Digeorge Syndrome: an Enigma in Mice and Men

Total Page:16

File Type:pdf, Size:1020Kb

Digeorge Syndrome: an Enigma in Mice and Men MOLECULAR MEDICINE TODAY, JANUARY 2000 (VOL. 6) Letters DiGeorge syndrome: an enigma in mice and men One out of 4000 children are born with cardiac and syndrome, including the pharyngeal arches, car- to the phenotype. The precise role of Ufd1l and facial abnormalities and are missing part of chro- diac outflow tract and aortic arch arteries (specif- Cdc45l in vertebrates remains unknown as well; mosome 22 (22q11.2). This condition, often called ically, the fourth aortic arch artery). A small 20 kb however, in yeast they are involved in a ubiquitin- DiGeorge syndrome (DGS), is the most common deletion in a ‘non-deleted’ DiGeorge patient (JF) dependent degradation pathway and cell cycle human deletion syndrome and the second most encompassing portions of UFD1L and a neigh- regulation, respectively. Numerous studies sug- common genetic cause of congenital heart de- boring gene, CDC45L, provided further evidence gest convergence of degradation and cell cycle fects1. Thymic and parathyroid anomalies are that UFD1L and/or CDC45L might be involved in pathways, raising the possibility that Ufd1l, Cdc45l often present, together with characteristic heart the etiology of 22q11 deletion syndrome6. As and perhaps HIRA function in a common pathway and facial defects. Because of the diverse clinical Novelli points out, this is not evidence that loss of during development. Whether heterozygosity of features, it is possible that more than one gene on one copy of UFD1L alone is sufficient to cause the the three genes in combination disrupts neural 22q11.2 contributes to the observed phenotype. phenotype; however, the results would suggest crest development will soon be testable in gen- However, many of the embryonic regions affected that a molecular pathway involving UFD1L con- etically engineered mice. in 22q11 deletion are derived from a common mi- tributes to the 22q11 phenotype. As the molecular basis for the 22q11 deletion gratory population of cells that arise from the The recently published contribution by Baldini’s syndrome becomes clearer, how will our findings neural folds (neural crest cells), suggesting that group7 has further clarified the potential etiologies impact individuals and families who have endured disruption of a molecular pathway regulating of 22q11 deletion syndrome. By generating a the hardships associated with chronic and often neural crest development might account for many mouse mutant lacking 14 of the genes in the pre- life-threatening disease? Unfortunately, fetal aspects of 22q11 deletion syndrome. sumed ‘DiGeorge critical region’ (DGCR), they gene therapy approaches remain in the distant fu- Substantial resources have been devoted to were able to recapitulate defects of the cardiac out- ture. However, the wide variance of clinical fea- identifying the genes involved in this syndrome. flow tract and fourth aortic arch artery observed in tures in individuals with 22q11 deletion might in- Genetic analyses of hundreds of patients suggest the syndrome. Unfortunately, no other phenotypic dicate that secondary genetic or environmental that the size of 22q11 deletion, typically 3 million features were found, and even the arch defects factors influence the final phenotype. Thus, regu- base pairs, does not correlate with the type of de- were incompletely penetrant (25%). It remains to lation of secondary factors during fetal develop- fects observed. As a result, the human genetic be determined whether genes outside this region ment in individuals genetically predisposed to ab- focus has shifted to studying patients with charac- are important for other features of the syndrome, normal neural crest development might allow teristic phenotypic features but without a de- or if species-specific factors influence the discor- attenuation of the disease. It is our hope that con- tectable 22q11 deletion. Surprisingly, extensive dant