1
|
Nunes EA, Silva HCD, Duarte NDAA, de Lima LE, Maraslis FT, Araújo MLD, Pedron T, Lange C, Freire BM, Matias AC, Batista BL, Barcelos GRM. Impact of DNA repair polymorphisms on DNA instability biomarkers induced by lead (Pb) in workers exposed to the metal. CHEMOSPHERE 2023:138897. [PMID: 37182709 DOI: 10.1016/j.chemosphere.2023.138897] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Revised: 05/05/2023] [Accepted: 05/07/2023] [Indexed: 05/16/2023]
Abstract
Although the mechanisms of Pb-induced genotoxicity are well established, a wide individual's variation response is seen in biomarkers related to Pb toxicity, despite similar levels of metal exposure. This may be related to intrinsic variations, such as genetic polymorphisms; moreover, very little is known about the impact of genetic variations related to DNA repair system on DNA instability induced by Pb. In this context, the present study aimed to assess the impact of SNPs in enzymes related to DNA repair system on biomarkers related to acute toxicity and DNA damage induced by Pb exposure, in individuals occupationally exposed to the metal. A cross-sectional study was run with 154 adults (males, >18 years) from an automotive batteries' factory, in Brazil. Blood lead levels (BLL) were determined by ICP-MS; biomarkers related to acute toxicity and DNA instability were monitored by the buccal micronucleus cytome (BMNCyt) assay and genotyping of polymorphisms of MLH1 (rs1799977), OGG1 (rs1052133), PARP1 (rs1136410), XPA (rs1800975), XPC (rs2228000) and XRCC1 (rs25487) were performed by TaqMan assays. BLL ranged from 2.0 to 51 μg dL-1 (mean 20 ± 12 μg dL-1) and significant associations between BLL and BMNCyt biomarkers related to cellular proliferation and cytokinetic, cell death and DNA damage were observed. Furthermore, SNPs from the OGG1, XPA and XPC genes were able to modulate interactions in nuclear bud formation (NBUDs) and micronucleus (MNi) events. Taken together, our data provide further evidence that polymorphisms related to DNA repair pathways may modulate Pb-induced DNA damage; studies that investigate the association between injuries to genetic material and susceptibilities in the workplace can provide additional information on the etiology of diseases and the determination of environmentally responsive genes.
Collapse
Affiliation(s)
- Emilene Arusievicz Nunes
- Department of Biosciences, Institute of Health and Society, Federal University of São Paulo, Rua XV de Novembro 195, CEP 11.060-001, Santos, Brazil.
| | - Heliton Camargo da Silva
- Department of Biosciences, Institute of Health and Society, Federal University of São Paulo, Rua XV de Novembro 195, CEP 11.060-001, Santos, Brazil.
| | - Nathália de Assis Aguilar Duarte
- Department of Biosciences, Institute of Health and Society, Federal University of São Paulo, Rua XV de Novembro 195, CEP 11.060-001, Santos, Brazil.
| | - Lindiane Eloisa de Lima
- Department of Biosciences, Institute of Health and Society, Federal University of São Paulo, Rua XV de Novembro 195, CEP 11.060-001, Santos, Brazil.
| | - Flora Troina Maraslis
- Department of Biosciences, Institute of Health and Society, Federal University of São Paulo, Rua XV de Novembro 195, CEP 11.060-001, Santos, Brazil.
| | - Marília Ladeira de Araújo
- Department of Biosciences, Institute of Health and Society, Federal University of São Paulo, Rua XV de Novembro 195, CEP 11.060-001, Santos, Brazil.
| | - Tatiana Pedron
- Center of Natural and Human Sciences, Federal University of ABC, Avenida Dos Estados, 5001, CEP 09210-580, Santo André, Brazil.
| | - Camila Lange
- Center of Natural and Human Sciences, Federal University of ABC, Avenida Dos Estados, 5001, CEP 09210-580, Santo André, Brazil.
| | - Bruna Moreira Freire
- Center of Natural and Human Sciences, Federal University of ABC, Avenida Dos Estados, 5001, CEP 09210-580, Santo André, Brazil.
| | - Andreza Cândido Matias
- Department of Cell and Developmental Biology, Institute of Biomedical Sciences, University of São Paulo, Avenida Professor Lineu Prestes, 1524, CEP 05508-000, São Paulo, Brazil.
| | - Bruno Lemos Batista
- Center of Natural and Human Sciences, Federal University of ABC, Avenida Dos Estados, 5001, CEP 09210-580, Santo André, Brazil.
| | - Gustavo Rafael Mazzaron Barcelos
- Department of Biosciences, Institute of Health and Society, Federal University of São Paulo, Rua XV de Novembro 195, CEP 11.060-001, Santos, Brazil.
| |
Collapse
|
2
|
Aspatwar A, Supuran CT, Waheed A, Sly WS, Parkkila S. Mitochondrial carbonic anhydrase VA and VB: properties and roles in health and disease. J Physiol 2023; 601:257-274. [PMID: 36464834 PMCID: PMC10107955 DOI: 10.1113/jp283579] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2022] [Accepted: 11/30/2022] [Indexed: 12/07/2022] Open
Abstract
Carbonic anhydrase V (CA V), a mitochondrial enzyme, was first isolated from guinea-pig liver and subsequently identified in mice and humans. Later, studies revealed that the mouse genome contains two mitochondrial CA sequences, named Car5A and Car5B. The CA VA enzyme is most highly expressed in the liver, whereas CA VB shows a broad tissue distribution. Car5A knockout mice demonstrated a predominant role for CA VA in ammonia detoxification, whereas the roles of CA VB in ureagenesis and gluconeogenesis were evident only in the absence of CA VA. Previous studies have suggested that CA VA is mainly involved in the provision of HCO3 - for biosynthetic processes. In children, mutations in the CA5A gene led to reduced CA activity, and the enzyme was sensitive to increased temperature. The metabolic profiles of these children showed a reduced supply of HCO3 - to the enzymes that take part in intermediary metabolism: carbamoylphosphate synthetase, pyruvate carboxylase, propionyl-CoA carboxylase and 3-methylcrotonyl-CoA carboxylase. Although the role of CA VB is still poorly understood, a recent study reported that it plays an essential role in human Sertoli cells, which sustain spermatogenesis. Metabolic disease associated with CA VA appears to be more common than other inborn errors of metabolism and responds well to treatment with N-carbamyl-l-glutamate. Therefore, early identification of hyperammonaemia will allow specific treatment with N-carbamyl-l-glutamate and prevent neurological sequelae. Carbonic anhydrase VA deficiency should therefore be considered a treatable condition in the differential diagnosis of hyperammonaemia in neonates and young children.
Collapse
Affiliation(s)
- Ashok Aspatwar
- Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland.,Fimlab Ltd and Tampere University Hospital, Tampere, Finland
| | - Claudiu T Supuran
- Neurofarba Department, Sezione di Chimica Farmaceutica e Nutraceutica, Università degli Studi di Firenze, Sesto Fiorentino, Firenze, Italy
| | - Abdul Waheed
- Department of Biochemistry and Molecular Biology, Edward A. Doisy Research Center, Saint Louis University School of Medicine, St Louis, MO, USA
| | - William S Sly
- Department of Biochemistry and Molecular Biology, Edward A. Doisy Research Center, Saint Louis University School of Medicine, St Louis, MO, USA
| | - Seppo Parkkila
- Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland.,Fimlab Ltd and Tampere University Hospital, Tampere, Finland
| |
Collapse
|
3
|
Beier S, Stiegler M, Hitzenhammer E, Monika S. Screening for genes involved in cellulase regulation by expression under the control of a novel constitutive promoter in Trichoderma reesei. CURRENT RESEARCH IN BIOTECHNOLOGY 2022. [DOI: 10.1016/j.crbiot.2022.04.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
|
4
|
Davis ES, Voss G, Miesfeld JB, Zarate-Sanchez J, Voss SR, Glaser T. The rax homeobox gene is mutated in the eyeless axolotl, Ambystoma mexicanum. Dev Dyn 2021; 250:807-821. [PMID: 32864847 PMCID: PMC8907009 DOI: 10.1002/dvdy.246] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2020] [Accepted: 08/11/2020] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND Vertebrate eye formation requires coordinated inductive interactions between different embryonic tissue layers, first described in amphibians. A network of transcription factors and signaling molecules controls these steps, with mutations causing severe ocular, neuronal, and craniofacial defects. In eyeless mutant axolotls, eye morphogenesis arrests at the optic vesicle stage, before lens induction, and development of ventral forebrain structures is disrupted. RESULTS We identified a 5-bp deletion in the rax (retina and anterior neural fold homeobox) gene, which was tightly linked to the recessive eyeless (e) axolotl locus in an F2 cross. This frameshift mutation, in exon 2, truncates RAX protein within the homeodomain (P154fs35X). Quantitative RNA analysis shows that mutant and wild-type rax transcripts are equally abundant in E/e embryos. Translation appears to initiate from dual start codons, via leaky ribosome scanning, a conserved feature among gnathostome RAX proteins. Previous data show rax is expressed in the optic vesicle and diencephalon, deeply conserved among metazoans, and required for eye formation in other species. CONCLUSION The eyeless axolotl mutation is a null allele in the rax homeobox gene, with primary defects in neural ectoderm, including the retinal and hypothalamic primordia.
Collapse
Affiliation(s)
- Erik S. Davis
- Department of Cell Biology and Human Anatomy, University of California Davis School of Medicine, Davis, California
| | - Gareth Voss
- Department of Neuroscience, Spinal Cord and Brain Injury Research Center, and Ambystoma Genetic Stock Center, University of Kentucky, Lexington, Kentucky
| | - Joel B. Miesfeld
- Department of Cell Biology and Human Anatomy, University of California Davis School of Medicine, Davis, California
| | - Juan Zarate-Sanchez
- Department of Cell Biology and Human Anatomy, University of California Davis School of Medicine, Davis, California
- Davis Senior High School, Davis, California
| | - S. Randal Voss
- Department of Neuroscience, Spinal Cord and Brain Injury Research Center, and Ambystoma Genetic Stock Center, University of Kentucky, Lexington, Kentucky
| | - Tom Glaser
- Department of Cell Biology and Human Anatomy, University of California Davis School of Medicine, Davis, California
| |
Collapse
|
5
|
Liu S, Yan L, Zhou X, Chen C, Wang D, Yuan G. Delayed-onset adrenal hypoplasia congenita and hypogonadotropic hypogonadism caused by a novel mutation in DAX1. J Int Med Res 2019; 48:300060519882151. [PMID: 31642359 PMCID: PMC7605007 DOI: 10.1177/0300060519882151] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
In this study, we described a male who presented with delayed-onset adrenal
hypoplasia congenita (AHC) and mild hypogonadotropic hypogonadism (HHG) without
a relevant family history. A novel mutation in the DAX1
(dosage-sensitive sex reversal, congenital adrenal hypoplasia critical region on
the X chromosome, gene 1) gene was shown to cause X-linked AHC and HHG. Genetic
analysis revealed a novel nonsense mutation, c.154G > T (p.Glu52Term), in the
DAX1 gene. Molecular testing demonstrated that the milder
phenotype caused by this mutation was due to expression of a partially
functional, amino-truncated DAX1 protein generated from an
alternate in-frame translation start site (methionine at codon 83). This unusual
case revealed a potential mechanism for a novel mutation that resulted in an
unusual delayed-onset mild clinical phenotype. It expands the spectrum of
adrenal hypoplasia congenita and hypogonadotropic hypogonadism.
Collapse
Affiliation(s)
- Siyue Liu
- Department of Internal Medicine, Tongji Hospital, Huazhong
University of Science and Technology, Wuhan, Hubei, P.R. China
| | - Libin Yan
- Department of Urology, Tongji Hospital, Huazhong University of
Science and Technology, Wuhan, Hubei, P.R. China
| | - Xinrong Zhou
- Department of Internal Medicine, Tongji Hospital, Huazhong
University of Science and Technology, Wuhan, Hubei, P.R. China
| | - Chen Chen
- Molecular Diagnostic Laboratory, Tongji Hospital, Huazhong
University of Science and Technology, Wuhan, Hubei, China
| | - Daowen Wang
- Department of Internal Medicine, Tongji Hospital, Huazhong
University of Science and Technology, Wuhan, Hubei, P.R. China
| | - Gang Yuan
- Department of Internal Medicine, Tongji Hospital, Huazhong
University of Science and Technology, Wuhan, Hubei, P.R. China
- Gang Yuan, Department of Internal Medicine,
Tongji Hospital, Tongji Medical College, Huazhong University of Science &
Technology, Wuhan 430030, China.
| |
Collapse
|
6
|
Geng H, Shi J, Fuerst EP, Wei J, Morris CF. Physical Mapping of Peroxidase Genes and Development of Functional Markers for TaPod-D1 on Bread Wheat Chromosome 7D. FRONTIERS IN PLANT SCIENCE 2019; 10:523. [PMID: 31068962 PMCID: PMC6491870 DOI: 10.3389/fpls.2019.00523] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/04/2019] [Accepted: 04/04/2019] [Indexed: 06/09/2023]
Abstract
Peroxidase (POD) activity in wheat (Triticum aestivum L.) grain influences natural carotenoid pigment content and is associated with the color of flour, and processing and product quality. Here, we report the molecular characterization and physical mapping of POD genes in bread wheat. The complete genomic DNA (gDNA) sequence of two POD genes (TaPod-A2 and TaPod-D1), and the partial gDNA sequence of two additional POD genes (TaPod-A3 and TaPod-B1) from wheat were characterized using in silico cloning and validated through laboratory experiments. Using a set of 21 nullisomic-tetrasomic (NT) lines, six group-7 ditelosomic (Dt) lines, and 38 group-7 deletion (Del) lines of Chinese Spring (CS), TaPod-A2 and TaPod-D1 were found to be physically located on 0.73-0.83 and on the most distal 0.39 fraction arm length (FL) of 7AS and 7DS in cv. CS, respectively; whereas, TaPod-A3 and TaPod-B1 were assigned to the 0.40-0.49 and 0.40-0.48 FL of 7AL and 7BL, respectively. Based on single nucleotide polymorphisms (SNPs) of two alleles at the TaPod-D1 locus, two functional markers POD-7D1 and POD-7D6 were developed, amplifying 540- and 640-bp, fragments in varieties with higher and lower POD activities, respectively. A total of 224 wheat varieties were analyzed and showed a significant association between the polymorphic fragments and POD activity using POD-7D1 and POD-7D6 markers. The analysis of variance (ANOVA) indicated the average POD activities of 115 varieties with TaPod-D1a were significantly lower than 109 varieties with TaPod-D1b (P < 0.01). This study provides useful information of the POD genes in bread wheat, insight into wheat genome synteny and structure, gene-specific markers, and contributes a valuable resource for quality improvement in wheat breeding programs.
