Using Hidden Markov Models to Investigate G- Quadruplex Motifs in Genomic Sequences

Total Page:16

File Type:pdf, Size:1020Kb

Using Hidden Markov Models to Investigate G- Quadruplex Motifs in Genomic Sequences Yano and Kato BMC Genomics 2014, 15(Suppl 9):S15 http://www.biomedcentral.com/1471-2164/15/S9/S15 RESEARCH Open Access Using hidden Markov models to investigate G- quadruplex motifs in genomic sequences Masato Yano1, Yuki Kato2* From Asia Pacific Bioinformatics Network (APBioNet) Thirteenth International Conference on Bioinformatics (InCoB2014) Sydney, Australia. 31 July - 2 August 2014 Abstract Background: G-quadruplexes are four-stranded structures formed in guanine-rich nucleotide sequences. Several functional roles of DNA G-quadruplexes have so far been investigated, where their putative functional roles during DNA replication and transcription have been suggested. A necessary condition for G-quadruplex formation is the presence of four regions of tandem guanines called G-runs and three nucleotide subsequences called loops that connect G-runs. A simple computational way to detect potential G-quadruplex regions in a given genomic sequence is pattern matching with regular expression. Although many putative G-quadruplex motifs can be found in most genomes by the regular expression-based approach, the majority of these sequences are unlikely to form G- quadruplexes because they are unstable as compared with canonical double helix structures. Results: Here we present elaborate computational models for representing DNA G-quadruplex motifs using hidden Markov models (HMMs). Use of HMMs enables us to evaluate G-quadruplex motifs quantitatively by a probabilistic measure. In addition, the parameters of HMMs can be trained by using experimentally verified data. Computational experiments in discriminating between positive and negative G-quadruplex sequences as well as reducing putative G- quadruplexes in the human genome were carried out, indicating that HMM-based models can discern bona fide G- quadruplex structures well and one of them has the possibility of reducing false positive G-quadruplexes predicted by existing regular expression-based methods. Furthermore, our results show that one of our models can be specialized to detect G-quadruplex sequences whose functional roles are expected to be involved in DNA transcription. Conclusions: The HMM-based method along with the conventional pattern matching approach can contribute to reducing costly and laborious wet-lab experiments to perform functional analysis on a given set of potential G- quadruplexes of interest. The C++ and Perl programs are available at http://tcs.cira.kyoto-u.ac.jp/~ykato/program/ g4hmm/. Background in the antiparallel way. In contrast, several in vitro experi- Deoxyribonucleic acids (DNAs) are macromolecules that ments reveal the existence of non-B-DNA structures hold genetic information in almost all of the organisms. caused by particular sequence motifs and DNA-protein The bulk of existing DNA molecules is assumed to form a interactions. Well investigated examples include G-quad- right-handed double helical structure called B-DNA [1], ruplex (G4), Z-DNA, cruciform and triplex. Recent where each constituent bases A and C selectively bind to advances in providing in vitro evidence of these specific bases T and G, respectively, between two strands arranged structures develop the hypothesis that these structures are considered to have some functional roles in living cells [2]. A G4 structure is one of the topological conformations * Correspondence: [email protected] 2Center for iPS Cell Research and Application (CiRA), Kyoto University, 53 that DNAs can adopt, where G-quartets, hydrogen-bonded Kawahara-cho, Shogoin, Sakyo-ku, Kyoto 606-8507, Japan square planar substructures between four guanines (Gs), Full list of author information is available at the end of the article © 2014 Yano and Kato; licensee BioMed Central Ltd. This is an Open Access article distributed under the terms of the Creative Commons Attribution License (http://creativecommons.org/licenses/by/4.0), which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated. Yano and Kato BMC Genomics 2014, 15(Suppl 9):S15 Page 2 of 11 http://www.biomedcentral.com/1471-2164/15/S9/S15 Since little is known about the functions of G4 struc- tures and genome-scale wet-lab experiments with nuclear magnetic resonance (NMR) spectroscopy for structural analysis [9] are not feasible, several computational efforts have been made on identifying the locations of potential G4 sequences in genomic DNAs and inferring their func- tions by comparative sequence analysis using related genes with known functions [10,11]. In principle, G4 motifs can be represented by a regular expression G+N *G+N*G+N*G+, where ‘N’ shows an arbitrary base including G, ‘+’ denotes ‘ ’ Figure 1 An example of G4 structure.