phenotypes observed in mice and men. Their tinued efforts to understand molecular pathways mutation analysis of such ‘non-deleted’ patients deletion (Df1) encompassed Ufd1l and Cdc45l but in mouse and chick neural crest development will has failed to reveal even a single individual, world- also removed 12 other genes. Interestingly, het- ultimately lead to novel approaches for preven- wide, who harbors a point mutation in any of the erozygosity of Ufd1l alone was not sufficient to tion of cardiac and craniofacial birth defects. Until genes (which number close to 30) in the commonly cause aortic arch defects in mice, although loss of then, scientists, clinicians and patients will con- deleted region. This finding might suggest that the both alleles of Ufd1l resulted in embryonic lethal- tinue to experience the roller coaster of hope and vast majority of non-deleted patients have genetic ity. As Baldini suggests, further studies will be re- despair that have become a hallmark of the 22q11 or epigenetic causes of neural crest disruption that quired to clarify the role of UFD1L in the 22q11.2 deletion syndrome. are unrelated to 22q11. Adding further complexity deletion phenotype. Whether heterozygosity of to the human genetic approach are the ten re- Ufd1l and Cdc45, as suggested by patient JF, is References ported patients that harbor large, atypical deletions sufficient to cause aortic arch defects in mice is 1 Emanuel, B.S. et al. (1998) The genetic basis of of 22q11 that do not overlap with one another2. currently being tested in our laboratory. conotruncal cardiac defects: The chromosome Two recent reports have begun to take alterna- Ultimately, the most important information will 22q11.2 deletion, in Heart Development (R.P. tive approaches to identifying the genes involved come from understanding the function of the vari- Harvey and N. Rosenthal, eds) pp. 463–478, in this enigmatic syndrome. Our group had been ous genes in the commonly deleted region. HIRA, Academic Press studying the function of a transcription factor, a gene located 50 kb away from UFD1L, encodes 2 Novelli, G. et al. (2000) Individual haploinsufficient dHAND, required for survival of the types of neural a protein that interacts with the homeodomain pro- loci and the complex phenotype of the DiGeorge crest-derived cells affected in 22q11 deletion syn- tein Pax3, and might play a functional role in syndrome. Mol. Med. Today 6, 10–11 drome3–5. In a screen for genes downregulated in neural crest development in chick embryos8,9. The 3 Srivastava, D. et al. (1995) A new subclass of bHLH dHAND mutant mice, we identified UFD1L, which yeast homolog of HIRA (Tup1) plays a role in cell proteins required for cardiac morphogenesis. had previously been mapped to 22q11 by Novelli’s cycle-dependent histone regulation. Although nei- Science 270, 1995–1999 group. We subsequently demonstrated specific ther Baldini’s nor our studies have addressed 4 Srivastava, D. et al. (1997) Regulation of cardiac UFD1L mRNA expression in the neural-crest-de- HIRA’s role in 22q11 deletion, it remains conceiv- mesoderm and neural crest by the bHLH protein, rived structures most affected in 22q11 deletion able that functional disruption of HIRA contributes dHAND. Nat. Genet. 16, 154–160 1357-4310/00/$ - see front matter © 2000 Elsevier Science Ltd. All rights reserved. PII: S1357-4310(99)01641 13 Letters MOLECULAR MEDICINE TODAY, JANUARY 2000 (VOL. 6) 5 Thomas, T. et al. (1998) A signaling cascade in- 8 Magnaghi P. et al. (1998) HIRA, a mammalian Depts of Pediatric Cardiology and Molecular volving endothelin-1, dHAND and Msx1 regulates homologue of Saccharomyces cerevisiae tran- Biology and Oncology, University of Texas development of neural crest-derived branchial arch scriptional co-repressors, interacts with Pax3. Nat. Southwestern Medical Center at Dallas, 6000 mesenchyme. Development 125, 3005–3014 Genet. 