Collapse
Affiliation(s)
- Hongwei Geng
- College of Agronomy, Xinjiang Agricultural University, Ürümqi, China
| | - Jia Shi
- College of Agronomy, Xinjiang Agricultural University, Ürümqi, China
| | - E. Patrick Fuerst
- Department of Crop and Soil Sciences, Washington State University, Pullman, WA, United States, Affiliated with the Western Wheat Quality Laboratory
| | - Jingxin Wei
- National Wheat Improvement Center, Institute of Crop Science, Chinese Academy of Agricultural Sciences (CAAS), Beijing, China
| | - Craig F. Morris
- USDA-ARS Western Wheat Quality Laboratory, E-202 Food Science and Human Nutrition Facility East, Washington State University, Pullman, WA, United States
| |
Collapse
|
7
|
Li A, Sun Y, Drummer C, Lu Y, Yu D, Zhou Y, Li X, Pearson SJ, Johnson C, Yu C, Yang WY, Mastascusa K, Jiang X, Sun J, Rogers T, Hu W, Wang H, Yang X. Increasing Upstream Chromatin Long-Range Interactions May Favor Induction of Circular RNAs in LysoPC-Activated Human Aortic Endothelial Cells. Front Physiol 2019; 10:433. [PMID: 31057422 PMCID: PMC6482593 DOI: 10.3389/fphys.2019.00433] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2018] [Accepted: 03/28/2019] [Indexed: 01/10/2023] Open
Abstract
Circular RNAs (circRNAs) are non-coding RNAs that form covalently closed continuous loops, and act as gene regulators in physiological and disease conditions. To test our hypothesis that proatherogenic lipid lysophosphatidylcholine (LPC) induce a set of circRNAs in human aortic endothelial cell (HAEC) activation, we performed circRNA analysis by searching our RNA-Seq data from LPC-activated HAECs, and found: (1) LPC induces significant modulation of 77 newly characterized cirRNAs, among which 47 circRNAs (61%) are upregulated; (2) 34 (72%) out of 47 upregulated circRNAs are upregulated when the corresponding mRNAs are downregulated, suggesting that the majority of circRNAs are upregulated presumably via LPC-induced “abnormal splicing” when the canonical splicing for generation of corresponding mRNAs is suppressed; (3) Upregulation of 47 circRNAs is temporally associated with mRNAs-mediated LPC-upregulated cholesterol synthesis-SREBP2 pathway and LPC-downregulated TGF-β pathway; (4) Increase in upstream chromatin long-range interaction sites to circRNA related genes is associated with preferred circRNA generation over canonical splicing for mRNAs, suggesting that shifting chromatin long-range interaction sites from downstream to upstream may promote induction of a list of circRNAs in lysoPC-activated HAECs; (5) Six significantly changed circRNAs may have sponge functions for miRNAs; and (6) 74% significantly changed circRNAs contain open reading frames, suggesting that putative short proteins may interfere with the protein interaction-based signaling. Our findings have demonstrated for the first time that a new set of LPC-induced circRNAs may contribute to homeostasis in LPC-induced HAEC activation. These novel insights may lead to identifications of new therapeutic targets for treating metabolic cardiovascular diseases, inflammations, and cancers.
Collapse
Affiliation(s)
- Angus Li
- Center for Metabolic Disease Research, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, United States.,Department of Biomedical Engineering, Pratt School of Engineering, Duke University, Durham, NC, United States
| | - Yu Sun
- Center for Metabolic Disease Research, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, United States
| | - Charles Drummer
- Center for Metabolic Disease Research, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, United States
| | - Yifan Lu
- Center for Metabolic Disease Research, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, United States
| | - Daohai Yu
- Department of Clinical Sciences, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, United States
| | - Yan Zhou
- Biostatistics and Bioinformatics Facility, Fox Chase Cancer Center, Temple Health, Philadelphia, PA, United States
| | - Xinyuan Li
- Center for Metabolic Disease Research, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, United States
| | - Simone J Pearson
- Center for Metabolic Disease Research, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, United States
| | - Candice Johnson
- Center for Metabolic Disease Research, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, United States
| | - Catherine Yu
- Geisinger Commonwealth School of Medicine, Scranton, PA, United States
| | - William Y Yang
- Center for Metabolic Disease Research, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, United States
| | - Kevin Mastascusa
- Center for Metabolic Disease Research, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, United States
| | - Xiaohua Jiang
- Center for Metabolic Disease Research, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, United States
| | - Jianxin Sun
- Center for Translational Medicine, Department of Medicine, Sidney Kimmel Medical College, Philadelphia University - Thomas Jefferson University, Philadelphia, PA, United States
| | - Thomas Rogers
- Center for Inflammation, Translational and Clinical Lung Research, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, United States
| | - Wenhui Hu
- Center for Metabolic Disease Research, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, United States
| | - Hong Wang
- Center for Metabolic Disease Research, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, United States
| | - Xiaofeng Yang
- Center for Metabolic Disease Research, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, United States.,Center for Inflammation, Translational and Clinical Lung Research, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, United States
| |
Collapse
|
8
|
Watson Levings RS, Broome TA, Smith AD, Rice BL, Gibbs EP, Myara DA, Hyddmark EV, Nasri E, Zarezadeh A, Levings PP, Lu Y, White ME, Dacanay EA, Foremny GB, Evans CH, Morton AJ, Winter M, Dark MJ, Nickerson DM, Colahan PT, Ghivizzani SC. Gene Therapy for Osteoarthritis: Pharmacokinetics of Intra-Articular Self-Complementary Adeno-Associated Virus Interleukin-1 Receptor Antagonist Delivery in an Equine Model. HUM GENE THER CL DEV 2019; 29:90-100. [PMID: 29869540 DOI: 10.1089/humc.2017.142] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Toward the treatment of osteoarthritis (OA), the authors have been investigating self-complementary adeno-associated virus (scAAV) for intra-articular delivery of therapeutic gene products. As OA frequently affects weight-bearing joints, pharmacokinetic studies of scAAV gene delivery were performed in the joints of the equine forelimb to identify parameters relevant to clinical translation in humans. Using interleukin-1 receptor antagonist (IL-1Ra) as a secreted therapeutic reporter, scAAV vector plasmids containing codon-optimized cDNA for equine IL-1Ra (eqIL-1Ra) were generated, which produced eqIL-1Ra at levels 30- to 50-fold higher than the native sequence. The most efficient cDNA was packaged in AAV2.5 capsid, and following characterization in vitro, the virus was injected into the carpal and metacarpophalangeal joints of horses over a 100-fold dose range. A putative ceiling dose of 5 × 1012 viral genomes was identified that elevated the steady-state eqIL-1Ra in the synovial fluids of injected joints by >40-fold over endogenous levels and was sustained for at least 6 months. No adverse effects were seen, and eqIL-1Ra in serum and urine remained at background levels throughout. Using the 5 × 1012 viral genome dose of scAAV, and green fluorescent protein as a cytologic marker, the local and systemic distribution of vector and transduced cells following intra-articular injection scAAV.GFP were compared in healthy equine joints and in those with late-stage, naturally occurring OA. In both cases, 99.7% of the vector remained within the injected joint. Strikingly, the pathologies characteristic of OA (synovitis, osteophyte formation, and cartilage erosion) were associated with a substantial increase in transgenic expression relative to tissues in healthy joints. This was most notable in regions of articular cartilage with visible damage, where foci of brilliantly fluorescent chondrocytes were observed. Overall, these data suggest that AAV-mediated gene transfer can provide relatively safe, sustained protein drug delivery to joints of human proportions.
Collapse
Affiliation(s)
| | - Ted A Broome
- 2 Department of Large Animal Clinical Sciences, University of Florida , Gainesville, Florida
| | - Andrew D Smith
- 2 Department of Large Animal Clinical Sciences, University of Florida , Gainesville, Florida
| | - Brett L Rice
- 2 Department of Large Animal Clinical Sciences, University of Florida , Gainesville, Florida
| | - Eric P Gibbs
- 1 Department of Orthopedics and Rehabilitation, University of Florida , Gainesville, Florida
| | - David A Myara
- 1 Department of Orthopedics and Rehabilitation, University of Florida , Gainesville, Florida
| | - E Viktoria Hyddmark
- 1 Department of Orthopedics and Rehabilitation, University of Florida , Gainesville, Florida
| | - Elham Nasri
- 1 Department of Orthopedics and Rehabilitation, University of Florida , Gainesville, Florida
| | - Ali Zarezadeh
- 1 Department of Orthopedics and Rehabilitation, University of Florida , Gainesville, Florida
| | - Padraic P Levings
- 1 Department of Orthopedics and Rehabilitation, University of Florida , Gainesville, Florida
| | - Yuan Lu
- 1 Department of Orthopedics and Rehabilitation, University of Florida , Gainesville, Florida
| | - Margaret E White
- 1 Department of Orthopedics and Rehabilitation, University of Florida , Gainesville, Florida
| | - E Anthony Dacanay
- 1 Department of Orthopedics and Rehabilitation, University of Florida , Gainesville, Florida
| | - Gregory B Foremny
- 1 Department of Orthopedics and Rehabilitation, University of Florida , Gainesville, Florida
| | - Christopher H Evans
- 3 Rehabilitation Medicine Research Center, Mayo Clinic , Rochester, Minnesota
| | - Alison J Morton
- 2 Department of Large Animal Clinical Sciences, University of Florida , Gainesville, Florida
| | - Mathew Winter
- 4 Department of Small Animal Clinical Sciences, University of Florida , Gainesville, Florida
| | - Michael J Dark
- 5 Department of Infectious Diseases and Pathology, University of Florida , Gainesville, Florida
| | - David M Nickerson
- 6 Department of Statistics and Actuarial Science, University of Central Florida , Orlando, Florida
| | - Patrick T Colahan
- 2 Department of Large Animal Clinical Sciences, University of Florida , Gainesville, Florida
| | - Steven C Ghivizzani
- 1 Department of Orthopedics and Rehabilitation, University of Florida , Gainesville, Florida
| |
Collapse
|
9
|
Zmajkovic J, Lundberg P, Nienhold R, Torgersen ML, Sundan A, Waage A, Skoda RC. A Gain-of-Function Mutation in EPO in Familial Erythrocytosis. N Engl J Med 2018. [PMID: 29514032 DOI: 10.1056/nejmoa1709064] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Familial erythrocytosis with elevated erythropoietin levels is frequently caused by mutations in genes that regulate oxygen-dependent transcription of the gene encoding erythropoietin ( EPO). We identified a mutation in EPO that cosegregated with disease with a logarithm of the odds (LOD) score of 3.3 in a family with autosomal dominant erythrocytosis. This mutation, a single-nucleotide deletion (c.32delG), introduces a frameshift in exon 2 that interrupts translation of the main EPO messenger RNA (mRNA) transcript but initiates excess production of erythropoietin from what is normally a noncoding EPO mRNA transcribed from an alternative promoter located in intron 1. (Funded by the Gebert Rüf Foundation and others.).
Collapse
Affiliation(s)
- Jakub Zmajkovic
- From the Department of Biomedicine, Experimental Hematology, University Hospital Basel and University of Basel (J.Z., P.L., R.N., R.C.S.), and Diagnostic Hematology, University Hospital Basel (P.L.), Basel, Switzerland; and the Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology (M.L.T., A.S., A.W.), and the Department of Hematology, St. Olavs Hospital (A.W.) - both in Trondheim, Norway
| | - Pontus Lundberg
- From the Department of Biomedicine, Experimental Hematology, University Hospital Basel and University of Basel (J.Z., P.L., R.N., R.C.S.), and Diagnostic Hematology, University Hospital Basel (P.L.), Basel, Switzerland; and the Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology (M.L.T., A.S., A.W.), and the Department of Hematology, St. Olavs Hospital (A.W.) - both in Trondheim, Norway
| | - Ronny Nienhold
- From the Department of Biomedicine, Experimental Hematology, University Hospital Basel and University of Basel (J.Z., P.L., R.N., R.C.S.), and Diagnostic Hematology, University Hospital Basel (P.L.), Basel, Switzerland; and the Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology (M.L.T., A.S., A.W.), and the Department of Hematology, St. Olavs Hospital (A.W.) - both in Trondheim, Norway
| | - Maria L Torgersen
- From the Department of Biomedicine, Experimental Hematology, University Hospital Basel and University of Basel (J.Z., P.L., R.N., R.C.S.), and Diagnostic Hematology, University Hospital Basel (P.L.), Basel, Switzerland; and the Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology (M.L.T., A.S., A.W.), and the Department of Hematology, St. Olavs Hospital (A.W.) - both in Trondheim, Norway
| | - Anders Sundan
- From the Department of Biomedicine, Experimental Hematology, University Hospital Basel and University of Basel (J.Z., P.L., R.N., R.C.S.), and Diagnostic Hematology, University Hospital Basel (P.L.), Basel, Switzerland; and the Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology (M.L.T., A.S., A.W.), and the Department of Hematology, St. Olavs Hospital (A.W.) - both in Trondheim, Norway
| | - Anders Waage
- From the Department of Biomedicine, Experimental Hematology, University Hospital Basel and University of Basel (J.Z., P.L., R.N., R.C.S.), and Diagnostic Hematology, University Hospital Basel (P.L.), Basel, Switzerland; and the Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology (M.L.T., A.S., A.W.), and the Department of Hematology, St. Olavs Hospital (A.W.) - both in Trondheim, Norway
| | - Radek C Skoda
- From the Department of Biomedicine, Experimental Hematology, University Hospital Basel and University of Basel (J.Z., P.L., R.N., R.C.S.), and Diagnostic Hematology, University Hospital Basel (P.L.), Basel, Switzerland; and the Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology (M.L.T., A.S., A.W.), and the Department of Hematology, St. Olavs Hospital (A.W.) - both in Trondheim, Norway
| |
Collapse
|
10
|
Biswas R, Gao S, Cultraro CM, Maity TK, Venugopalan A, Abdullaev Z, Shaytan AK, Carter CA, Thomas A, Rajan A, Song Y, Pitts S, Chen K, Bass S, Boland J, Hanada KI, Chen J, Meltzer PS, Panchenko AR, Yang JC, Pack S, Giaccone G, Schrump DS, Khan J, Guha U. Genomic profiling of multiple sequentially acquired tumor metastatic sites from an "exceptional responder" lung adenocarcinoma patient reveals extensive genomic heterogeneity and novel somatic variants driving treatment response. Cold Spring Harb Mol Case Stud 2017; 2:a001263. [PMID: 27900369 PMCID: PMC5111000 DOI: 10.1101/mcs.a001263] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
We used next-generation sequencing to identify somatic alterations in multiple metastatic sites from an “exceptional responder” lung adenocarcinoma patient during his 7-yr course of ERBB2-directed therapies. The degree of heterogeneity was unprecedented, with ∼1% similarity between somatic alterations of the lung and lymph nodes. One novel translocation, PLAG1-ACTA2, present in both sites, up-regulated ACTA2 expression. ERBB2, the predominant driver oncogene, was amplified in both sites, more pronounced in the lung, and harbored an L869R mutation in the lymph node. Functional studies showed increased proliferation, migration, metastasis, and resistance to ERBB2-directed therapy because of L869R mutation and increased migration because of ACTA2 overexpression. Within the lung, a nonfunctional CDK12, due to a novel G879V mutation, correlated with down-regulation of DNA damage response genes, causing genomic instability, and sensitivity to chemotherapy. We propose a model whereby a subclone metastasized early from the primary site and evolved independently in lymph nodes.