(a)Fourguaninesform at least one repeat of the preceding symbol and * means hydrogen bonds to their adjacent guanines, resulting in a G-quartet. at least zero repeats. Due to this simple pattern of G4 (b) Several G-quartets are stacked onto each other to form a G- motifs, several in silico methods have been proposed to quadruplex (G4) structure. A G4 structure has its sequence as detect G4 sequences in genomes using pattern matching alternate G-runs and loops. with regular expression [12-16]. Moreover, regular expres- sion-based methods that incorporate a simple scoring scheme are proposed [17-19]. Another computational study focuses on thermodynamic stability of G4 structures are stacked onto each other (see Figure 1). At the using Gaussian process regression [20]. Although the pat- sequence level, a G4 sequence can be represented by four tern matching approaches can detect many G4 motifs in regions of consecutive Gs that form G-quartets, called G- genomic sequences quite fast, it is pointed out that the runs,andthreeregionsofnucleotide subsequences that majorityofthesemotifsmaybefalsepositiveG4 connect G-runs, called loops, which can have varying sequences [21,22]. length including lack of loop [3]. Note that consecutive Gs In this contribution, we present more elaborate compu- may stretch over G-run and loop regions, making the pro- tational models than regular expression to represent G4 blem of predicting G-run regions somewhat complex. motifs, employing hidden Markov models (HMMs). Loops are known to play an important role in stability of HMMs are so flexible in modeling linear dependence that G4 structures [4]. In addition, it is pointed out that the they are widely used in bioinformatics including protein loop length affects therapeutic selectivity to target a G4 secondary structure prediction [23,24] and sequence motif instead of the topology of the G4 structure [5]. G4 struc- search [25]. To model G4 motifs, we provide four HMM- tures are also stabilized by monovalent cations, especially based models from the viewpoint of the number of hidden K+, located in the central cavities in the stack. states that describe G-runs and loops, and compare with Eukyariotic telomeric sequences include G-rich regions each other in three computational experiments. The first and they can form G4 structures in vitro. However, the preliminary experiment in predicting G-run regions in a question of how many such G-rich regions can actually set of 100 real G4 sequences in the literature [20] indicates form G4 structures in vivo has not been resolved. The that each HMM-based model can represent actual G-run potential to form G4 structures in telomeric sequences in regions well. The subsequent experiment in discriminating vivo can be shown by in vitro DNA binding experiments real and shuffled G4 sequences by using HMMs shows with those sequences [2]. For example, telomere end-bind- that the models considering detailed distributions of G- ing proteins in ciliates can control the formation of G4 run and loop lengths can outperform the simple probabil- DNA structures at telomeres [6]. Interestingly, however, a istic extension of regular expression. In the third test with recent study suggests that endogenous G4 structures in statistical analysis in discriminating highly likely G4 struc- human cells are present largely outside the telomeres [7]. tures from putative G4 motifs in the human pre-mRNA Another work reports that protruding nucleotides in sequences [26], the results show that the HMM-based human telomeric sequences destabilize the G4 structure model that can represent elaborate length distribution of and overhanging sequences influence the folding of the G-run regions outperforms the other three models pre- quadruplex [8]. Other examples of G-rich regions in gen- sented in this work. Moreover, the above model can be omes are transcriptional start sites, mitotic and meiotic specialized to detect G4 sequences whose functional roles double strand break sites. Although G4 structures have are expected to be involved in DNA transcription. Finally, stability with higher temperature than that of canonical this model in conjunction with pattern search is applied to double helix structures, many functional regions in geno- G4 screening in the whole human genome, producing a mic sequences have not a few G-rich motifs [2], motivat- considerably smaller number of G4 candidates with statis- ing us to investigate further the functional roles of G4 tical significance than that of G4 sequences predicted by structures. pattern matching alone. Yano and Kato BMC Genomics 2014, 15(Suppl 9):S15 Page 3 of 11
Recommended publications
  • The N-Cadherin Interactome in Primary Cardiomyocytes As Defined Using Quantitative Proximity Proteomics Yang Li1,*, Chelsea D