20, 74–77 Harry Hines Boulevard, Dallas, 6 Yamagishi, H. et al. (1999) A molecular pathway re- 9 Farrell M.J. et al. (1999) HIRA, a DiGeorge syn- TX 75235-9148, USA. vealing a genetic basis for human cardiac and cran- drome candidate gene, is required for cardiac out- iofacial defects. Science 283, 1158–1161 flow tract septation. Circ. Res. 84, 127–135 7 Lindsay, E.A. et al. (1999) Congenital heart disease Tel: +1 214 648 1246 in mice deficient for the DiGeorge syndrome region. Deepak Srivastava MD Fax: +1 214 648 1196 Nature 401, 379–383 Assistant Professor e-mail: [email protected] UFD1L is not the monogenic basis for heart defects associated with the CATCH phenotype* It is clearly possible that some aspects of the im- further reinforced the rejection of a defect in forward to generate a fragment spanning the portant and highly variable CATCH phenotype* UFD1L as ‘the cause’; using the Cre–Lox system, deleted region in order to clarify the breakpoints. are attributable to loss of a single allele. However, they have generated mice that are deleted for a We are not aware of these results being pub- we and others have devoted much effort to 1.2 Mb section of the murine region homologous lished. The one certainty is that this story is not searching for mutations in candidate genes in to the human deleted region, from Es2 to Ufd1l. yet complete. individuals with features suggestive of the Mice heterozygous for the Lox site inserts, in CATCH phenotype, without success until the ex- which the only genetic defect is disruption of Es2 References citing report by Yamagishi et al.1 of a 20 kb de- and Ufd1l, do not develop heart defects, but hemi- 01 Yamagishi, H. et al. (1999) A molecular pathway re- letion involving the first three exons of UFD1L. zygous cases manifest the classic heart malfor- vealing a genetic basis for human cardiac and cran- The fact that this gene shows appropriate ex- mations associated with DiGeorge syndrome, in iofacial defects. Science 283, 1158–1161 pression and was absent in a large series of particular, hypoplasia or apparent absence of the 02 Novelli, G. et al. (2000) Individual haploinsufficient cases was important supportive evidence but not fourth arch artery.
Recommended publications
  • Hira Peracha a Thesis
    DIAGNOSIS METHOD FOR MUCOPOLYSACCHARIDOSES by Hira Peracha A thesis submitted to the Faculty of the University of Delaware in partial fulfillment of the requirements for the degree of Bachelor of Arts in Biological Sciences with Distinction Spring 2018 © 2018 Hira Peracha All Rights Reserved i DIAGNOSIS METHOD FOR MUCOPOLYSACCHARIDOSES by Hira Peracha Approved: __________________________________________________________ Shunji Tomatsu, MD, Ph.D. Professor in charge of thesis on behalf of the Advisory Committee Approved: __________________________________________________________ Deni Galileo, Ph.D. Professor in charge of thesis on behalf of the Advisory Committee Approved: __________________________________________________________ Jessica Tanis, Ph.D. Committee member from the Department of Department Name Approved: __________________________________________________________ Barbara Settles, Ph.D. Committee member from the Board of Senior Thesis Readers Approved: __________________________________________________________ Michael Chajes, Ph.D. Chair of the University Committee on Student and Faculty Honors ii “Education is the key to unlocking the world, a passport to freedom.” -Oprah Winfrey iii ACKNOWLEDGMENTS I dedicate my thesis to my late grandfather, who taught me the value of life, hard work and sacrifice- especially in sickness and in health. I would like to express my deepest acknowledgements to my family. I could not have achieved all of my dreams and goals in life without my parents; my mom and my dad, whose sacrifices, love and support have been endless and infinite. Thank you for raising me to believe that anything was possible, and for spending your entire lives to better mine. To my younger siblings, Saarah, Hafsah and Arif, who have been the light in the dark times in my life and have always brought joy and happiness in my life.