Collapse
Affiliation(s)
- Romi Biswas
- Thoracic and Gastrointestinal Oncology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland 20892, USA
| | - Shaojian Gao
- Thoracic and Gastrointestinal Oncology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland 20892, USA
| | - Constance M Cultraro
- Thoracic and Gastrointestinal Oncology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland 20892, USA
| | - Tapan K Maity
- Thoracic and Gastrointestinal Oncology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland 20892, USA
| | - Abhilash Venugopalan
- Thoracic and Gastrointestinal Oncology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland 20892, USA
| | - Zied Abdullaev
- Laboratory of Pathology, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland 20892, USA
| | - Alexey K Shaytan
- National Center for Biotechnology Information, National Library of Medicine, National Institutes of Health, Bethesda, Maryland 20894, USA
| | - Corey A Carter
- Walter Reed National Military Medical Center, Bethesda, Maryland 20889, USA
| | - Anish Thomas
- Thoracic and Gastrointestinal Oncology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland 20892, USA
| | - Arun Rajan
- Thoracic and Gastrointestinal Oncology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland 20892, USA
| | - Young Song
- Genetics Branch, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland 20892, USA
| | - Stephanie Pitts
- Thoracic and Gastrointestinal Oncology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland 20892, USA
| | - Kevin Chen
- Thoracic and Gastrointestinal Oncology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland 20892, USA
| | - Sara Bass
- Cancer Genomics Research Laboratory, Division of Cancer Epidemiology and Genetics, National Cancer Institute, Gaithersburg, Maryland 20848, USA
| | - Joseph Boland
- Cancer Genomics Research Laboratory, Division of Cancer Epidemiology and Genetics, National Cancer Institute, Gaithersburg, Maryland 20848, USA
| | - Ken-Ichi Hanada
- Surgery Branch, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland 20892, USA
| | - Jinqiu Chen
- Laboratory of Cell Biology, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland 20892, USA
| | - Paul S Meltzer
- Genetics Branch, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland 20892, USA
| | - Anna R Panchenko
- National Center for Biotechnology Information, National Library of Medicine, National Institutes of Health, Bethesda, Maryland 20894, USA
| | - James C Yang
- Surgery Branch, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland 20892, USA
| | - Svetlana Pack
- Laboratory of Pathology, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland 20892, USA
| | - Giuseppe Giaccone
- Lombardi Comprehensive Cancer Center, Georgetown University, Washington, D.C. 20057, USA
| | - David S Schrump
- Thoracic and Gastrointestinal Oncology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland 20892, USA
| | - Javed Khan
- Genetics Branch, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland 20892, USA
| | - Udayan Guha
- Thoracic and Gastrointestinal Oncology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland 20892, USA
| |
Collapse
|
11
|
Single nucleotide polymorphisms in DNA repair genes and putative cancer risk. Arch Toxicol 2016; 90:2369-88. [PMID: 27334373 DOI: 10.1007/s00204-016-1771-2] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2016] [Accepted: 06/15/2016] [Indexed: 02/07/2023]
Abstract
Single nucleotide polymorphisms (SNPs) are the most frequent type of genetic alterations between individuals. An SNP located within the coding sequence of a gene may lead to an amino acid substitution and in turn might alter protein function. Such a change in protein sequence could be functionally relevant and therefore might be associated with susceptibility to human diseases, such as cancer. DNA repair mechanisms are known to play an important role in cancer development, as shown in various human cancer syndromes, which arise due to mutations in DNA repair genes. This leads to the question whether subtle genetic changes such as SNPs in DNA repair genes may contribute to cancer susceptibility. In numerous epidemiological studies, efforts have been made to associate specific SNPs in DNA repair genes with altered DNA repair and cancer. The present review describes some of the common and most extensively studied SNPs in DNA repair genes and discusses whether they are functionally relevant and subsequently increase the likelihood that cancer will develop.
Collapse
|
12
|
Nik-Zainal S, Davies H, Staaf J, Ramakrishna M, Glodzik D, Zou X, Martincorena I, Alexandrov LB, Martin S, Wedge DC, Van Loo P, Ju YS, Smid M, Brinkman AB, Morganella S, Aure MR, Lingjærde OC, Langerød A, Ringnér M, Ahn SM, Boyault S, Brock JE, Broeks A, Butler A, Desmedt C, Dirix L, Dronov S, Fatima A, Foekens JA, Gerstung M, Hooijer GKJ, Jang SJ, Jones DR, Kim HY, King TA, Krishnamurthy S, Lee HJ, Lee JY, Li Y, McLaren S, Menzies A, Mustonen V, O’Meara S, Pauporté I, Pivot X, Purdie CA, Raine K, Ramakrishnan K, Rodríguez-González FG, Romieu G, Sieuwerts AM, Simpson PT, Shepherd R, Stebbings L, Stefansson OA, Teague J, Tommasi S, Treilleux I, Van den Eynden GG, Vermeulen P, Vincent-Salomon A, Yates L, Caldas C, van’t Veer L, Tutt A, Knappskog S, Tan BKT, Jonkers J, Borg Å, Ueno NT, Sotiriou C, Viari A, Futreal PA, Campbell PJ, Span PN, Van Laere S, Lakhani SR, Eyfjord JE, Thompson AM, Birney E, Stunnenberg HG, van de Vijver MJ, Martens JW, Børresen-Dale AL, Richardson AL, Kong G, Thomas G, Stratton MR. Landscape of somatic mutations in 560 breast cancer whole-genome sequences. Nature 2016; 534:47-54. [PMID: 27135926 PMCID: PMC4910866 DOI: 10.1038/nature17676] [Citation(s) in RCA: 1522] [Impact Index Per Article: 169.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2015] [Accepted: 03/17/2016] [Indexed: 02/06/2023]
Abstract
We analysed whole-genome sequences of 560 breast cancers to advance understanding of the driver mutations conferring clonal advantage and the mutational processes generating somatic mutations. We found that 93 protein-coding cancer genes carried probable driver mutations. Some non-coding regions exhibited high mutation frequencies, but most have distinctive structural features probably causing elevated mutation rates and do not contain driver mutations. Mutational signature analysis was extended to genome rearrangements and revealed twelve base substitution and six rearrangement signatures. Three rearrangement signatures, characterized by tandem duplications or deletions, appear associated with defective homologous-recombination-based DNA repair: one with deficient BRCA1 function, another with deficient BRCA1 or BRCA2 function, the cause of the third is unknown. This analysis of all classes of somatic mutation across exons, introns and intergenic regions highlights the repertoire of cancer genes and mutational processes operating, and progresses towards a comprehensive account of the somatic genetic basis of breast cancer.
Collapse
Affiliation(s)
- Serena Nik-Zainal
- Wellcome Trust Sanger Institute, Hinxton, Cambridge CB10 1SA, UK
- East Anglian Medical Genetics Service, Cambridge University Hospitals NHS Foundation Trust, Cambridge CB2 9NB, UK
| | - Helen Davies
- Wellcome Trust Sanger Institute, Hinxton, Cambridge CB10 1SA, UK
| | - Johan Staaf
- Division of Oncology and Pathology, Department of Clinical Sciences Lund, Lund University, Lund, Sweden
| | | | - Dominik Glodzik
- Wellcome Trust Sanger Institute, Hinxton, Cambridge CB10 1SA, UK
| | - Xueqing Zou
- Wellcome Trust Sanger Institute, Hinxton, Cambridge CB10 1SA, UK
| | | | - Ludmil B. Alexandrov
- Wellcome Trust Sanger Institute, Hinxton, Cambridge CB10 1SA, UK
- Theoretical Biology and Biophysics (T-6), Los Alamos National Laboratory, Los Alamos, New Mexico, United States of America
- Center for Nonlinear Studies, Los Alamos National Laboratory, Los Alamos, New Mexico, United States of America
| | - Sancha Martin
- Wellcome Trust Sanger Institute, Hinxton, Cambridge CB10 1SA, UK
| | - David C. Wedge
- Wellcome Trust Sanger Institute, Hinxton, Cambridge CB10 1SA, UK
| | - Peter Van Loo
- Wellcome Trust Sanger Institute, Hinxton, Cambridge CB10 1SA, UK
- Department of Human Genetics, University of Leuven, B-3000 Leuven, Belgium
| | - Young Seok Ju
- Wellcome Trust Sanger Institute, Hinxton, Cambridge CB10 1SA, UK
| | - Marcel Smid
- Erasmus MC Cancer Institute and Cancer Genomics Netherlands, Erasmus University Medical Center, Department of Medical Oncology, Rotterdam, The Netherlands
| | - Arie B Brinkman
- Radboud University, Department of Molecular Biology, Faculties of Science and Medicine, Nijmegen, Netherlands
| | - Sandro Morganella
- European Molecular Biology Laboratory, European Bioinformatics Institute, Wellcome Trust Genome Campus,Hinxton, Cambridgeshire, CB10 1SD
| | - Miriam R. Aure
- Department of Cancer Genetics, Institute for Cancer Research, Oslo University Hospital The Norwegian Radiumhospital
- K.G. Jebsen Centre for Breast Cancer Research, Institute for Clinical Medicine, University of Oslo, Oslo, Norway
| | - Ole Christian Lingjærde
- K.G. Jebsen Centre for Breast Cancer Research, Institute for Clinical Medicine, University of Oslo, Oslo, Norway
- Department of Computer Science, University of Oslo, Oslo, Norway
| | - Anita Langerød
- Department of Cancer Genetics, Institute for Cancer Research, Oslo University Hospital The Norwegian Radiumhospital
- K.G. Jebsen Centre for Breast Cancer Research, Institute for Clinical Medicine, University of Oslo, Oslo, Norway
| | - Markus Ringnér
- Division of Oncology and Pathology, Department of Clinical Sciences Lund, Lund University, Lund, Sweden
| | - Sung-Min Ahn
- Gachon Institute of Genome Medicine and Science, Gachon University Gil Medical Center, Incheon, South Korea
| | - Sandrine Boyault
- Translational Research Lab, Centre Léon Bérard, 28, rue Laënnec, 69373 Lyon Cedex 08, France
| | - Jane E. Brock
- Department of Pathology, Brigham and Women’s Hospital, Boston, MA 02115 USA
| | - Annegien Broeks
- The Netherlands Cancer Institute, 1066CX Amsterdam, The Netherlands
| | - Adam Butler
- Wellcome Trust Sanger Institute, Hinxton, Cambridge CB10 1SA, UK
| | - Christine Desmedt
- Breast Cancer Translational Research Laboratory, Université Libre de Bruxelles, Institut Jules Bordet, Bd de Waterloo 121, B-1000 Brussels, Belgium
| | - Luc Dirix
- Translational Cancer Research Unit, Center for Oncological Research, Faculty of Medicine and Health Sciences, University of Antwerp, Antwerp, Belgium
| | - Serge Dronov
- Wellcome Trust Sanger Institute, Hinxton, Cambridge CB10 1SA, UK
| | | | - John A. Foekens
- Erasmus MC Cancer Institute and Cancer Genomics Netherlands, Erasmus University Medical Center, Department of Medical Oncology, Rotterdam, The Netherlands
| | - Moritz Gerstung
- Wellcome Trust Sanger Institute, Hinxton, Cambridge CB10 1SA, UK
| | - Gerrit KJ Hooijer
- Department of Pathology, Academic Medical Center, Meibergdreef 9, 1105 AZ Amsterdam, the Netherlands
| | - Se Jin Jang
- Department of Pathology, Asan Medical Center, College of Medicine, Ulsan University, South Korea
| | - David R. Jones
- Wellcome Trust Sanger Institute, Hinxton, Cambridge CB10 1SA, UK
| | - Hyung-Yong Kim
- Department of Pathology, College of Medicine, Hanyang University, Seoul, South Korea
| | - Tari A. King
- Memorial Sloan Kettering Cancer Center, 1275 York Ave, New York, NY 10065, United States
| | - Savitri Krishnamurthy
- Morgan Welch Inflammatory Breast Cancer Research Program and Clinic, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd., Houston, TX 77030
| | - Hee Jin Lee
- Department of Pathology, Asan Medical Center, College of Medicine, Ulsan University, South Korea
| | - Jeong-Yeon Lee
- Institute for Bioengineering and Biopharmaceutical Research (IBBR), Hanyang University, Seoul, South Korea
| | - Yilong Li
- Wellcome Trust Sanger Institute, Hinxton, Cambridge CB10 1SA, UK
| | - Stuart McLaren
- Wellcome Trust Sanger Institute, Hinxton, Cambridge CB10 1SA, UK
| | - Andrew Menzies
- Wellcome Trust Sanger Institute, Hinxton, Cambridge CB10 1SA, UK
| | - Ville Mustonen
- Wellcome Trust Sanger Institute, Hinxton, Cambridge CB10 1SA, UK
| | - Sarah O’Meara
- Wellcome Trust Sanger Institute, Hinxton, Cambridge CB10 1SA, UK
| | - Iris Pauporté
- Institut National du Cancer, Research Division, Clinical Research Department, 52 avenue Morizet, 92513 Boulogne-Billancourt, France
| | - Xavier Pivot
- University Hospital of Minjoz, INSERM UMR 1098, Bd Fleming, Besançon 25000, France
| | - Colin A. Purdie