    © 2019. Published by The Company of Biologists Ltd | Journal of Cell Science (2019) 132, jcs221606. doi:10.1242/jcs.221606 TOOLS AND RESOURCES The N-cadherin interactome in primary cardiomyocytes as defined using quantitative proximity proteomics Yang Li1,*, Chelsea D. Merkel1,*, Xuemei Zeng2, Jonathon A. Heier1, Pamela S. Cantrell2, Mai Sun2, Donna B. Stolz1, Simon C. Watkins1, Nathan A. Yates1,2,3 and Adam V. Kwiatkowski1,‡ ABSTRACT requires multiple adhesion, cytoskeletal and signaling proteins, The junctional complexes that couple cardiomyocytes must transmit and mutations in these proteins can cause cardiomyopathies (Ehler, the mechanical forces of contraction while maintaining adhesive 2018). However, the molecular composition of ICD junctional homeostasis. The adherens junction (AJ) connects the actomyosin complexes remains poorly defined. – networks of neighboring cardiomyocytes and is required for proper The core of the AJ is the cadherin catenin complex (Halbleib and heart function. Yet little is known about the molecular composition of the Nelson, 2006; Ratheesh and Yap, 2012). Classical cadherins are cardiomyocyte AJ or how it is organized to function under mechanical single-pass transmembrane proteins with an extracellular domain that load. Here, we define the architecture, dynamics and proteome of mediates calcium-dependent homotypic interactions. The adhesive the cardiomyocyte AJ. Mouse neonatal cardiomyocytes assemble properties of classical cadherins are driven by the recruitment of stable AJs along intercellular contacts with organizational and cytosolic catenin proteins to the cadherin tail, with p120-catenin β structural hallmarks similar to mature contacts. We combine (CTNND1) binding to the juxta-membrane domain and -catenin β quantitative mass spectrometry with proximity labeling to identify the (CTNNB1) binding to the distal part of the tail.
    [Show full text]
  • Ahnaks Are a Class of Giant Propeller-Like Proteins That Associate with Calcium Channel Proteins of Cardiomyocytes and Other Cells
    The AHNAKs are a class of giant propeller-like proteins that associate with calcium channel proteins of cardiomyocytes and other cells Akihiko Komuro*, Yutaka Masuda*, Koichi Kobayashi, Roger Babbitt, Murat Gunel, Richard A. Flavell, and Vincent T. Marchesi† Departments of Pathology and Immunobiology, Boyer Center for Molecular Medicine, Yale University School of Medicine, New Haven, CT 06510 Contributed by Vincent T. Marchesi, December 31, 2003 To explore the function of the giant AHNAK molecule, first de- mechanisms, one operating at the cell surface in collaboration with scribed in 1992 [Shtivelman, E., Cohen, F. E. & Bishop, J. M. (1992) calcium channels, and the second, PLC activation, which is a process Proc. Natl. Acad. Sci. USA 89, 5472–5476], we created AHNAK null that could potentially take place at multiple points throughout the mice by homologous recombination. Homozygous knockouts cell. showed no obvious phenotype, but revealed instead a second The arrangement of channel proteins at the cell surface is AHNAK-like molecule, provisionally designated AHNAK2. Like the believed to be controlled by multidomain polypeptides known as original AHNAK, AHNAK2 is a 600-kDa protein composed of a large scaffolding proteins that link together activated channels at specific number of highly conserved repeat segments. Structural predic- points on the membrane surface. Scaffolding proteins also coordi- tions suggest that the repeat segments of both AHNAKs may have nate the activities of multienzyme complexes by physically linking as their basic framework a series of linked, antiparallel ␤-strands them together, and as in the case with AHNAK, they are often similar to those found in ␤-propeller proteins.
    [Show full text]
  • Identification of Cis-Regulatory Sequence
    Worsley-Hunt et al. Genome Medicine 2011, 3:65 http://genomemedicine.com/content/3/9/65 REVIEW Identication of cis-regulatory sequence variations in individual genome sequences Rebecca Worsley-Hunt1,2, Virginie Bernard1 and Wyeth W Wasserman1* Abstract for the appropriate patterning or magnitude of gene expression. Similarly, mutations can disrupt other sequence- Functional contributions of cis-regulatory sequence specific regulatory controls, such as elements regulating variations to human genetic disease are numerous. For RNA splicing or stability. Although much emphasis in the instance, disrupting variations in a HNF4A transcription age of exome sequencing has been placed on variation factor binding site upstream of the Factor IX gene within protein-encoding sequences, it is apparent that contributes causally to hemophilia B Leyden. Although regulatory sequence disruptions will become a key focus clinical genome sequence analysis currently focuses as full genome sequences become widely accessible for on the identication of protein-altering variation, the medical genetics research. impact of cis-regulatory mutations can be similarly e emerging collection of cis-regulatory variations strong. New technologies are now enabling genome that cause human disease or altered phenotype is grow- sequencing beyond exomes, revealing variation across ing [2-4]. Reports identify cis-acting, expression-altering the non-coding 98% of the genome responsible for mutations observed within introns, far upstream of developmental and physiological patterns of gene genes, at splicing sites or within microRNA target sites. activity. The capacity to identify causal regulatory For example, cis-regulatory mutations have roles in hemo- mutations is improving, but predicting functional philia, Gilbert’s syndrome, Bernard-Soulier syndrome, changes in regulatory DNA sequences remains a great irritable bowel syndrome, beta-thalassemia, cholesterol challenge.