    [Show full text]
  • Anti-Histone H2B Antibody (ARG41373)
    Product datasheet [email protected] ARG41373 Package: 100 μl anti-Histone H2B antibody Store at: -20°C Summary Product Description Rabbit Polyclonal antibody recognizes Histone H2B Tested Reactivity Hu, Ms, Rat Tested Application FACS, ICC/IF, IHC-P, IP, WB Host Rabbit Clonality Polyclonal Isotype IgG Target Name Histone H2B Antigen Species Human Immunogen Synthetic peptide derived from Human Histone H2B. Conjugation Un-conjugated Alternate Names Histone H2B type 1-K; H2BFAiii; H2BFT; H2B/S; H2B K; H2BK; HIRA-interacting protein 1 Application Instructions Application table Application Dilution FACS 1:50 ICC/IF 1:50 - 1:200 IHC-P 1:50 - 1:200 IP 1:30 WB 1:5000 - 1:20000 Application Note * The dilutions indicate recommended starting dilutions and the optimal dilutions or concentrations should be determined by the scientist. Calculated Mw 14 kDa Observed Size ~ 14 kDa Properties Form Liquid Purification Affinity purified. Buffer PBS (pH 7.4), 150 mM NaCl, 0.02% Sodium azide and 50% Glycerol. Preservative 0.02% Sodium azide Stabilizer 50% Glycerol Storage instruction For continuous use, store undiluted antibody at 2-8°C for up to a week. For long-term storage, aliquot www.arigobio.com 1/2 and store at -20°C. Storage in frost free freezers is not recommended. Avoid repeated freeze/thaw cycles. Suggest spin the vial prior to opening. The antibody solution should be gently mixed before use. Note For laboratory research only, not for drug, diagnostic or other use. Bioinformation Gene Symbol HIST1H2BK Gene Full Name histone cluster 1, H2bk Background Histones are basic nuclear proteins that are responsible for the nucleosome structure of the chromosomal fiber in eukaryotes.
    [Show full text]
  • Differential Regulation of the Histone Chaperone HIRA During Muscle Cell Differentiation by a Phosphorylation Switch
    OPEN Experimental & Molecular Medicine (2016) 48, e252; doi:10.1038/emm.2016.68 & 2016 KSBMB. All rights reserved 2092-6413/16 www.nature.com/emm ORIGINAL ARTICLE Differential regulation of the histone chaperone HIRA during muscle cell differentiation by a phosphorylation switch Jae-Hyun Yang1,4,5, Tae-Yang Song1,4, Chanhee Jo1,4, Jinyoung Park1, Han-Young Lee1, Ilang Song1, Suji Hong1, Kwan Young Jung1, Jaehoon Kim2, Jeung-Whan Han1, Hong-Duk Youn3 and Eun-Jung Cho1 Replication-independent incorporation of variant histone H3.3 has a profound impact on chromatin function and numerous cellular processes, including the differentiation of muscle cells. The histone chaperone HIRA and H3.3 have essential roles in MyoD regulation during myoblast differentiation. However, the precise mechanism that determines the onset of H3.3 deposition in response to differentiation signals is unclear. Here we show that HIRA is phosphorylated by Akt kinase, an important signaling modulator in muscle cells. By generating a phosphospecific antibody, we found that a significant amount of HIRA was phosphorylated in myoblasts. The phosphorylation level of HIRA and the occupancy of phosphorylated protein on muscle genes gradually decreased during cellular differentiation. Remarkably, the forced expression of the phosphomimic form of HIRA resulted in reduced H3.3 deposition and suppressed the activation of muscle genes in myotubes. Our data show that HIRA phosphorylation limits the expression of myogenic genes, while the dephosphorylation of HIRA is required for proficient H3.3 deposition and gene activation, demonstrating that the phosphorylation switch is exploited to modulate HIRA/H3.3-mediated muscle gene regulation during myogenesis.
    [Show full text]
  • Ubinuclein-1 Confers Histone H3.3-Specific-Binding by the HIRA
    ARTICLE Received 22 Apr 2015 | Accepted 1 Jun 2015 | Published 10 Jul 2015 DOI: 10.1038/ncomms8711 OPEN Ubinuclein-1 confers histone H3.3-specific-binding by the HIRA histone chaperone complex M. Daniel Ricketts1,2, Brian Frederick3, Henry Hoff3, Yong Tang3, David C. Schultz3, Taranjit Singh Rai4,5, Maria Grazia Vizioli4, Peter D. Adams4 & Ronen Marmorstein1,2,6 Histone chaperones bind specific histones to mediate their storage, eviction or deposition from/or into chromatin. The HIRA histone chaperone complex, composed of HIRA, ubinuclein-1 (UBN1) and CABIN1, cooperates with the histone chaperone ASF1a to mediate H3.3-specific binding and chromatin deposition. Here we demonstrate that the conserved UBN1 Hpc2-related domain (HRD) is a novel H3.3-specific-binding domain. Biochemical and biophysical studies show the UBN1-HRD preferentially binds H3.3/H4 over H3.1/H4. X-ray crystallographic and mutational studies reveal that conserved residues within the UBN1-HRD and H3.3 G90 as key determinants of UBN1–H3.3-binding specificity. Comparison of the structure with the unrelated H3.3-specific chaperone DAXX reveals nearly identical points of contact between the chaperone and histone in the proximity of H3.3 G90, although the mechanism for H3.3 G90 recognition appears to be distinct. This study points to UBN1 as the determinant of H3.3-specific binding and deposition by the HIRA complex. 1 Department of Biochemistry and Biophysics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA. 2 Graduate Group in Biochemistry and Molecular Biophysics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA.