- Pathology Department, Ninewells Hospital & Medical School, Dundee DD1 9SY, UK
| | - Keiran Raine
- Wellcome Trust Sanger Institute, Hinxton, Cambridge CB10 1SA, UK
| | | | - F. Germán Rodríguez-González
- Erasmus MC Cancer Institute and Cancer Genomics Netherlands, Erasmus University Medical Center, Department of Medical Oncology, Rotterdam, The Netherlands
| | - Gilles Romieu
- Oncologie Sénologie, ICM Institut Régional du Cancer, Montpellier, France
| | - Anieta M. Sieuwerts
- Erasmus MC Cancer Institute and Cancer Genomics Netherlands, Erasmus University Medical Center, Department of Medical Oncology, Rotterdam, The Netherlands
| | - Peter T Simpson
- The University of Queensland: UQ Centre for Clinical Research and School of Medicine, Brisbane, Australia
| | - Rebecca Shepherd
- Wellcome Trust Sanger Institute, Hinxton, Cambridge CB10 1SA, UK
| | - Lucy Stebbings
- Wellcome Trust Sanger Institute, Hinxton, Cambridge CB10 1SA, UK
| | - Olafur A Stefansson
- Cancer Research Laboratory, Faculty of Medicine, University of Iceland, 101 Reykjavik, Iceland
| | - Jon Teague
- Wellcome Trust Sanger Institute, Hinxton, Cambridge CB10 1SA, UK
| | | | - Isabelle Treilleux
- Department of Pathology, Centre Léon Bérard, 28 rue Laënnec, 69373 Lyon Cédex 08, France
| | - Gert G. Van den Eynden
- Translational Cancer Research Unit, Center for Oncological Research, Faculty of Medicine and Health Sciences, University of Antwerp, Antwerp, Belgium
- Department of Pathology, GZA Hospitals Sint-Augustinus, Antwerp, Belgium
| | - Peter Vermeulen
- Translational Cancer Research Unit, Center for Oncological Research, Faculty of Medicine and Health Sciences, University of Antwerp, Antwerp, Belgium
- Department of Pathology, GZA Hospitals Sint-Augustinus, Antwerp, Belgium
| | - Anne Vincent-Salomon
- Institut Curie, Department of Pathology and INSERM U934, 26 rue d’Ulm, 75248 Paris Cedex 05, France
| | - Lucy Yates
- Wellcome Trust Sanger Institute, Hinxton, Cambridge CB10 1SA, UK
| | - Carlos Caldas
- Cancer Research UK Cambridge Institute, University of Cambridge, Li Ka Shing Centre, Robinson Way, Cambridge CB2 0RE, United Kingdom
| | - Laura van’t Veer
- The Netherlands Cancer Institute, 1066CX Amsterdam, The Netherlands
| | - Andrew Tutt
- Breast Cancer Now Toby Research Unit, King’s College London
- Breast Cancer Now Toby Robin’s Research Centre, Institute of Cancer Research, London
| | - Stian Knappskog
- Department of Clinical Science, University of Bergen, 5020 Bergen, Norway
- Department of Oncology, Haukeland University Hospital, 5021 Bergen, Norway
| | - Benita Kiat Tee Tan
- National Cancer Centre Singapore, 11 Hospital Drive, Singapore 169610
- Singapore General Hospital, Outram Road, Singapore 169608
| | - Jos Jonkers
- The Netherlands Cancer Institute, 1066CX Amsterdam, The Netherlands
| | - Åke Borg
- Division of Oncology and Pathology, Department of Clinical Sciences Lund, Lund University, Lund, Sweden
| | - Naoto T Ueno
- Morgan Welch Inflammatory Breast Cancer Research Program and Clinic, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd., Houston, TX 77030
| | - Christos Sotiriou
- Breast Cancer Translational Research Laboratory, Université Libre de Bruxelles, Institut Jules Bordet, Bd de Waterloo 121, B-1000 Brussels, Belgium
| | - Alain Viari
- Equipe Erable, INRIA Grenoble-Rhône-Alpes, 655, Av. de l’Europe, 38330 Montbonnot-Saint Martin, France
- Synergie Lyon Cancer, Centre Léon Bérard, 28 rue Laënnec, Lyon Cedex 08, France
| | - P. Andrew Futreal
- Wellcome Trust Sanger Institute, Hinxton, Cambridge CB10 1SA, UK
- Department of Genomic Medicine, UT MD Anderson Cancer Center, Houston, TX, 77230
| | - Peter J Campbell
- Wellcome Trust Sanger Institute, Hinxton, Cambridge CB10 1SA, UK
| | - Paul N. Span
- Department of Radiation Oncology, and department of Laboratory Medicine, Radboud university medical center, Nijmegen, the Netherlands
| | - Steven Van Laere
- Translational Cancer Research Unit, Center for Oncological Research, Faculty of Medicine and Health Sciences, University of Antwerp, Antwerp, Belgium
| | - Sunil R Lakhani
- The University of Queensland: UQ Centre for Clinical Research and School of Medicine, Brisbane, Australia
- Pathology Queensland, The Royal Brisbane and Women’s Hospital, Brisbane, Australia
| | - Jorunn E. Eyfjord
- Cancer Research Laboratory, Faculty of Medicine, University of Iceland, 101 Reykjavik, Iceland
| | - Alastair M. Thompson
- Morgan Welch Inflammatory Breast Cancer Research Program and Clinic, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd., Houston, TX 77030
- Department of Surgical Oncology, University of Texas MD Anderson Cancer Center, 1400 Pressler Street,Houston, Texas 77030
| | - Ewan Birney
- European Molecular Biology Laboratory, European Bioinformatics Institute, Wellcome Trust Genome Campus,Hinxton, Cambridgeshire, CB10 1SD
| | - Hendrik G Stunnenberg
- Radboud University, Department of Molecular Biology, Faculties of Science and Medicine, Nijmegen, Netherlands
| | - Marc J van de Vijver
- Department of Pathology, Academic Medical Center, Meibergdreef 9, 1105 AZ Amsterdam, the Netherlands
| | - John W.M. Martens
- Erasmus MC Cancer Institute and Cancer Genomics Netherlands, Erasmus University Medical Center, Department of Medical Oncology, Rotterdam, The Netherlands
| | - Anne-Lise Børresen-Dale
- Department of Cancer Genetics, Institute for Cancer Research, Oslo University Hospital The Norwegian Radiumhospital
- K.G. Jebsen Centre for Breast Cancer Research, Institute for Clinical Medicine, University of Oslo, Oslo, Norway
| | - Andrea L. Richardson
- Department of Pathology, Brigham and Women’s Hospital, Boston, MA 02115 USA
- Dana-Farber Cancer Institute, Boston, MA 02215 USA
| | - Gu Kong
- Department of Pathology, College of Medicine, Hanyang University, Seoul, South Korea
| | - Gilles Thomas
- Synergie Lyon Cancer, Centre Léon Bérard, 28 rue Laënnec, Lyon Cedex 08, France
| | | |
Collapse
|
13
|
Felker A, Mosimann C. Contemporary zebrafish transgenesis with Tol2 and application for Cre/lox recombination experiments. Methods Cell Biol 2016; 135:219-44. [PMID: 27443928 DOI: 10.1016/bs.mcb.2016.01.009] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/09/2022]
Abstract
Spatiotemporal transgene regulation by transgenic DNA recombinases is a central tool for reverse genetics in multicellular organisms, with excellent applications for misexpression and lineage tracing experiments. One of the most widespread technologies for this purpose is Cre recombinase-controlled lox site recombination that is attracting increasing interest in the zebrafish field. Tol2-mediated zebrafish transgenesis provides a stable platform to integrate lox cassette transgenes, while the amenability of the zebrafish embryo to drug treatments makes the model an ideal candidate for tamoxifen-inducible CreERT2 experiments. In addition, advanced transgenesis technologies such as phiC31 or CRISPR-Cas9-based knock-ins are even further promoting zebrafish transgenesis for Cre/lox applications. In this chapter, we will first introduce the basics of Cre/lox methodology, CreERT2 regulation by tamoxifen, as well as the utility of Tol2 and other contemporary transgenesis techniques for Cre/lox experiments. We will then outline in detail practical experimental steps for efficient transgenesis toward the creation of single-insertion transgenes and will introduce protocols for 4-hydroxytamoxifen-mediated CreERT2 induction to perform spatiotemporal lox transgene regulation experiments in zebrafish embryos. Last, we will discuss advanced experimental applications of Cre/lox beyond traditional lineage tracing approaches.
Collapse
Affiliation(s)
- A Felker
- University of Zürich, Zürich, Switzerland
| | - C Mosimann
- University of Zürich, Zürich, Switzerland
| |
Collapse
|
14
|
Pérez-Fernández J, Megías M, Pombal MA. Expression of a Novel D4 Dopamine Receptor in the Lamprey Brain. Evolutionary Considerations about Dopamine Receptors. Front Neuroanat 2016; 9:165. [PMID: 26778974 PMCID: PMC4701969 DOI: 10.3389/fnana.2015.00165] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2015] [Accepted: 12/08/2015] [Indexed: 12/28/2022] Open
Abstract
Numerous data reported in lampreys, which belong to the phylogenetically oldest branch of vertebrates, show that the dopaminergic system was already well developed at the dawn of vertebrate evolution. The expression of dopamine in the lamprey brain is well conserved when compared to other vertebrates, and this is also true for the D2 receptor. Additionally, the key role of dopamine in the striatum, modulating the excitability in the direct and indirect pathways through the D1 and D2 receptors, has also been recently reported in these animals. The moment of divergence regarding the two whole genome duplications occurred in vertebrates suggests that additional receptors, apart from the D1 and D2 previously reported, could be present in lampreys. We used in situ hybridization to characterize the expression of a novel dopamine receptor, which we have identified as a D4 receptor according to the phylogenetic analysis. The D4 receptor shows in the sea lamprey a more restricted expression pattern than the D2 subtype, as reported in mammals. Its main expression areas are the striatum, lateral and ventral pallial sectors, several hypothalamic regions, habenula, and mesencephalic and rhombencephalic motoneurons. Some expression areas are well conserved through vertebrate evolution, as is the case of the striatum or the habenula, but the controversies regarding the D4 receptor expression in other vertebrates hampers for a complete comparison, especially in rhombencephalic regions. Our results further support that the dopaminergic system in vertebrates is well conserved and suggest that at least some functions of the D4 receptor were already present before the divergence of lampreys.
Collapse
Affiliation(s)
- Juan Pérez-Fernández
- Neurolam Group, Department of Functional Biology and Health Sciences, Faculty of Biology - Centro de Investigaciones Biomédicas - Instituto de Investigación Biomédica de Vigo, Uiversity of Vigo Vigo, Spain
| | - Manuel Megías
- Neurolam Group, Department of Functional Biology and Health Sciences, Faculty of Biology - Centro de Investigaciones Biomédicas - Instituto de Investigación Biomédica de Vigo, Uiversity of Vigo Vigo, Spain
| | - Manuel A Pombal
- Neurolam Group, Department of Functional Biology and Health Sciences, Faculty of Biology - Centro de Investigaciones Biomédicas - Instituto de Investigación Biomédica de Vigo, Uiversity of Vigo Vigo, Spain
| |
Collapse
|
15
|
Lu WJ, Zhang CL, Wang XQ, Yang Q, Li ZM, Zhao GY. XPA A23G polymorphism and risk of esophageal cancer: A meta-analysis. Shijie Huaren Xiaohua Zazhi 2015; 23:5348-5355. [DOI: 10.11569/wcjd.v23.i33.5348] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
AIM: To perform a Meta-analysis to evaluate the relationship between the A23G polymorphism of the xeroderma pigmentosum group A (XPA) gene and susceptibility to esophageal cancer.
METHODS: PubMed, EMBASE, The Cochrane Library, Web of Science, CNKI, CBM, Wanfang, and VIP were searched for case-control studies evaluating the relationship between the A23G polymorphism of the XPA gene and susceptibility to esophageal cancer. Articles were assessed using pre-designed eligibility forms, according to the pre-defined eligibility criteria. Results were pooled using the STATA12.1 software to yield odds ratios (ORs) and 95% confidence intervals (CIs).
RESULTS: A total of nine case-control studies involving 2065 esophageal cancer cases and 3552 non-tumor controls were included. Meta-analysis suggested that susceptibility to esophageal cancer had no significant relationship with allele model (G vs A) (OR = 1.01, 95%CI: 0.76-1.34), dominant model (GA + AA vs GG) (OR = 0.87, 95%CI: 0.61-1.23), recessive model (AA vs GA + GG) (OR = 0.97, 95%CI: 0.66-1.42), or additive model (GG vs AA) (OR = 1.12, 95%CI: 0.65-1.92) of the XPA A23G polymorphism, but was significantly associated with the codominant model (GA vs AA + GG) (OR = 1.20, 95%CI: 1.07-1.35). The result of subgroup analysis based on ethnicity and source of controls showed that in Asian populations and source controlled populations, the codominant model (GA vs AA + GG) increased the risk of esophageal cancer (OR = 1.25, 95%CI: 1.10-1.44; OR = 1.26, 95%CI: 1.08-1.48).
CONCLUSION: Susceptibility to esophageal cancer has no significant relationship with allele model, dominant model, recessive model or additive model of the XPA A23G polymorphism, but significantly associates with the codominant model.