    [Show full text]
  • Transcriptome-Wide Profiling of Cerebral Cavernous Malformations
    www.nature.com/scientificreports OPEN Transcriptome-wide Profling of Cerebral Cavernous Malformations Patients Reveal Important Long noncoding RNA molecular signatures Santhilal Subhash 2,8, Norman Kalmbach3, Florian Wegner4, Susanne Petri4, Torsten Glomb5, Oliver Dittrich-Breiholz5, Caiquan Huang1, Kiran Kumar Bali6, Wolfram S. Kunz7, Amir Samii1, Helmut Bertalanfy1, Chandrasekhar Kanduri2* & Souvik Kar1,8* Cerebral cavernous malformations (CCMs) are low-fow vascular malformations in the brain associated with recurrent hemorrhage and seizures. The current treatment of CCMs relies solely on surgical intervention. Henceforth, alternative non-invasive therapies are urgently needed to help prevent subsequent hemorrhagic episodes. Long non-coding RNAs (lncRNAs) belong to the class of non-coding RNAs and are known to regulate gene transcription and involved in chromatin remodeling via various mechanism. Despite accumulating evidence demonstrating the role of lncRNAs in cerebrovascular disorders, their identifcation in CCMs pathology remains unknown. The objective of the current study was to identify lncRNAs associated with CCMs pathogenesis using patient cohorts having 10 CCM patients and 4 controls from brain. Executing next generation sequencing, we performed whole transcriptome sequencing (RNA-seq) analysis and identifed 1,967 lncRNAs and 4,928 protein coding genes (PCGs) to be diferentially expressed in CCMs patients. Among these, we selected top 6 diferentially expressed lncRNAs each having signifcant correlative expression with more than 100 diferentially expressed PCGs. The diferential expression status of the top lncRNAs, SMIM25 and LBX2-AS1 in CCMs was further confrmed by qRT-PCR analysis. Additionally, gene set enrichment analysis of correlated PCGs revealed critical pathways related to vascular signaling and important biological processes relevant to CCMs pathophysiology.
    [Show full text]
  • Mir-17-92 Fine-Tunes MYC Expression and Function to Ensure
    ARTICLE Received 31 Mar 2015 | Accepted 22 Sep 2015 | Published 10 Nov 2015 DOI: 10.1038/ncomms9725 OPEN miR-17-92 fine-tunes MYC expression and function to ensure optimal B cell lymphoma growth Marija Mihailovich1, Michael Bremang1, Valeria Spadotto1, Daniele Musiani1, Elena Vitale1, Gabriele Varano2,w, Federico Zambelli3, Francesco M. Mancuso1,w, David A. Cairns1,w, Giulio Pavesi3, Stefano Casola2 & Tiziana Bonaldi1 The synergism between c-MYC and miR-17-19b, a truncated version of the miR-17-92 cluster, is well-documented during tumor initiation. However, little is known about miR-17-19b function in established cancers. Here we investigate the role of miR-17-19b in c-MYC-driven lymphomas by integrating SILAC-based quantitative proteomics, transcriptomics and 30 untranslated region (UTR) analysis upon miR-17-19b overexpression. We identify over one hundred miR-17-19b targets, of which 40% are co-regulated by c-MYC. Downregulation of a new miR-17/20 target, checkpoint kinase 2 (Chek2), increases the recruitment of HuR to c- MYC transcripts, resulting in the inhibition of c-MYC translation and thus interfering with in vivo tumor growth. Hence, in established lymphomas, miR-17-19b fine-tunes c-MYC activity through a tight control of its function and expression, ultimately ensuring cancer cell homeostasis. Our data highlight the plasticity of miRNA function, reflecting changes in the mRNA landscape and 30 UTR shortening at different stages of tumorigenesis. 1 Department of Experimental Oncology, European Institute of Oncology, Via Adamello 16, Milan 20139, Italy. 2 Units of Genetics of B cells and lymphomas, IFOM, FIRC Institute of Molecular Oncology Foundation, Milan 20139, Italy.
