.; [email protected] et al.
    [Show full text]
  • Deletion of the Sclerotome-Enriched Lncrna PEAT Augments Ribosomal Protein Expression
    Deletion of the sclerotome-enriched lncRNA PEAT augments ribosomal protein expression David A. Stafforda, Darwin S. Dichmanna, Jessica K. Changb, and Richard M. Harlanda,1 aDepartment of Molecular and Cell Biology, University of California, Berkeley, CA 94720; and bDepartment of Genetics, Stanford University, Stanford, CA 94305-5120 Contributed by Richard M. Harland, September 23, 2016 (sent for review July 22, 2016; reviewed by Margaret Buckingham and Chen-Ming Fan) To define a complete catalog of the genes that are activated during developing sclerotome by dissection from early embryos at 8- to mouse sclerotome formation, we sequenced RNA from embryonic 9.5-d postconception is technically difficult. Although a model mouse tissue directed to form sclerotome in culture. In addition to for presomitic mesoderm culture from stem cells has been de- well-known early markers of sclerotome, such as Pax1, Pax9,andthe veloped (10), this has not been exploited as an in vitro model of Bapx2/Nkx3-2 homolog Nkx3-1, the long-noncoding RNA PEAT (Pax1 the sclerotome. To obtain sufficient sclerotomal transcripts for enhancer antisense transcript) was induced in sclerotome-directed RNA-seq, we chemically treated explants of embryonic tissue to samples. Strikingly, PEAT is located just upstream of the Pax1 gene. bias differentiation of fluorescently marked presomitic mesoderm Using CRISPR/Cas9, we generated a mouse line bearing a complete toward the sclerotome. Following FACS, we compared control deletion of the PEAT-transcribed unit. RNA-seq on PEAT mutant and induced cultures to define the sclerotome transcriptome. In embryos showed that loss of PEAT modestly increases bone mor- addition to the previously described transcription factors, we identi- phogenetic protein target gene expression and also elevates the fied an uncharacterized noncoding RNA transcript that we named expression of a large subset of ribosomal protein mRNAs.
    [Show full text]
  • The Pennsylvania State University the Graduate School College Of
    The Pennsylvania State University The Graduate School College of Agricultural Sciences GENOMIC AND IMMUNOLOGICAL APPROACHES TO CONTROL GRAM- NEGATIVE INTRAMAMMARY INFECTIONS IN DAIRY CATTLE A Thesis in Animal Science by Annapoorani Chockalingam © 2006 Annapoorani Chockalingam Submitted in Partial Fulfillment of the Requirements for the Degree of Doctor of Philosophy May 2006 The thesis of Annapoorani Chockalingam was reviewed and approved* by the following: Cindy E. McKinney Assistant Professor of Transgenic Biology Thesis Advisor Co-Chair of Committee Douglas D. Bannerman Research Scientist, BFGL-ANRI, BARC, USDA-ARS Co-Chair of Committee Guy F. Barbato Associate Professor of Poultry Science Kenneth M. Weiss Evan Pugh Professor of Anthropology and Genetics Eric T. Harvill Assistant Professor of Veterinary Science Robert F. Paulson Associate Professor of Veterinary Science Terry D. Etherton Distinguished Professor of Animal Nutrition Head of the Department of Dairy and Animal Science *Signatures are on file in the Graduate School iii ABSTRACT The incidence of mastitis due to Gram-negative bacteria continues to impact the dairy industry due to the ubiquitous nature of the pathogens. Gram-negative bacteria account for nearly 40% of clinical mastitis cases resulting in an estimated loss to the U.S. dairy industry of $800 million per year. The research presented here focuses on host- mediated genetic and immunological factors involved in Gram-negative bacteria induced mastitis during the early phases of disease onset. Array analysis of mammary gland gene expression in a global context in response to E. coli is currently unavailable. A mouse mastitis model was used to study the transcriptional response in the mammary gland.
    [Show full text]
  • Metabolic and Behavioral Effects of Mutant Huntingtin Deletion in Sim1
    www.nature.com/scientificreports OPEN Metabolic and behavioral effects of mutant huntingtin deletion in Sim1 neurons in the BACHD mouse Received: 08 April 2016 Accepted: 31 May 2016 model of Huntington’s disease Published: 23 June 2016 Rana Soylu-Kucharz, Barbara Baldo & Åsa Petersén Hypothalamic pathology, metabolic dysfunction and psychiatric symptoms are part of Huntington disease (HD), which is caused by an expanded CAG repeat in the huntingtin (HTT) gene. Inactivation of mutant HTT selectively in the hypothalamus prevents the development of metabolic dysfunction and depressive-like behavior in the BACHD mouse model. The hypothalamic paraventricular nucleus (PVN) is implicated in metabolic and emotional control, therefore we here tested whether inactivation of mutant HTT in the PVN affects metabolic and psychiatric manifestations of HD in BACHD mice. BACHD mice were crossed with mice expressing Cre-recombinase under the Sim1 promoter (Sim1- Cre) to inactivate mutant HTT in Sim1 expressing cells, i.e. the PVN of the hypothalamus. We found that inactivation of mutant HTT in Sim1 cells had a sex-specific effect on both the metabolic and the psychiatric phenotype, as these phenotypes were no longer different in male BACHD/Sim1-Cre mice compared to wild-type littermates. We also found a reduced number of GnRH neurons specifically in the anterior hypothalamus and an increased testes weight in male BACHD mice compared to wild- type littermates. Taken together, expression of mutant HTT in Sim1 cells may play a role for the development of metabolic dysfunction and depressive-like behavior in male BACHD mice. Huntington’s disease (HD) is a neurodegenerative disorder with formal diagnosis based on prominent motor symptoms and a positive gene test for an expansion of the CAG repeat in the huntingtin (HTT) gene1,2.
    [Show full text]
  • Ventral Neural Patterning by Nkx Homeobox Genes: Nkx6.1 Controls Somatic Motor Neuron and Ventral Interneuron Fates
    Downloaded from genesdev.cshlp.org on September 30, 2021 - Published by Cold Spring Harbor Laboratory Press RESEARCH COMMUNICATION vitro in response to two- to threefold changes in the Ventral neural patterning by concentration of Shh, and the position at which each Nkx homeobox genes: Nkx6.1 neuronal class is generated in vivo is predicted by the controls somatic motor neuron concentration required for their induction in vivo (Eric- son et al. 1997a,b; Briscoe et al. 2000). Thus, neurons and ventral interneuron fates generated in more ventral regions of the neural tube re- 1,2,6 3,6 quire progressively higher concentrations of Shh for their Maike Sander, Sussan Paydar, induction. 4,5 4 Johan Ericson, James Briscoe, The genetic programs activated in neural progenitor 3 1 Elizabeth Berber, Michael German, cells in response to Shh signaling, however, remain in- Thomas M. Jessell,4 and John L.R. Rubenstein3,7 completely defined. Emerging evidence suggests that homeobox genes function as critical intermediaries in 1Hormone Research Institute, Department of Medicine, the neural response to Shh signals (Lumsden and Krum- University of California–San Francisco, San Franscisco, California 94143 USA; 2Center for Molecular Neurobiology, lauf 1996; Tanabe and Jessell 1996; Ericson et al. 1997a,b; Hamburg, Germany; 3Nina Ireland Laboratory of Hammerschmidt et al. 1997; Rubenstein et al. 1998). Developmental Neurobiology, University of California–San Several homeobox genes are expressed by ventral pro- Francisco, San Francisco, California 94143 USA;
    [Show full text]