    [Show full text]
  • HIRA Orchestrates a Dynamic Chromatin Landscape in Senescence and Is Required for Suppression of Neoplasia
    Downloaded from genesdev.cshlp.org on September 25, 2021 - Published by Cold Spring Harbor Laboratory Press HIRA orchestrates a dynamic chromatin landscape in senescence and is required for suppression of neoplasia Taranjit Singh Rai,1,2,3 John J. Cole,1,2,5 David M. Nelson,1,2,5 Dina Dikovskaya,1,2 William J. Faller,1 Maria Grazia Vizioli,1,2 Rachael N. Hewitt,1,2 Orchi Anannya,1 Tony McBryan,1,2 Indrani Manoharan,1,2 John van Tuyn,1,2 Nicholas Morrice,1 Nikolay A. Pchelintsev,1,2 Andre Ivanov,1,2,4 Claire Brock,1,2 Mark E. Drotar,1,2 Colin Nixon,1 William Clark,1 Owen J. Sansom,1 Kurt I. Anderson,1 Ayala King,1 Karen Blyth,1 and Peter D. Adams1,2 1Beatson Institute for Cancer Research, Bearsden, Glasgow G61 1BD, United Kingdom; 2Institute of Cancer Sciences, College of Medical, Veterinary, and Life Sciences, University of Glasgow, Glasgow G61 1BD, United Kingdom; 3Institute of Biomedical and Environmental Health Research, University of West of Scotland, Paisley PA1 2BE, United Kingdom Cellular senescence is a stable proliferation arrest that suppresses tumorigenesis. Cellular senescence and associated tumor suppression depend on control of chromatin. Histone chaperone HIRA deposits variant histone H3.3 and histone H4 into chromatin in a DNA replication-independent manner. Appropriately for a DNA replication-independent chaperone, HIRA is involved in control of chromatin in nonproliferating senescent cells, although its role is poorly defined. Here, we show that nonproliferating senescent cells express and incorporate histone H3.3 and other canonical core histones into a dynamic chromatin landscape.
    [Show full text]
  • A Catalog of Hemizygous Variation in 127 22Q11 Deletion Patients
    A catalog of hemizygous variation in 127 22q11 deletion patients. Matthew S Hestand, KU Leuven, Belgium Beata A Nowakowska, KU Leuven, Belgium Elfi Vergaelen, KU Leuven, Belgium Jeroen Van Houdt, KU Leuven, Belgium Luc Dehaspe, UZ Leuven, Belgium Joshua A Suhl, Emory University Jurgen Del-Favero, University of Antwerp Geert Mortier, Antwerp University Hospital Elaine Zackai, The Children's Hospital of Philadelphia Ann Swillen, KU Leuven, Belgium Only first 10 authors above; see publication for full author list. Journal Title: Human Genome Variation Volume: Volume 3 Publisher: Nature Publishing Group: Open Access Journals - Option B | 2016-01-14, Pages 15065-15065 Type of Work: Article | Final Publisher PDF Publisher DOI: 10.1038/hgv.2015.65 Permanent URL: https://pid.emory.edu/ark:/25593/rncxx Final published version: http://dx.doi.org/10.1038/hgv.2015.65 Copyright information: © 2016 Official journal of the Japan Society of Human Genetics This is an Open Access work distributed under the terms of the Creative Commons Attribution 4.0 International License (http://creativecommons.org/licenses/by/4.0/). Accessed September 28, 2021 7:41 PM EDT OPEN Citation: Human Genome Variation (2016) 3, 15065; doi:10.1038/hgv.2015.65 Official journal of the Japan Society of Human Genetics 2054-345X/16 www.nature.com/hgv ARTICLE A catalog of hemizygous variation in 127 22q11 deletion patients Matthew S Hestand1, Beata A Nowakowska1,2,Elfi Vergaelen1, Jeroen Van Houdt1,3, Luc Dehaspe3, Joshua A Suhl4, Jurgen Del-Favero5, Geert Mortier6, Elaine Zackai7,8, Ann Swillen1, Koenraad Devriendt1, Raquel E Gur8, Donna M McDonald-McGinn7,8, Stephen T Warren4, Beverly S Emanuel7,8 and Joris R Vermeesch1 The 22q11.2 deletion syndrome is the most common microdeletion disorder, with wide phenotypic variability.