    [Show full text]
  • CCM1 and CCM2 Variants in Patients with Cerebral Cavernous
    www.nature.com/scientificreports OPEN CCM1 and CCM2 variants in patients with cerebral cavernous malformation in an ethnically Received: 19 January 2018 Accepted: 11 July 2019 Chinese population in Taiwan Published: xx xx xxxx Chun-Wei Chang 1, Peng-Wei Hsu2,3, Kuo-Chen Wei2,3, Chia-Wen Chang1, Hon-Chung Fung1, Mo-Song Hsih1, Wen-Chuin Hsu1,3, Long-Sun Ro1,3, Chen-Nen Chang2,4, Jiun-Jie Wang5,6, Yih-Ru Wu1,3 & Sien-Tsong Chen1 Cerebral cavernous malformation (CCM) is a vascular malformation characterized by clustered enlarged capillary-like channels in the central nervous system. The genes harboring variants in patients with CCM include CCM1/Krev interaction trapped-1, CCM2/MGC4607, and CCM3/programmed cell death protein 10. We aimed to identify pathogenic variants in an ethnic Chinese population in Taiwan. We recruited 95 patients with multiple CCMs or a single lesion with a relevant family history. Sanger sequencing was performed for 41 patients. Variants were identifed using sequence alignment tools, and the clinical signifcance of these variants was determined using American College of Medical Genetics and Genomics standards and guidelines. Several pathogenic variants were found in six patients, including three unrelated patients and three afected members of one family. Two novel pathogenic variants leading to early truncation comprised a deletion variant in exon 18 of CCM1 (c.1846delA; p.Glu617LysfsTer44) and an insertion variant in exon 4 of CCM2 (c.401_402insGCCC; p.Ile136AlafsTer4). One novel pathogenic splice site variant was c.485 + 1G > C at the beginning of intron 8 of CCM1. In this study, we identifed novel variants related to CCM in an ethnically Chinese population in Taiwan.
    [Show full text]
  • Overexpression of RNF38 Facilitates TGF-Β Signaling by Ubiquitinating
    Peng et al. Journal of Experimental & Clinical Cancer Research (2019) 38:113 https://doi.org/10.1186/s13046-019-1113-3 RESEARCH Open Access Overexpression of RNF38 facilitates TGF-β signaling by Ubiquitinating and degrading AHNAK in hepatocellular carcinoma Rui Peng1,2†, Peng-Fei Zhang1,3†, Xuan Yang1†, Chuan-Yuan Wei1†, Xiao-Yong Huang1, Jia-Bin Cai1, Jia-Cheng Lu1, Chao Gao1, Hai-Xiang Sun1, Qiang Gao1, Dou-Sheng Bai2, Guo-Ming Shi1*, Ai-Wu Ke1* and Jia Fan1,4* Abstract Background: RING finger protein 38 (RNF38), a member of the RNF protein family, has just emerged as a vital driver of cancer progression. However, the oncogenic mechanisms of RNF38 remain unexplored. Methods: Using frozen tumor tissue and tissue microarray from hepatocellular carcinoma (HCC) patients, we tried to probe the expression of RNF38 in HCC and its clinical value. Then the biological functions of RNF38 were analyzed in vivo and vitro. Stable isotope labeling with amino acids (SILAC) in cell culture and co-immunoprecipitation proteomic analyses were combined to reveal the potential mechanism of RNF38 in HCC progression. Results: We report that RNF38 expression was markedly higher in HCC tissues than in peritumor tissues. Correspondingly, RNF38 overexpression promoted the HCC cell migration and invasion and inhibited apoptosis both in vitro and in vivo. And elevated RNF38 expression induced HCC cell epithelial-mesenchymal transition by facilitating transforming growth factor-β (TGF-β) signaling via ubiquitinating and degrading neuroblast differentiation-associated protein (AHNAK), a well-established inhibitor of TGF-β signaling. Furthermore, AHNAK interference restored the HCC cell invasion and metastasis deprived by RNF38 downregulation.