Collapse
|
16
|
Leon DR, Ytterberg AJ, Boontheung P, Kim U, Loo JA, Gunsalus RP, Ogorzalek Loo RR. Mining proteomic data to expose protein modifications in Methanosarcina mazei strain Gö1. Front Microbiol 2015; 6:149. [PMID: 25798134 PMCID: PMC4350412 DOI: 10.3389/fmicb.2015.00149] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2014] [Accepted: 02/09/2015] [Indexed: 12/11/2022] Open
Abstract
Proteomic tools identify constituents of complex mixtures, often delivering long lists of identified proteins. The high-throughput methods excel at matching tandem mass spectrometry data to spectra predicted from sequence databases. Unassigned mass spectra are ignored, but could, in principle, provide valuable information on unanticipated modifications and improve protein annotations while consuming limited quantities of material. Strategies to "mine" information from these discards are presented, along with discussion of features that, when present, provide strong support for modifications. In this study we mined LC-MS/MS datasets of proteolytically-digested concanavalin A pull down fractions from Methanosarcina mazei Gö1 cell lysates. Analyses identified 154 proteins. Many of the observed proteins displayed post-translationally modified forms, including O-formylated and methyl-esterified segments that appear biologically relevant (i.e., not artifacts of sample handling). Interesting cleavages and modifications (e.g., S-cyanylation and trimethylation) were observed near catalytic sites of methanogenesis enzymes. Of 31 Methanosarcina protein N-termini recovered by concanavalin A binding or from a previous study, only M. mazei S-layer protein MM1976 and its M. acetivorans C2A orthologue, MA0829, underwent signal peptide excision. Experimental results contrast with predictions from algorithms SignalP 3.0 and Exprot, which were found to over-predict the presence of signal peptides. Proteins MM0002, MM0716, MM1364, and MM1976 were found to be glycosylated, and employing chromatography tailored specifically for glycopeptides will likely reveal more. This study supplements limited, existing experimental datasets of mature archaeal N-termini, including presence or absence of signal peptides, translation initiation sites, and other processing. Methanosarcina surface and membrane proteins are richly modified.
Collapse
Affiliation(s)
- Deborah R Leon
- Department of Chemistry and Biochemistry, University of California, Los Angeles Los Angeles, CA, USA
| | - A Jimmy Ytterberg
- Department of Chemistry and Biochemistry, University of California, Los Angeles Los Angeles, CA, USA
| | - Pinmanee Boontheung
- Department of Chemistry and Biochemistry, University of California, Los Angeles Los Angeles, CA, USA
| | - Unmi Kim
- Microbiology, Immunology, and Molecular Genetics, University of California, Los Angeles Los Angeles, CA, USA
| | - Joseph A Loo
- Department of Chemistry and Biochemistry, University of California, Los Angeles Los Angeles, CA, USA ; Department of Biological Chemistry, David Geffen School of Medicine, University of California, Los Angeles Los Angeles, CA, USA ; UCLA-DOE Institute for Genomics and Proteomics, University of California, Los Angeles Los Angeles, CA, USA
| | - Robert P Gunsalus
- Microbiology, Immunology, and Molecular Genetics, University of California, Los Angeles Los Angeles, CA, USA ; UCLA-DOE Institute for Genomics and Proteomics, University of California, Los Angeles Los Angeles, CA, USA
| | - Rachel R Ogorzalek Loo
- Department of Biological Chemistry, David Geffen School of Medicine, University of California, Los Angeles Los Angeles, CA, USA ; UCLA-DOE Institute for Genomics and Proteomics, University of California, Los Angeles Los Angeles, CA, USA
| |
Collapse
|
17
|
He L, Deng T, Luo H. XPA A23G polymorphism and risk of digestive system cancers: a meta-analysis. Onco Targets Ther 2015; 8:385-94. [PMID: 25709470 PMCID: PMC4332261 DOI: 10.2147/ott.s75767] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Background Several studies have reported an association between the A23G polymorphism (rs 1800975) in the xeroderma pigmentosum group A (XPA) gene and risk of digestive system cancers. However, the results are inconsistent. In this study, we performed a meta-analysis to assess the association between XPA A23G polymorphism and the risk of digestive system cancers. Methods Relevant studies were identified using the PubMed, Web of Science, China National Knowledge Infrastructure, WanFang, and VIP databases up to August 30, 2014. The pooled odds ratio (OR) with a 95% confidence interval (CI) was calculated using the fixed or random effects model. Results A total of 18 case-control studies from 16 publications with 4,170 patients and 6,929 controls were included. Overall, no significant association was found between XPA A23G polymorphism and the risk of digestive system cancers (dominant model: GA + AA versus GG, OR 0.89, 95% CI 0.74–1.08; recessive model: AA versus GA + GG, OR 0.94, 95% CI 0.74–1.20; GA versus GG, OR 0.89, 95% CI 0.77–1.03; and AA versus GG, OR 0.87, 95% CI 0.64–1.19). When the analysis was stratified by ethnicity, similar results were observed among Asians and Caucasians in all genetic models. In stratified analysis based on tumor type, we also failed to detect any association between XPA A23G polymorphism and the risk of esophageal, gastric, or colorectal cancers. Conclusion This meta-analysis indicates that the XPA A23G polymorphism is not associated with a risk of digestive system cancers.
Collapse
Affiliation(s)
- Lei He
- Department of Gastroenterology, Renmin Hospital of Wuhan University, Wuhan, People's Republic of China
| | - Tao Deng
- Department of Gastroenterology, Renmin Hospital of Wuhan University, Wuhan, People's Republic of China
| | - Hesheng Luo
- Department of Gastroenterology, Renmin Hospital of Wuhan University, Wuhan, People's Republic of China
| |
Collapse
|
18
|
Lou Y, Li R, Zhang Y, Zhong R, Pei J, Xiong L, Zhang X, Han B. XPA gene rs1800975 single nucleotide polymorphism and lung cancer risk: a meta-analysis. Tumour Biol 2014; 35:6607-17. [PMID: 24696258 DOI: 10.1007/s13277-014-1824-1] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2013] [Accepted: 03/05/2014] [Indexed: 12/26/2022] Open
Abstract
No clear consensus has been reached on the XPA gene rs1800975 polymorphism and lung cancer risk. We performed a meta-analysis in an effort to systematically explore the possible association. We conducted a computer retrieval of PubMed, Embase, Wanfang, China National Knowledge Infrastructure Platform, and VIP databases prior to November 2013. References of retrieved articles were also screened. The fixed- and the random-effects model were applied for dichotomous outcomes to combine the results of the individual studies. According to the inclusion criteria, 10 articles (11 studies) were finally included. In overall, statistical association could be found between rs1800975 polymorphism and lung cancer in recessive genetic model [AA vs. (AG + GG): P = 0.02, OR = 1.16, 95% CI 1.02-1.31, P heterogeneity = 0.14, fixed-effects model]. In the East Asians, significant association was found in allele comparison model (A vs. G: P = 0.03, OR = 1.13, 95% CI 1.01-1.26, P heterogeneity = 0.39, fixed-effects model), in recessive genetic model [AA vs. (AG + GG): P = 0.005, OR = 1.30, 95% CI 1.08-1.56, P heterogeneity = 0.58, fixed-effects model] and in the homozygote comparison (AA vs. GG: P = 0.02, OR = 1.30, 95% CI 1.04-1.63, P heterogeneity = 0.39, fixed-effects model). No evidence suggested that rs1800975 polymorphism might associate with lung cancer in other ethnicities. Stratification analysis performed by histologic types indicated that AA genotype might represent a risk factor for squamous cell carcinoma [AA vs. (AG + GG): P = 0.01, OR = 1.42, 95% CI 1.08-1.86, P heterogeneity = 0.27, fixed-effects model; AA vs. GG: P = 0.03, OR = 1.43, 95% CI 1.04-1.96, P heterogeneity = 0.21, fixed-effects model]. No association was observed in adenocarcinoma subgroup. Our study suggested that XPA rs1800975 polymorphism might associate with lung cancer risk in overall and in East Asians. This polymorphism might also associate with squamous cell carcinoma.
Collapse
Affiliation(s)
- Yuqing Lou
- Department of Pulmonary, Shanghai Chest Hospital, Shanghai Jiaotong University, 241 West Huaihai Road, Shanghai, 200030, People's Republic of China
| | | | | | | | | | | | | | | |
Collapse
|
19
|
O'Carroll AM, Lolait SJ, Harris LE, Pope GR. The apelin receptor APJ: journey from an orphan to a multifaceted regulator of homeostasis. J Endocrinol 2013; 219:R13-35. [PMID: 23943882 DOI: 10.1530/joe-13-0227] [Citation(s) in RCA: 246] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The apelin receptor (APJ; gene symbol APLNR) is a member of the G protein-coupled receptor gene family. Neural gene expression patterns of APJ, and its cognate ligand apelin, in the brain implicate the apelinergic system in the regulation of a number of physiological processes. APJ and apelin are highly expressed in the hypothalamo-neurohypophysial system, which regulates fluid homeostasis, in the hypothalamic-pituitary-adrenal axis, which controls the neuroendocrine response to stress, and in the forebrain and lower brainstem regions, which are involved in cardiovascular function. Recently, apelin, synthesised and secreted by adipocytes, has been described as a beneficial adipokine related to obesity, and there is growing awareness of a potential role for apelin and APJ in glucose and energy metabolism. In this review we provide a comprehensive overview of the structure, expression pattern and regulation of apelin and its receptor, as well as the main second messengers and signalling proteins activated by apelin. We also highlight the physiological and pathological roles that support this system as a novel therapeutic target for pharmacological intervention in treating conditions related to altered water balance, stress-induced disorders such as anxiety and depression, and cardiovascular and metabolic disorders.
Collapse
Affiliation(s)
- Anne-Marie O'Carroll
- Henry Wellcome Laboratories for Integrative Neuroscience and Endocrinology, School of Clinical Sciences, University of Bristol, Dorothy Hodgkin Building, Whitson Street, Bristol BS1 3NY, UK
| | | | | | | |
Collapse
|
20
|
Rao YS, Wang ZF, Chai XW, Nie QH, Zhang XQ. Relationship between 5′ UTR length and gene expression pattern in chicken. Genetica 2013; 141:311-8. [DOI: 10.1007/s10709-013-9730-9] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2013] [Accepted: 08/11/2013] [Indexed: 11/29/2022]
|
21
|
Hu MC, Di Sole F, Zhang J, McLeroy P, Moe OW. Chronic regulation of the renal Na(+)/H(+) exchanger NHE3 by dopamine: translational and posttranslational mechanisms. Am J Physiol Renal Physiol 2013; 304:F1169-80. [PMID: 23427139 DOI: 10.1152/ajprenal.00630.2012] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
The intrarenal autocrine/paracrine dopamine (DA) system contributes to natriuresis in response to both acute and chronic Na(+) loads. While the acute DA effect is well described, how DA induces natriuresis chronically is not known. We used an animal and a cell culture model to study the chronic effect of DA on a principal renal Na(+) transporter, Na(+)/H(+) exchanger-3 (NHE3). Intraperitoneal injection of Gludopa in rats for 2 days elevated DA excretion and decreased total renal cortical and apical brush-border NHE3 antigen. Chronic treatment of an opossum renal proximal cell line with DA decreased NHE3 activity, cell surface and total cellular NHE3 antigen, but not NHE3 transcript. The decrease in NHE3 antigen was dose and time dependent with maximal inhibition at 16-24 h and half maximal effect at 3 × 10(-7) M. This is in contradistinction to the acute effect of DA on NHE3 (half maximal at 2 × 10(-6) M), which was not associated with changes in total cellular NHE3 protein. The DA-induced decrease in total NHE3 protein was associated with decrease in NHE3 translation and mediated by cis-sequences in the NHE3 5'-untranslated region. DA also decreased cell surface and total cellular NHE3 protein half-life. The DA-induced decrease in total cellular NHE3 was partially blocked by proteasome inhibition but not by lysosome inhibition, and DA increased ubiquitylation of total and surface NHE3. In summary, chronic DA inhibits NHE3 with mechanisms distinct from its acute action and involves decreased NHE3 translation and increased NHE3 degradation, which are novel mechanisms for NHE3 regulation.
Collapse
Affiliation(s)
- Ming Chang Hu
- Dept. of Internal Medicine, Univ. of Texas Southwestern Medical Center, 5323 Harry Hines Blvd., Dallas, TX 75390-8885, USA
| | | | | | | | | |
Collapse
|
22
|
Pérez-Fernández J, Megías M, Pombal MA. Distribution of a Y1 receptor mRNA in the brain of two Lamprey species, the sea lamprey (Petromyzon marinus) and the river Lamprey (Lampetra fluviatilis). J Comp Neurol 2013; 521:426-47. [PMID: 22740099 DOI: 10.1002/cne.23180] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2012] [Revised: 05/23/2012] [Accepted: 06/22/2012] [Indexed: 11/09/2022]
Abstract
The neuropeptide Y system consists of several neuropeptides acting through a broad number of receptor subtypes, the NPY family of receptors. NPY receptors are divided into three subfamilies (Y1, Y2, and Y5) that display a complex evolutionary history due to local and large-scale gene duplication events and gene losses. Lampreys emerged from a basal branch of the tree of vertebrates and they are in a key position to shed light on the evolutionary history of the NPY system. One member of the Y1 subfamily has been reported in agnathans, but the phylogenetic tree of the Y1 subfamily is not yet clear. We cloned the sequences of the Y1-subtype receptor of Petromyzon marinus and Lampetra fluviatilis to study the expression pattern of this receptor in lampreys by in situ hybridization and to analyze the phylogeny of the Y1-subfamily receptors in vertebrates. The phylogenetic study showed that the Y1 receptor of lampreys is basal to the Y1/6 branch of the Y1-subfamily receptors. In situ hybridization showed that the Y1 receptor is widely expressed throughout the brain of lampreys, with some regions showing numerous positive neurons, as well as the presence of numerous cerebrospinal fluid-contacting cells in the spinal cord. This broad distribution of the lamprey Y1 receptor is more similar to that found in other vertebrates for the Y1 receptor than that of the other members of the Y1 subfamily: Y4, Y8, and Y6 receptors. Both phylogenetic relationship and expression pattern suggest that this receptor is a Y1 receptor.