    [Show full text]
  • Unraveling the Genetic and Developmental Mysteries of 22Q11 Deletion Syndrome
    Review TRENDS in Molecular Medicine Vol.9 No.9 September 2003 383 Unraveling the genetic and developmental mysteries of 22q11 deletion syndrome Hiroyuki Yamagishi1 and Deepak Srivastava2 1Department of Pediatrics, Keio University School of Medicine, 35 Shinanomachi, Shinjyuku-ku, Tokyo 160-8582, Japan 2Departments of Pediatrics and Molecular Biology, University of Texas Southwestern Medical Center, 6000 Harry Hines Blvd, Rm NA8.124, Dallas, TX 75390-9148, USA Birth defects occur in nearly 5% of all live births and are susceptibilities that predispose individuals to a variety the major cause of infant mortality and morbidity. of birth defects. Here, we review the recent advances that Despite the recent progress in molecular and develop- have begun to reveal the human genetic, developmental mental biology, the underlying genetic etiology of most biology and molecular biology underpinnings of this congenital anomalies remains unknown. Heterozygous common syndrome. deletion of the 22q11.2 locus results in the most com- mon human genetic deletion syndrome, known as Clinical features of 22q11DS DiGeorge syndrome, and has served as an entry to The clinical findings associated with del22q11 are highly understanding the basis for numerous congenital heart variable. Approximately 75% of patients with 22q11DS are and craniofacial anomalies, among many other defects. born with congenital heart defects (CHD), mainly of the Extensive human genetic analyses, mouse modeling cardiac outflow tract and aortic arch. Other common and studies of developmental molecular cascades features of 22q11DS include a characteristic facial involved in 22q11 deletion syndrome are revealing com- appearance, immunodeficiency from thymic hypoplasia, plex networks of signaling and transcriptional events velopharyngeal dysfunction with or without cleft palate, that are essential for normal embryonic development.
    [Show full text]
  • The Death-Associated Protein DAXX Is a Novel Histone Chaperone Involved in the Replication-Independent Deposition of H3.3
    Downloaded from genesdev.cshlp.org on October 3, 2021 - Published by Cold Spring Harbor Laboratory Press The death-associated protein DAXX is a novel histone chaperone involved in the replication-independent deposition of H3.3 Pascal Drane´,1 Khalid Ouararhni, Arnaud Depaux, Muhammad Shuaib, and Ali Hamiche2 IGMBC (Institut de Ge´ne´tique et de Biologie Mole´culaire et Cellulaire), Illkirch F-67400, France; CNRS, UMR7104, Illkirch F-67404, France; Inserm, U964, Illkirch F-67400, France; and Universite´ de Strasbourg, Strasbourg F-67000, France The histone variant H3.3 marks active chromatin by replacing the conventional histone H3.1. In this study, we investigate the detailed mechanism of H3.3 replication-independent deposition. We found that the death domain- associated protein DAXX and the chromatin remodeling factor ATRX (a-thalassemia/mental retardation syndrome protein) are specifically associated with the H3.3 deposition machinery. Bacterially expressed DAXX has a marked binding preference for H3.3 and assists the deposition of (H3.3–H4)2 tetramers on naked DNA, thus showing that DAXX is a H3.3 histone chaperone. In DAXX-depleted cells, a fraction of H3.3 was found associated with the replication-dependent machinery of deposition, suggesting that cells adapt to the depletion. The reintroduced DAXX in these cells colocalizes with H3.3 into the promyelocytic leukemia protein (PML) bodies. Moreover, DAXX associates with pericentric DNA repeats, and modulates the transcription from these repeats through assembly of H3.3 nucleosomes. These findings establish a new link between the PML bodies and the regulation of pericentric DNA repeat chromatin structure. Taken together, our data demonstrate that DAXX functions as a bona fide histone chaperone involved in the replication-independent deposition of H3.3.