Collapse
Affiliation(s)
- Juan Pérez-Fernández
- Neurolam Group, Department of Functional Biology and Health Sciences, University of Vigo, Vigo, Spain
| | | | | |
Collapse
|
23
|
Evidence for an alternative genomic structure, mRNA and protein sequence of human ABCA13. Gene 2013; 515:298-307. [DOI: 10.1016/j.gene.2012.11.072] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2011] [Revised: 10/21/2012] [Accepted: 11/29/2012] [Indexed: 11/21/2022]
|
24
|
Human calmodulin methyltransferase: expression, activity on calmodulin, and Hsp90 dependence. PLoS One 2012; 7:e52425. [PMID: 23285036 PMCID: PMC3527508 DOI: 10.1371/journal.pone.0052425] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2012] [Accepted: 11/15/2012] [Indexed: 12/12/2022] Open
Abstract
Deletion of the first exon of calmodulin-lysine N-methyltransferase (CaM KMT, previously C2orf34) has been reported in two multigene deletion syndromes, but additional studies on the gene have not been reported. Here we show that in the cells from 2p21 deletion patients the loss of CaM KMT expression results in accumulation of hypomethylated calmodulin compared to normal controls, suggesting that CaM KMT is essential for calmodulin methylation and there are no compensatory mechanisms for CaM methylation in humans. We have further studied the expression of this gene at the transcript and protein levels. We have identified 2 additional transcripts in cells of the 2p21 deletion syndrome patients that start from alternative exons positioned outside the deletion region. One of them starts in the 2nd known exon, the other in a novel exon. The transcript starting from the novel exon was also identified in a variety of tissues from normal individuals. These new transcripts are not expected to produce proteins. Immunofluorescent localization of tagged CaM KMT in HeLa cells indicates that it is present in both the cytoplasm and nucleus of cells whereas the short isoform is localized to the Golgi apparatus. Using Western blot analysis we show that the CaM KMT protein is broadly expressed in mouse tissues. Finally we demonstrate that the CaM KMT interacts with the middle portion of the Hsp90 molecular chaperon and is probably a client protein since it is degraded upon treatment of cells with the Hsp90 inhibitor geldanamycin. These findings suggest that the CaM KMT is the major, possibly the single, methyltransferase of calmodulin in human cells with a wide tissue distribution and is a novel Hsp90 client protein. Thus our data provides basic information for a gene potentially contributing to the patient phenotype of two contiguous gene deletion syndromes.
Collapse
|
25
|
Fyfe JC, Lynch M, Olsen J, Louёr E. A thyroid peroxidase (TPO) mutation in dogs reveals a canid-specific gene structure. Mamm Genome 2012; 24:127-33. [PMID: 23223904 DOI: 10.1007/s00335-012-9442-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2012] [Accepted: 11/17/2012] [Indexed: 11/30/2022]
Abstract
Congenital hypothyroidism with goiter (CHG) occurring as an autosomal recessive disorder is typically due to a defect of thyroid hormone synthesis (aka dyshormonogenesis). Thyroid peroxidase (TPO) is a multifunctional, heme-containing enzyme whose activity is required, and several inactivating TPO mutations causing CHG in humans and dogs have been described. Recently, two half-sib Spanish water dog (SWD) pups were diagnosed with CHG based on clinical signs, endocrine testing, and thyroid histology. TPO enzyme activity was absent, and immuno-cross-reactive TPO was undetectable in affected-dog thyroid tissue. A single guanosine insertion was observed in the first exon of the affected-dog TPO cDNA at a site not previously thought to be within the coding sequence. The insertion allele segregated with the deduced disease allele in the SWD breed and was not observed in unrelated dogs of various breeds. Comparison of the insertion site (an 8-nt poly-G tract) with the orthologous sequences of other mammalian reference genomes revealed that the octa-G tract obliterated the intron 1 splice acceptor site and the exon 2 translation initiation codon found at that position in other species. An in-frame ATG in strong Kozak consensus context was observed in the normal dog sequence 12 codons 5' of the usual mammalian start site, suggesting that dogs have lost the noncoding exon 1 demonstrated in human and mouse. A survey of TPO sequences in other carnivore species indicates that the poly-G tract necessitating an alternative translation initiation site is a canid-specific feature.
Collapse
Affiliation(s)
- John C Fyfe
- Laboratory of Comparative Medical Genetics, Biomedical & Physical Sciences, Michigan State University, East Lansing, MI 48824, USA.
| | | | | | | |
Collapse
|
26
|
DUXO, a novel double homeobox transcription factor, is a regulator of the gastrula organizer in human embryonic stem cells. Stem Cell Res 2012; 9:261-9. [PMID: 23010573 DOI: 10.1016/j.scr.2012.08.003] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/17/2012] [Revised: 08/03/2012] [Accepted: 08/09/2012] [Indexed: 01/04/2023] Open
Abstract
Human embryonic stem cells differentiate into gastrula organizer cells that express typical markers and induce secondary axes when injected into frog embryos. Here, we report that these human organizer cells express DUXO (DUX of the Organizer), a novel member of the double-homeobox (DUX) family of transcription factors, a group of genes unique to placental mammals. Both of DUXO's homeodomains share high similarity with those of Siamois and Twin, the initial inducers of the amphibian gastrula organizer. DUXO overexpression in human embryoid bodies induces organizer related genes, whereas its knock down hampers formation of the organizer and its derivatives. Finally, we show that DUXO regulates GOOSECOID, the canonical organizer marker, in a direct manner, suggesting that DUXO is a major regulator of human organizer formation.
Collapse
|
27
|
Wang W, Luo J, Zhong Y, Lin XZ, Shi HB, Zhu JJ, Li J, Sun YT, Zhao WS. Goat liver X receptor α, molecular cloning, functional characterization and regulating fatty acid synthesis in epithelial cells of goat mammary glands. Gene 2012; 505:114-20. [DOI: 10.1016/j.gene.2012.05.028] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2011] [Revised: 05/03/2012] [Accepted: 05/10/2012] [Indexed: 10/28/2022]
|
28
|
Butkiewicz D, Drosik A, Suwiński R, Krześniak M, Rusin M, Kosarewicz A, Rachtan J, Matuszczyk I, Gawkowska-Suwińska M. Influence of DNA repair gene polymorphisms on prognosis in inoperable non-small cell lung cancer patients treated with radiotherapy and platinum-based chemotherapy. Int J Cancer 2012; 131:E1100-8. [PMID: 22511383 DOI: 10.1002/ijc.27596] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2012] [Accepted: 04/05/2012] [Indexed: 12/18/2022]
Abstract
Polymorphisms in DNA repair genes may modulate not only an individual DNA repair capacity, DNA damage levels and cancer risk but also clinical outcome after DNA damage-inducing anticancer therapy. In this study, we analyzed the association between the XPA -4G>A, XPD Asp312Asn, hOGG1 Ser326Cys, XRCC1 Arg399Gln, XRCC2 -4234G>C, XRCC3 -4541A>G and Thr241Met polymorphisms and prognosis in 250 inoperable non-small cell lung cancer (NSCLC) patients treated with radiotherapy and platinum-based chemotherapy. In univariate model, the XPA-4A and XRCC1 399Gln alleles alone and in combination influenced survival only in stage III group. In multivariate analysis, the XPA-4 GA/AA was associated with poor survival (HR 1.55, p = 0.011 overall and HR 1.72, p = 0.008 in stage III). In chemoradiotherapy group, the XPA-4A carriers were at increased risk of death and progression (HR 1.73, p = 0.013 and HR 1.65, p = 0.016, respectively), especially in stage III (p = 0.008). Moreover, individuals with ≥ 2 XPA/XRCC1 adverse alleles showed a higher risk of death (HR 1.46, p = 0.036 overall; HR 1.85, p = 0.004 in stage III and HR 1.71, p = 0.022 in chemoradiotherapy group) and progression (HR 1.75, p = 0.011 overall and HR 1.93, p = 0.005 in stage III). The XPA-4 GA/AA genotype individually and together with the XRCC1 399Gln was an independent unfavorable prognostic factor in our study. Thus, our findings indicate a prognostic potential of the XPA-4G>A in unresected NSCLC treated with radiotherapy and chemoradiotherapy. The results require validation in an independent population.
Collapse
Affiliation(s)
- Dorota Butkiewicz
- Center for Translational Research and Molecular Biology of Cancer, M Skłodowska-Curie Memorial Cancer Center and Institute of Oncology, Gliwice Branch, 44-101 Gliwice, Poland.
| | | | | | | | | | | | | | | | | |
Collapse
|
29
|
The dopamine D2 receptor gene in lamprey, its expression in the striatum and cellular effects of D2 receptor activation. PLoS One 2012; 7:e35642. [PMID: 22563388 PMCID: PMC3338520 DOI: 10.1371/journal.pone.0035642] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2012] [Accepted: 03/19/2012] [Indexed: 11/19/2022] Open
Abstract
All basal ganglia subnuclei have recently been identified in lampreys, the phylogenetically oldest group of vertebrates. Furthermore, the interconnectivity of these nuclei is similar to mammals and tyrosine hydroxylase-positive (dopaminergic) fibers have been detected within the input layer, the striatum. Striatal processing is critically dependent on the interplay with the dopamine system, and we explore here whether D2 receptors are expressed in the lamprey striatum and their potential role. We have identified a cDNA encoding the dopamine D2 receptor from the lamprey brain and the deduced protein sequence showed close phylogenetic relationship with other vertebrate D2 receptors, and an almost 100% identity within the transmembrane domains containing the amino acids essential for dopamine binding. There was a strong and distinct expression of D2 receptor mRNA in a subpopulation of striatal neurons, and in the same region tyrosine hydroxylase-immunoreactive synaptic terminals were identified at the ultrastructural level. The synaptic incidence of tyrosine hydroxylase-immunoreactive boutons was highest in a region ventrolateral to the compact layer of striatal neurons, a region where most striatal dendrites arborise. Application of a D2 receptor agonist modulates striatal neurons by causing a reduced spike discharge and a diminished post-inhibitory rebound. We conclude that the D2 receptor gene had already evolved in the earliest group of vertebrates, cyclostomes, when they diverged from the main vertebrate line of evolution (560 mya), and that it is expressed in striatum where it exerts similar cellular effects to that in other vertebrates. These results together with our previous published data (Stephenson-Jones et al. 2011, 2012) further emphasize the high degree of conservation of the basal ganglia, also with regard to the indirect loop, and its role as a basic mechanism for action selection in all vertebrates.
Collapse
|
30
|
Zallocchi M, Meehan DT, Delimont D, Rutledge J, Gratton MA, Flannery J, Cosgrove D. Role for a novel Usher protein complex in hair cell synaptic maturation. PLoS One 2012; 7:e30573. [PMID: 22363448 PMCID: PMC3281840 DOI: 10.1371/journal.pone.0030573] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2011] [Accepted: 12/22/2011] [Indexed: 12/22/2022] Open
Abstract
The molecular mechanisms underlying hair cell synaptic maturation are not well understood. Cadherin-23 (CDH23), protocadherin-15 (PCDH15) and the very large G-protein coupled receptor 1 (VLGR1) have been implicated in the development of cochlear hair cell stereocilia, while clarin-1 has been suggested to also play a role in synaptogenesis. Mutations in CDH23, PCDH15, VLGR1 and clarin-1 cause Usher syndrome, characterized by congenital deafness, vestibular dysfunction and retinitis pigmentosa. Here we show developmental expression of these Usher proteins in afferent spiral ganglion neurons and hair cell synapses. We identify a novel synaptic Usher complex comprised of clarin-1 and specific isoforms of CDH23, PCDH15 and VLGR1. To establish the in vivo relevance of this complex, we performed morphological and quantitative analysis of the neuronal fibers and their synapses in the Clrn1−/− mouse, which was generated by incomplete deletion of the gene. These mice showed a delay in neuronal/synaptic maturation by both immunostaining and electron microscopy. Analysis of the ribbon synapses in Ames waltzerav3J mice also suggests a delay in hair cell synaptogenesis. Collectively, these results show that, in addition to the well documented role for Usher proteins in stereocilia development, Usher protein complexes comprised of specific protein isoforms likely function in synaptic maturation as well.
Collapse
Affiliation(s)
- Marisa Zallocchi
- Boys Town National Research Hospital, Omaha, Nebraska, United States of America
| | - Daniel T. Meehan
- Boys Town National Research Hospital, Omaha, Nebraska, United States of America
| | - Duane Delimont
- Boys Town National Research Hospital, Omaha, Nebraska, United States of America
| | - Joseph Rutledge
- Otolaryngology-Head, Neck Surgery, St Louis University, St Louis, Missouri, United States of America
| | - Michael Anne Gratton
- Otolaryngology-Head, Neck Surgery, St Louis University, St Louis, Missouri, United States of America
| | - John Flannery
- Helen Wills Neuroscience Institute, University of California, Berkeley, California, United States of America
| | - Dominic Cosgrove
- Boys Town National Research Hospital, Omaha, Nebraska, United States of America
- University of Nebraska Medical Center, Omaha, Nebraska, United States of America
- * E-mail:
| |
Collapse
|
31
|
Salgado MC, Metón I, Anemaet IG, González JD, Fernández F, Baanante IV. Hepatocyte nuclear factor 4α transactivates the mitochondrial alanine aminotransferase gene in the kidney of Sparus aurata. MARINE BIOTECHNOLOGY (NEW YORK, N.Y.) 2012; 14:46-62. [PMID: 21607544 DOI: 10.1007/s10126-011-9386-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/14/2010] [Accepted: 05/03/2011] [Indexed: 05/30/2023]
Abstract
Alanine aminotransferase (ALT) plays an important role in amino acid metabolism and gluconeogenesis. The preference of carnivorous fish for protein amino acids instead of carbohydrates as a source of energy lead us to study the transcriptional regulation of the mitochondrial ALT (mALT) gene and to characterize the enzyme kinetics and modulation of mALT expression in the kidney of gilthead sea bream (Sparus aurata) under different nutritional and hormonal conditions. 5'-Deletion analysis of mALT promoter in transiently transfected HEK293 cells, site-directed mutagenesis and electrophoretic mobility shift assays allowed us to identify HNF4α as a new factor involved in the transcriptional regulation of mALT expression. Quantitative RT-PCR assays showed that starvation and the administration of streptozotocin (STZ) decreased HNF4α levels in the kidney of S. aurata, leading to the downregulation of mALT transcription. Analysis of the tissue distribution showed that kidney, liver, and intestine were the tissues with higher mALT and HNF4α expression. Kinetic analysis indicates that mALT enzyme is more efficient in catalyzing the conversion of L: -alanine to pyruvate than the reverse reaction. From these results, we conclude that HNF4α transactivates the mALT promoter and that the low levels of mALT expression found in the kidney of starved and STZ-treated fish result from a decreased expression of HNF4α. Our findings suggest that the mALT isoenzyme plays a major role in oxidazing dietary amino acids, and points to ALT as a target for a biotechnological action to spare protein and optimize the use of dietary nutrients for fish culture.