    [Show full text]
  • Modeling Chromosomes in Mouse to Explore the Function of Genes
    Review Modeling Chromosomes in Mouse to Explore the Function of Genes, Genomic Disorders, and Chromosomal Organization Ve´ronique Brault, Patricia Pereira, Arnaud Duchon, Yann He´rault* ABSTRACT chromosomes using microcell-mediated chromosome transfer (MMCT) offers the opportunity to study the function ne of the challenges of genomic research after the of large genes or clusters of genes and provides more and completion of the human genome project is to more mouse models to study human pathologies such as O assign a function to all the genes and to understand contiguous gene syndromes. In this review, we describe the their interactions and organizations. Among the various panel of techniques available for chromosome engineering in techniques, the emergence of chromosome engineering tools the mouse, some of their applications for studying gene with the aim to manipulate large genomic regions in the function and genomic organization and for modeling human mouse model offers a powerful way to accelerate the diseases, and the implications for future research. discovery of gene functions and provides more mouse models to study normal and pathological developmental processes Chemical and Radiation-Induced Chromosome associated with aneuploidy. The combination of gene Rearrangements targeting in ES cells, recombinase technology, and other Historically, various types of rearrangements including techniques makes it possible to generate new chromosomes deletions, inversions, and reciprocal translocations were carrying specific and defined deletions, duplications, obtained through irradiation or chemical mutagenesis. Such inversions, and translocations that are accelerating functional chromosomal configurations are important tools for looking analysis. This review presents the current status of at recessive lethal mutations in mice [16] or to obtain mouse chromosome engineering techniques and discusses the models of partial aneuploidy.
    [Show full text]
  • 13307 HIRA (D2A5E) Rabbit Mab
    Revision 1 C 0 2 - t HIRA (D2A5E) Rabbit mAb a e r o t S Orders: 877-616-CELL (2355) [email protected] 7 Support: 877-678-TECH (8324) 0 3 Web: [email protected] 3 www.cellsignal.com 1 # 3 Trask Lane Danvers Massachusetts 01923 USA For Research Use Only. Not For Use In Diagnostic Procedures. Applications: Reactivity: Sensitivity: MW (kDa): Source/Isotype: UniProt ID: Entrez-Gene Id: WB H M Mk Endogenous 112 Rabbit IgG P54198 7290 Product Usage Information 4. Jin, C. and Felsenfeld, G. (2007) Genes Dev 21, 1519-29. 5. Goldberg, A.D. et al. (2010) Cell 140, 678-91. Application Dilution 6. Wong, L.H. et al. (2010) Genome Res 20, 351-60. 7. Wong, L.H. et al. (2009) Genome Res 19, 404-14. Western Blotting 1:1000 8. Zhang, R. et al. (2007) Mol Cell Biol 27, 2343-58. 9. Wilming, L.G. et al. (1997) Hum Mol Genet 6, 247-58. Storage Supplied in 10 mM sodium HEPES (pH 7.5), 150 mM NaCl, 100 µg/ml BSA, 50% glycerol and less than 0.02% sodium azide. Store at –20°C. Do not aliquot the antibody. Specificity / Sensitivity HIRA (D2A5E) Rabbit mAb recognizes endogenous levels of total HIRA protein. Species Reactivity: Human, Mouse, Monkey Source / Purification Monoclonal antibody is produced by immunizing animals with a synthetic peptide corresponding to residues surrounding Leu593 of human HIRA protein. Background Histone cell cycle regulation defective homolog A (HIRA), also known as TUP1-like enhancer of split protein 1 (TUPLE1), is the mammalian homolog of the yeast HIR1 and HIR2 transcriptional repressor proteins (1).