Collapse
Affiliation(s)
- María C Salgado
- Departament de Bioquímica i Biologia Molecular, Facultat de Farmàcia, Universitat de Barcelona, Joan XXIII s/n, 08028 Barcelona, Spain
| | | | | | | | | | | |
Collapse
|
32
|
Barris W, Harrison BE, McWilliam S, Bunch RJ, Goddard ME, Barendse W. Next generation sequencing of African and Indicine cattle to identify single nucleotide polymorphisms. ANIMAL PRODUCTION SCIENCE 2012. [DOI: 10.1071/an11095] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
We sequenced the genomes of a Brahman, an Africander and a Tuli bull because tropically adapted breeds of cattle have so far not been well characterised at the level of DNA variation. In excess of 16 Gb of Illumina GA-II sequence was obtained for each animal in the form of 75-bp paired-end reads, generating more than 6× coverage of each genome, and between 86.7 and 88.8% of the bases of each genome sequence was covered by one or more sequence reads. A total of 6.35 million single nucleotide polymorphisms (SNP) were discovered in the three animals, adding 3.56 million new SNP to dbSNP. The Brahman animal had nearly twice as many SNP as either the Tuli or the Africander. Comparing genome sequence to genotypic array data, genotype accuracy from sequencing was more than 98% for homozygotes that had at least six high quality sequence reads and for heterozygotes that had at least two high quality reads containing the alternative allele. Intergenic and intronic SNP were found at higher densities closer to coding sequences, and there was a reduction in numbers of SNP within 5 bp of a splice site, features consistent with genetic selection. On average, slightly more SNP per Mb, and slightly higher average reads per SNP per Mb, were found towards the ends of chromosomes, especially towards the telomeric end of the chromosome. At least one autosome in each animal showed a large stretch of homozygosity, the largest was 58 Mb long in the Tuli, although the animals are not known to have recent inbreeding.
Collapse
|
33
|
Ding D, Zhang Y, Yu H, Guo Y, Jiang L, He X, Ma W, Zheng W. Genetic variation of XPA gene and risk of cancer: a systematic review and pooled analysis. Int J Cancer 2011; 131:488-96. [PMID: 21866550 DOI: 10.1002/ijc.26391] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2011] [Accepted: 08/15/2011] [Indexed: 12/17/2022]
Abstract
XPA, a zinc-finger DNA-binding protein, play an important role in both global genome and transcription-coupled repair pathways. XPA -4G>A polymorphism was identified in the 5' noncoding region, located four nucleotides upstream of the ATG start codon. Previous studies have shown that this polymorphism may affect mRNA tertiary structure and stability and play a role in susceptibility to cancer. However, the results remained controversial. To derive a more precise estimation of association between this polymorphism and risk of different types of cancer, we performed a meta-analysis based on 36 case-control or case-cohort studies, including a total of 11,700 cases and 15,033 controls. We used odds ratios with 95% confidence intervals to assess the strength of the association. Overall, no significantly elevated cancer risk was found in all genetic models when eligible studies were pooled into the meta-analysis. In the stratified analyses, we found that individuals with A-allele had a higher risk of lung cancer (AA versus GG: OR = 1.25, 95% CI = 1.09-1.43; recessive model: OR = 1.31, 95% CI = 1.16-1.48). When stratified by ethnicity, significantly elevated risks were observed among Asian populations (AA versus GG: OR = 1.31, 95% CI = 1.01-1.70; dominant model: OR = 1.14, 95% CI = 1.00-1.30). This meta-analysis suggests that XPA -4G>A polymorphism is associated with increased lung cancer risk and may be a low-penetrant risk factor in Asian ethnicity for cancer development.
Collapse
Affiliation(s)
- Dapeng Ding
- Institute of Genetic Engineering, Southern Medical University, Guangzhou, People's Republic of China
| | | | | | | | | | | | | | | |
Collapse
|
34
|
Wang JQ, Hou L, Yi N, Zhang RF, Zou XY. Molecular analysis and its expression of a pou homeobox protein gene during development and in response to salinity stress from brine shrimp, Artemia sinica. Comp Biochem Physiol A Mol Integr Physiol 2011; 161:36-43. [PMID: 21911072 DOI: 10.1016/j.cbpa.2011.08.016] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2011] [Revised: 08/29/2011] [Accepted: 08/29/2011] [Indexed: 10/17/2022]
Abstract
Brine shrimps of the genus Artemia are aquatic species of economic importance because of their important significance to aquaculture and are used as a model species in physiology and developmental biology. Research on Artemia POU homeobox gene function will enhance our understanding of the physiological and developmental processes of POU homeobox gene in animals. Herein, a full-length cDNA encoding an Artemia POU homeobox protein gene 1 (APH-1) from Artemia sinica (designated as As-APH-1) was cloned and characterized by a reverse-transcription polymerase chain reaction (RT-PCR) and rapid amplification of cDNA end (RACE) method. The As-APH-1 gene encoded a protein of 388 amino acid polypeptide with a calculated molecular mass of 42.85kDa and an isoelectric point of 6.90 and the protein belongs to the POU III family. Multiple sequence alignments revealed that A. sinica As-APH-1 protein sequence shared a conserved POU homeobox domain with other species. The early and persistent expression of As-APH-1 in the naupliar stages by semi-quantitative RT-PCR and whole-mount embryonic immunohistochemistry suggest that As-APH-1 functions very early in the salt gland and may be required continuously in this organ. Later in development, expression of As-APH-1 begins to dramatically decrease and disappear in salt gland of the sub-adult Artemia. In addition, we also discovered that As-APH-1 increased obviously as the salinity increased, indicating that As-APH-1 might be used as a good indicator of salinity stress. In summary, we are the first to identify the As-APH-1 gene and to determine its gene expression patterns in early embryogenesis stages and in different salinity stress in brine shrimp, A. sinica. The result of expression of As-APH-1 affected by salinity changes will provide us further understanding of the underlying mechanisms of osmoregulation in Artemia early embryonic development.
Collapse
Affiliation(s)
- Jia-Qing Wang
- College of Science and Technology, Shenyang Agricultural University, Fushun, PR China
| | | | | | | | | |
Collapse
|
35
|
O'Driscoll KE, Pipe RA, Britton FC. Increased complexity of Tmem16a/Anoctamin 1 transcript alternative splicing. BMC Mol Biol 2011; 12:35. [PMID: 21824394 PMCID: PMC3170211 DOI: 10.1186/1471-2199-12-35] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2011] [Accepted: 08/08/2011] [Indexed: 01/09/2023] Open
Abstract
Background TMEM16A (Anoctamin 1; ANO1) is an eight transmembrane protein that functions as a calcium-activated chloride channel. TMEM16A in human exhibits alternatively spliced exons (6b, 13 and 15), which confer important roles in the regulation of channel function. Mouse Tmem16a is reported to consist of 25 exons that code for a 956 amino acid protein. In this study our aim was to provide details of mouse Tmem16a genomic structure and to investigate if Tmem16a transcript undergoes alternative splicing to generate channel diversity. Results We identified Tmem16a transcript variants consisting of alternative exons 6b, 10, 13, 14, 15 and 18. Our findings indicate that many of these exons are expressed in various combinations and that these splicing events are mostly conserved between mouse and human. In addition, we confirmed the expression of these exon variants in other mouse tissues. Additional splicing events were identified including a novel conserved exon 13b, tandem splice sites of exon 1 and 21 and two intron retention events. Conclusion Our results suggest that Tmem16a gene is significantly more complex than previously described. The complexity is especially evident in the region spanning exons 6 through 16 where a number of the alternative splicing events are thought to affect calcium sensitivity, voltage dependence and the kinetics of activation and deactivation of this calcium-activated chloride channel. The identification of multiple Tmem16a splice variants suggests that alternative splicing is an exquisite mechanism that operates to diversify TMEM16A channel function in both physiological and pathophysiological conditions.
Collapse
Affiliation(s)
- Kate E O'Driscoll
- Department of Physiology and Cell Biology, 1664 North Virginia Street, University of Nevada School of Medicine, Reno, Nevada 89557-0046, USA
| | | | | |
Collapse
|
36
|
|
37
|
Advanced zebrafish transgenesis with Tol2 and application for Cre/lox recombination experiments. Methods Cell Biol 2011; 104:173-94. [PMID: 21924163 DOI: 10.1016/b978-0-12-374814-0.00010-0] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Spatio-temporal transgene regulation by transgenic DNA recombinases is a central tool for genetic research in multicellular organisms, with excellent applications for misexpression and lineage tracing experiments. Cre recombinase-controlled lox site recombination is a cornerstone of contemporary mouse genetics, and Cre/lox techniques therefore attract increasing interest in the zebrafish field. Tol2-mediated zebrafish transgenesis now provides a stable platform for lox cassette transgenes, while the ease of drug treatments in zebrafish makes the model an ideal candidate for Tamoxifen/4-hydroxytamoxifen-inducible CreER(T2) experiments. In this chapter, we will first introduce the basics of Cre/lox methodology, CreER(T2) regulation by Tamoxifen/4-hydroxytamoxifen, as well as the benefits of Tol2 transgenesis for Cre/lox experiments. We will then in detail outline practical experimental steps for Tol2 transgenesis toward the creation of single-insertion transgenes. Lastly, we will introduce protocols for 4-hydroxytamoxifen-mediated CreER(T2) induction to perform spatio-temporal lox transgene regulation experiments in zebrafish embryos.
Collapse
|
38
|
Wu SL, Tsai MS, Wong SH, Hsieh-Li HM, Tsai TS, Chang WT, Huang SL, Chiu CC, Wang SH. Characterization of genomic structures and expression profiles of three tandem repeats of a mouse double homeobox gene: Duxbl. Dev Dyn 2010; 239:927-40. [PMID: 20063414 DOI: 10.1002/dvdy.22210] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
We identified and cloned a mouse double homeobox gene (Duxbl), which encodes two homeodomains. Duxbl gene, a tandem triplicate produces two major transcripts, Duxbl and Duxbl-s. The amino acid sequences of Duxbl homeodomains are most similar to those of human DUX4 protein, associated with facioscapulohumeral muscular dystrophy. In adult tissues, Duxbl is predominantly expressed in female reproductive organs and eyes, and slightly expressed in brain and testes. During gonad development, Duxbl is expressed from embryonic to adult stages and specifically expressed in oocytes and spermatogonia. During embryonic development, Duxbl is transcribed in limbs and tail. However, Duxbl proteins were only detected in trunk and limb muscles and in elongated myocytes and myotubes. In C2C12 muscle cell line, Duxbl expression pattern is similar to differentiated marker gene, Myogenin, increased in expression from 2 days onward in differentiating medium. We suggest that Duxbl proteins play regulatory roles during myogenesis and reproductive developments.
Collapse
Affiliation(s)
- Shey-Lin Wu
- Department of Neurology, Chang-Hua Christian Hospital, and Department of Bioindustry Technology, Da-Yeh University, Dacun, Changhua, Taiwan
| | | | | | | | | | | | | | | | | |
Collapse
|
39
|
A functional analysis of G23A polymorphism and the alternative splicing in the expression of the XPA gene. Cell Mol Biol Lett 2010; 15:611-29. [PMID: 20865363 PMCID: PMC6275895 DOI: 10.2478/s11658-010-0032-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2010] [Accepted: 09/07/2010] [Indexed: 12/31/2022] Open
Abstract
The XPA gene has a commonly occurring polymorphism (G23A) associated with cancer risk. This study assessed the functional significance of this polymorphism, which is localised near the translation start codon. Lymphoblastoid cell lines with alternative homozygous genotypes showed no significant differences in their XPA levels. The luciferase reporter assay detected no functional difference between the two sequences. Unexpectedly, we found that the alternatively spliced form of XPA mRNA lacked a part of exon 1. Only the reading frame downstream of codon Met59 was preserved. The alternative mRNA is expressed in various human tissues. The analysis of the 5’cDNA ends showed similar transcription start sites for the two forms. The in vitro expression of the alternative XPA labelled with the red fluorescent protein (mRFP) showed a lack of preferential nuclear accumulation of the XPA isoform. The biological role of the alternative XPA mRNA form remains to be elucidated.
Collapse
|
40
|
Campanella GSV, Colvin RA, Luster AD. CXCL10 can inhibit endothelial cell proliferation independently of CXCR3. PLoS One 2010; 5:e12700. [PMID: 20856926 PMCID: PMC2938333 DOI: 10.1371/journal.pone.0012700] [Citation(s) in RCA: 68] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2010] [Accepted: 08/17/2010] [Indexed: 01/22/2023] Open
Abstract
CXCL10 (or Interferon-inducible protein of 10 kDa, IP-10) is an interferon-inducible chemokine with potent chemotactic activity on activated effector T cells and other leukocytes expressing its high affinity G protein-coupled receptor CXCR3. CXCL10 is also active on other cell types, including endothelial cells and fibroblasts. The mechanisms through which CXCL10 mediates its effects on non-leukocytes is not fully understood. In this study, we focus on the anti-proliferative effect of CXCL10 on endothelial cells, and demonstrate that CXCL10 can inhibit endothelial cell proliferation in vitro independently of CXCR3. Four main findings support this conclusion. First, primary mouse endothelial cells isolated from CXCR3-deficient mice were inhibited by CXCL10 as efficiently as wildtype endothelial cells. We also note that the proposed alternative splice form CXCR3-B, which is thought to mediate CXCL10's angiostatic activity, does not exist in mice based on published mouse CXCR3 genomic sequences as an in-frame stop codon would terminate the proposed CXCR3-B splice variant in mice. Second, we demonstrate that human umbilical vein endothelial cells and human lung microvascular endothelial cells that were inhibited by CXL10 did not express CXCR3 by FACS analysis. Third, two different neutralizing CXCR3 antibodies did not inhibit the anti-proliferative effect of CXCL10. Finally, fourth, utilizing a panel of CXCL10 mutants, we show that the ability to inhibit endothelial cell proliferation correlates with CXCL10's glycosaminoglycan binding affinity and not with its CXCR3 binding and signaling. Thus, using a very defined system, we show that CXCL10 can inhibit endothelial cell proliferation through a CXCR3-independent mechanism.