    [Show full text]
  • Atypical Deletions Suggest Five 22Q11.2 Critical Regions
    European Journal of Human Genetics (1999) 7, 903–909 © 1999 Stockton Press All rights reserved 1018–4813/99 $15.00 t http://www.stockton-press.co.uk/ejhg ARTICLE Atypical deletions suggest five 22q11.2 critical regions related to the DiGeorge/velo-cardio- facial syndrome Francesca Amati1, Emanuela Conti1, Antonio Novelli1, Mario Bengala1, Maria Cristina Digilio2, Bruno Marino2, Aldo Giannotti2, Orazio Gabrielli3, Giuseppe Novelli1 and Bruno Dallapiccola1 1Dipartimento di Biopatologia e Diagnostica per Immagini, Universit`a Tor Vergata and CSS-Mendel, Rome 2Ospedale Pediatrico Bambino Ges`u, Rome 3Clinica Pediatrica, Universit`a di Ancona, Italy Deletions of chromosome 22q11.2 have been associated with distinct phenotypes including DiGeorge syndrome (DGS) and velo-cardio-facial (VCFS) syndrome. These diseases result from a failure to form derivatives of the third and fourth branchial arches during development. DGS/VCFS deletions usually encompass about 3 Mb of genomic DNA in more than 90% of patients. However, deletion mapping studies have failed to demonstrate the existence of a single small region of overlap (SRO) and ruled out any obvious correlation between site or size of deletion and severity of clinical phenotype. We describe three patients carrying ‘atypical’ deletions presenting the DGS/VCFS phenotype. A comparative analysis of deletions in our patients and those previously published has suggested the existence of five distinct critical regions within the 22q11.2 locus. This observation argues that DGS/VCFS results from haploinsufficiency
    [Show full text]
  • Molecular Cloning of the B4GALNT2 Gene and Its Single Nucleotide Polymorphisms Association with Litter Size in Small Tail Han Sheep
    animals Article Molecular Cloning of the B4GALNT2 Gene and Its Single Nucleotide Polymorphisms Association with Litter Size in Small Tail Han Sheep Xiaofei Guo 1,2,† , Xiangyu Wang 1,†, Benmeng Liang 1, Ran Di 1, Qiuyue Liu 1, Wenping Hu 1, Xiaoyun He 1, Jinlong Zhang 2, Xiaosheng Zhang 2 and Mingxing Chu 1,* 1 Key Laboratory of Animal Genetics and Breeding and Reproduction of Ministry of Agriculture, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing 100193, China; [email protected] (X.G.); [email protected] (X.W.); [email protected] (B.L.); [email protected] (R.D.); [email protected] (Q.L.); [email protected] (W.H.); [email protected] (X.H.) 2 Tianjin Institute of Animal Sciences, Tianjin 300381, China; [email protected] (J.Z.); [email protected] (X.Z.) * Correspondence: [email protected]; Tel.: +86-01-062-819-850 † These authors contributed equally to this study. Received: 1 August 2018; Accepted: 15 September 2018; Published: 20 September 2018 Simple Summary: In French Lacaune sheep, the B4GALNT2 (beta-1, 4-N-acetyl-galactosaminyl transferase 2) gene was considered as the potential gene for a FecL (mutation), which regulates the ovine ovulation rate. Three specific mutation sites linked with the FecL mutation have not been previously found in 11 sheep breeds. However, two mutations of g.36946470C > T and g.36933082C > T in the exon of B4GALNT2 were found to have had a significant effect on the litter size in the first parity for Small Tail Han (STH) Sheep (p < 0.05). B4GALNT2, which is mainly expressed in ovine ovary, also plays an important role in sheep reproduction.
    [Show full text]