Collapse
Affiliation(s)
- Gabriele S. V. Campanella
- Division of Rheumatology, Allergy and Immunology, Center for Immunology and Inflammatory Diseases, Massachusetts General Hospital, Harvard Medical School, Charlestown, Massachusetts, United States of America
| | - Richard A. Colvin
- Division of Rheumatology, Allergy and Immunology, Center for Immunology and Inflammatory Diseases, Massachusetts General Hospital, Harvard Medical School, Charlestown, Massachusetts, United States of America
| | - Andrew D. Luster
- Division of Rheumatology, Allergy and Immunology, Center for Immunology and Inflammatory Diseases, Massachusetts General Hospital, Harvard Medical School, Charlestown, Massachusetts, United States of America
- * E-mail:
| |
Collapse
|
41
|
Schmoll M, Seibel C, Tisch D, Dorrer M, Kubicek CP. A novel class of peptide pheromone precursors in ascomycetous fungi. Mol Microbiol 2010; 77:1483-501. [PMID: 20735770 PMCID: PMC3068285 DOI: 10.1111/j.1365-2958.2010.07295.x] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/06/2010] [Indexed: 11/30/2022]
Abstract
Recently, sexual development in the heterothallic ascomycete Trichoderma reesei (anamorph of Hypocrea jecorina) has been achieved and thus initiated attempts to elucidate regulation and determinants of this process. While the α-type pheromone of this fungus fits the consensus known from other fungi, the assumed a-type peptide pheromone precursor shows remarkably unusual characteristics: it comprises three copies of the motif (LI)GC(TS)VM thus constituting a CAAX domain at the C-terminus and two Kex2-protease sites. This structure shares characteristics of both a- and α-type peptide pheromone precursors. Presence of hybrid-type peptide pheromone precursor 1 (hpp1) is essential for male fertility, thus indicating its functionality as a peptide pheromone precursor, while its phosphorylation site is not relevant for this process. However, sexual development in a female fertile background is not perturbed in the absence of hpp1, which rules out a higher order function in this process. Open reading frames encoding proteins with similar characteristics to HPP1 were also found in Fusarium spp., of which Fusarium solani still retains a putative a-factor-like protein, but so far in no other fungal genome available. We therefore propose the novel class of h-type (hybrid) peptide pheromone precursors with H. jecorina HPP1 as the first member of this class.
Collapse
Affiliation(s)
- Monika Schmoll
- Research Area Gene Technology and Applied Biochemistry, Institute of Chemical Engineering, Vienna University of Technology, Getreidemarkt 9/1665, Vienna 1060, Austria.
| | | | | | | | | |
Collapse
|
42
|
Hendrickson RC, Wang C, Hatcher EL, Lefkowitz EJ. Orthopoxvirus genome evolution: the role of gene loss. Viruses 2010; 2:1933-1967. [PMID: 21994715 PMCID: PMC3185746 DOI: 10.3390/v2091933] [Citation(s) in RCA: 146] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2010] [Revised: 08/25/2010] [Accepted: 09/01/2010] [Indexed: 12/26/2022] Open
Abstract
Poxviruses are highly successful pathogens, known to infect a variety of hosts. The family Poxviridae includes Variola virus, the causative agent of smallpox, which has been eradicated as a public health threat but could potentially reemerge as a bioterrorist threat. The risk scenario includes other animal poxviruses and genetically engineered manipulations of poxviruses. Studies of orthologous gene sets have established the evolutionary relationships of members within the Poxviridae family. It is not clear, however, how variations between family members arose in the past, an important issue in understanding how these viruses may vary and possibly produce future threats. Using a newly developed poxvirus-specific tool, we predicted accurate gene sets for viruses with completely sequenced genomes in the genus Orthopoxvirus. Employing sensitive sequence comparison techniques together with comparison of syntenic gene maps, we established the relationships between all viral gene sets. These techniques allowed us to unambiguously identify the gene loss/gain events that have occurred over the course of orthopoxvirus evolution. It is clear that for all existing Orthopoxvirus species, no individual species has acquired protein-coding genes unique to that species. All existing species contain genes that are all present in members of the species Cowpox virus and that cowpox virus strains contain every gene present in any other orthopoxvirus strain. These results support a theory of reductive evolution in which the reduction in size of the core gene set of a putative ancestral virus played a critical role in speciation and confining any newly emerging virus species to a particular environmental (host or tissue) niche.
Collapse
Affiliation(s)
- Robert Curtis Hendrickson
- Department of Microbiology, University of Alabama at Birmingham, BBRB 276/11, 845 19th St S, Birmingham, AL 35222, USA; E-Mails: (R.C.H.); (E.L.H.)
| | - Chunlin Wang
- Stanford Genome Technology Center, Stanford University, 855 California Ave, Palo Alto, CA 94304, USA; E-Mail:
| | - Eneida L. Hatcher
- Department of Microbiology, University of Alabama at Birmingham, BBRB 276/11, 845 19th St S, Birmingham, AL 35222, USA; E-Mails: (R.C.H.); (E.L.H.)
| | - Elliot J. Lefkowitz
- Department of Microbiology, University of Alabama at Birmingham, BBRB 276/11, 845 19th St S, Birmingham, AL 35222, USA; E-Mails: (R.C.H.); (E.L.H.)
| |
Collapse
|
43
|
Cloning and characterization of p8 homolog cDNA in the Atlantic halibut (Hippoglossus hippoglossus). Biochem Genet 2010; 48:504-15. [PMID: 20454949 DOI: 10.1007/s10528-010-9334-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2009] [Accepted: 01/25/2010] [Indexed: 10/19/2022]
Abstract
The p8 gene encodes a transcription factor known to modulate cell growth, division, and apoptosis and influences gene expression. In this study, an Atlantic halibut (Hippoglossus hippoglossus) homolog of the p8 gene was cloned, sequenced, and characterized. The full-length p8 cDNA consists of 601 bp and encodes 76 amino acids with a molecular mass of 9 kD. The bHLH region is well conserved between Atlantic halibut and other animals. Analysis by RT-PCR showed that the p8 transcript is constitutively expressed in 9 of the 12 tissues tested: pancreas, intestine, stomach, gill, head kidney, heart, liver, ovary, and spleen. A predicted microRNA target site was found in the 3'UTR of Atlantic halibut p8 mRNA. We speculate that the target site may pair to microRNA molecules because the target site resides in a big loop, a space large enough for the binding of microRNA molecules.
Collapse
|
44
|
Diekmann S, Henneke M, Burckhardt BC, Gärtner J. Pelizaeus-Merzbacher-like disease is caused not only by a loss of connexin47 function but also by a hemichannel dysfunction. Eur J Hum Genet 2010; 18:985-92. [PMID: 20442743 DOI: 10.1038/ejhg.2010.61] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
Autosomal recessive mutations in the GJA12/GJC2 gene encoding the gap junction protein connexin47 (C x 47) cause a form of Pelizaeus-Merzbacher-like disease (PMLD) with hypomyelination, nystagmus, impaired psychomotor development and progressive spasticity. We investigated the functional consequences of four C x 47 missense mutations (G149S, G236R, T265A, and T398I) and one C x 47 complex mutation (A98G_V99insT) by immunoblot analysis and immunocytochemistry in transfected communication-incompetent HeLa cells and in OLI-neu cells. All studied C x 47 mutants, except G236R, generated stable proteins in transfected HeLa cells and OLI-neu cells. The mutants T265A and A98G_V99insT were retained in the ER, T398I formed gap junctional plaques at the plasma membrane, and G149S showed both, structures at the plasma membrane and ER localization. Two-microelectrode voltage clamp analyses in Xenopus laevis oocytes injected with wild-type and mutant C x 47 cRNA revealed reduced hemichannel currents for G236R, T265A, and A98G_V99insT. In contrast, T398I revealed hemichannel currents comparable to wild-type. For C x 47 mutant T398I, our results indicate a defect in hemichannel function, whereas C x 47 mutants G149S, G236R, T265A, and A98G_V99insT are predicted to result in a loss of C x 47 hemichannel function. Thus, PMLD is likely to be caused by two different disease mechanisms: a loss of function and a dysfunction.
Collapse
Affiliation(s)
- Simone Diekmann
- Department of Pediatrics and Pediatric Neurology, Georg August University, Göttingen, Germany
| | | | | | | |
Collapse
|
45
|
An overview of the current status of eukaryote gene prediction strategies. Gene 2010; 461:1-4. [PMID: 20430068 DOI: 10.1016/j.gene.2010.04.008] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2010] [Revised: 04/15/2010] [Accepted: 04/16/2010] [Indexed: 01/12/2023]
Abstract
As sequence data continues to be generated at a logarithmic rate our dependence on effective in silico gene prediction methods is also increasing. Herein, I review the current state of eukaryote gene prediction methods; their strengths, weaknesses and future directions.
Collapse
|
46
|
Genomic organization and regulation of the human orexin (hypocretin) receptor 2 gene: identification of alternative promoters. Biochem J 2010; 427:377-90. [DOI: 10.1042/bj20091755] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Orexins (hypocretins), acting via their receptors, are involved in the control of feeding behaviour, sleep, arousal and energy homoeostasis. However, regulation of the human orexin receptor 2 (hOX2R) gene remains unknown. We have identified four transcripts arising from alternative splicing from three exons. These exon 1 variants were designated exons 1A, 1B and 1C on the basis of their 5′–3′ order. RT (reverse transcription)–PCR demonstrates the differential expression in various human tissues. The alternative 5′-UTRs (untranslated regions) possessed by these isoforms have different translational efficiencies, which regulate the level of protein expression. In the present study, we have demonstrated that the hOX2R gene is regulated by two promoters and the novel transcripts are regulated by the distal promoter located upstream of exon 1A. We have demonstrated that the AP-1 (activator protein 1) motif is critical for sustaining the basal activity of distal promoter. Analysis of the proximal promoter revealed the region regulating promoter activity contained putative binding elements including those for CREB (cAMP-response-element-binding protein), GATA-2 and Oct-1. Using the chromatin immunoprecipitation assay, we demonstrated that CREB, GATA-2 and Oct-1 transcription factors bind to these critical regulatory promoter elements. Mutational studies suggested that these motifs functioned independently, but have a compound effect regulating hOX2R gene transcription. Furthermore, proximal promoter activity is enhanced by both PKA (protein kinase A) and PKC (protein kinase C) pathway activation, via binding of CREB and GATA-2 transcription factors. In conclusion, we have demonstrated that expression of hOX2R is regulated by a complex involving a proximal PKA/PKC-regulated promoter and a distal promoter regulating tissue-specific expression of alternative transcripts which in turn post-transcriptionally regulate receptor levels.
Collapse
|
47
|
Huang WB, Ren HL, Gopalakrishnan S, Xu DD, Qiao K, Wang KJ. First molecular cloning of a molluscan caspase from variously colored abalone (Haliotis diversicolor) and gene expression analysis with bacterial challenge. FISH & SHELLFISH IMMUNOLOGY 2010; 28:587-595. [PMID: 20045058 DOI: 10.1016/j.fsi.2009.12.016] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/06/2009] [Revised: 12/13/2009] [Accepted: 12/15/2009] [Indexed: 05/28/2023]
Abstract
Mammal caspases have been demonstrated to possess important functions in apoptosis and immune signaling, but there is less knowledge available on abalone caspases. In the present study, a molluscan caspase gene, abCaspase, was cloned for the first time from the variously colored abalone (Haliotis diversicolor) and its full-length cDNA sequence was 2427 bp, with a 1008 bp of open reading frame encoding a protein of 336 aa. The molecular mass of the deduced protein was approximately 36.97 kDa with an estimated pI of 5.28. The predicted amino acid sequence of abCaspase contained two domains of p20 and p10 which were conserved in the caspase family, including the cysteine active site pentapeptide "QSCRG" and the histidine active site signature "HTVYDCVVVIFLTHG". Homology analysis showed that abCaspase shared high similarity with apoptotic caspases and it was grouped together with vertebrate caspase-8s and caspase-10s using phylogenetic analysis, suggesting that abCaspase belonged to a typical apoptotic caspase and might possess the characteristic of human caspase-8 and -10. The mRNA transcripts of abCaspase were widely distributed in various tissues of H. diversicolor. Expression of the abCaspase gene was significantly induced in the tissues tested, especially in the hemocytes, gill and mantle with bacterial challenge. This study suggested that abCaspase may be an initiator caspase associated with the induction of apoptosis which is potentially involved in the immune defense of H. diversicolor.
Collapse
Affiliation(s)
- Wei-Bin Huang
- State Key Laboratory of Marine Environmental Science, College of Oceanography and Environmental Science, Xiamen University, Xiamen 361005, China
| | | | | | | | | | | |
Collapse
|
48
|
Wong JWY, Chan CL, Tang WK, Cheng CHK, Fong WP. Is antiquitin a mitochondrial Enzyme? J Cell Biochem 2010; 109:74-81. [PMID: 19885858 DOI: 10.1002/jcb.22381] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Antiquitin is an aldehyde dehydrogenase involved in the catabolism of lysine. Mutations of antiquitin have been linked with the disease pyridoxine-dependent seizures. While it is well established that lysine metabolism takes place in the mitochondrial matrix, evidence for the mitochondrial localization of antiquitin has been lacking. In the present study, the subcellular localization of antiquitin was investigated using human embryonic kidney HEK293 cells. Three different approaches were used. First, confocal microscopic analysis was carried out on cells transiently transfected with fusion constructs containing enhanced green fluorescent protein with different lengths of antiquitin based on the different potential start codons of translation. Second, immunofluorescence staining was used to detect the localization of antiquitin directly in the cells. Third, subcellular fractionation was carried out and the individual fraction was analyzed for the presence of antiquitin by Western blot and flow cytometric analyses. All the results showed that antiquitin was present not only in the cytosol but also in the mitochondria.
Collapse
Affiliation(s)
- Judy Wei-Yan Wong
- Department of Biochemistry, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong, China
| | | | | | | | | |
Collapse
|
49
|
|
50
|
Galloway JN, Nelson DL. Evidence for RNA-mediated toxicity in the fragile X-associated tremor/ataxia syndrome. FUTURE NEUROLOGY 2009; 4:785. [PMID: 20161676 DOI: 10.2217/fnl.09.44] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Fragile X premutation carriers are at risk for developing a late-onset, progressive neurodegenerative disorder termed fragile X-associated tremor/ataxia syndrome (FXTAS). A growing body of evidence suggests the characteristic excess CGG repeat containing FMR1 mRNA observed in premutation carriers is pathogenic and leads to clinical features of FXTAS. The current model suggests premutation mRNA transcripts can induce the formation of intranuclear inclusions by the sequestration of RNA-binding proteins and other proteins. The sequestered proteins are prevented from performing their normal functions, which is thought to lead to the neuropathology-observed FXTAS. This paper discusses the existing evidence that microsatellite expansions at the level of RNA play a role in the disease pathogenesis of FXTAS and some of the approaches that may uncover downstream effects of expanded riboCGG expression.
Collapse
Affiliation(s)
- Jocelyn N Galloway
- Baylor College of Medicine, Interdepartmental Program in Cell & Molecular Biology, One Baylor Plaza, Room 904E, Houston, TX 77030, USA, Tel.: +1 713 798 7898, Fax.: +1 713 798 1116
| | | |
Collapse
|