    [Show full text]
  • CD29 Identifies IFN-Γ–Producing Human CD8+ T Cells with an Increased Cytotoxic Potential
    + CD29 identifies IFN-γ–producing human CD8 T cells with an increased cytotoxic potential Benoît P. Nicoleta,b, Aurélie Guislaina,b, Floris P. J. van Alphenc, Raquel Gomez-Eerlandd, Ton N. M. Schumacherd, Maartje van den Biggelaarc,e, and Monika C. Wolkersa,b,1 aDepartment of Hematopoiesis, Sanquin Research, 1066 CX Amsterdam, The Netherlands; bLandsteiner Laboratory, Oncode Institute, Amsterdam University Medical Center, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands; cDepartment of Research Facilities, Sanquin Research, 1066 CX Amsterdam, The Netherlands; dDivision of Molecular Oncology and Immunology, Oncode Institute, The Netherlands Cancer Institute, 1066 CX Amsterdam, The Netherlands; and eDepartment of Molecular and Cellular Haemostasis, Sanquin Research, 1066 CX Amsterdam, The Netherlands Edited by Anjana Rao, La Jolla Institute for Allergy and Immunology, La Jolla, CA, and approved February 12, 2020 (received for review August 12, 2019) Cytotoxic CD8+ T cells can effectively kill target cells by producing therefore developed a protocol that allowed for efficient iso- cytokines, chemokines, and granzymes. Expression of these effector lation of RNA and protein from fluorescence-activated cell molecules is however highly divergent, and tools that identify and sorting (FACS)-sorted fixed T cells after intracellular cytokine + preselect CD8 T cells with a cytotoxic expression profile are lacking. staining. With this top-down approach, we performed an un- + Human CD8 T cells can be divided into IFN-γ– and IL-2–producing biased RNA-sequencing (RNA-seq) and mass spectrometry cells. Unbiased transcriptomics and proteomics analysis on cytokine- γ– – + + (MS) analyses on IFN- and IL-2 producing primary human producing fixed CD8 T cells revealed that IL-2 cells produce helper + + + CD8 Tcells.
    [Show full text]
  • Cerebral Cavernous Malformations: from Genes to Proteins to Disease
    See the corresponding editorial in this issue, pp 119–121. J Neurosurg 116:122–132, 2012 Cerebral cavernous malformations: from genes to proteins to disease Clinical article DANIEL D. CAVALCANTI, M.D., M. YASHAR S. KALANI, M.D., PH.D., NIKOLAY L. MARTIROSYAN, M.D., JUSTIN EALES, B.S., ROBERT F. SPETZLER, M.D., AND MARK C. PREUL, M.D. Division of Neurological Surgery, Barrow Neurological Institute, St. Joseph’s Hospital and Medical Center, Phoenix, Arizona Over the past half century molecular biology has led to great advances in our understanding of angio- and vas- culogenesis and in the treatment of malformations resulting from these processes gone awry. Given their sporadic and familial distribution, their developmental and pathological link to capillary telangiectasias, and their observed chromosomal abnormalities, cerebral cavernous malformations (CCMs) are regarded as akin to cancerous growths. Although the exact pathological mechanisms involved in the formation of CCMs are still not well understood, the identification of 3 genetic loci has begun to shed light on key developmental pathways involved in CCM pathogen- esis. Cavernous malformations can occur sporadically or in an autosomal dominant fashion. Familial forms of CCMs have been attributed to mutations at 3 different loci implicated in regulating important processes such as proliferation and differentiation of angiogenic precursors and members of the apoptotic machinery. These processes are important for the generation, maintenance, and pruning of every vessel in the body. In this review the authors highlight the lat- est discoveries pertaining to the molecular genetics of CCMs, highlighting potential new therapeutic targets for the treatment of these lesions.
    [Show full text]
  • A Focus on Genetic Features of Cerebral Cavernous Malformations and Brain Arteriovenous Malformations Pathogenesis
    Neurological Sciences (2019) 40:243–251 https://doi.org/10.1007/s10072-018-3674-x REVIEW ARTICLE Vis-à-vis: a focus on genetic features of cerebral cavernous malformations and brain arteriovenous malformations pathogenesis Concetta Scimone1,2 & Luigi Donato 1,2 & Silvia Marino3 & Concetta Alafaci1 & Rosalia D’Angelo1 & Antonina Sidoti1,2 Received: 7 September 2018 /Accepted: 1 December 2018 /Published online: 6 December 2018 # Fondazione Società Italiana di Neurologia 2018 Abstract Cerebrovascular malformations include a wide range of blood vessel disorders affecting brain vasculature. Neuroimaging differential diagnosis can result unspecific due to similar phenotypes of lesions and their deep locali- zation. Next-generation sequencing (NGS) platforms simultaneously analyze several hundreds of genes and can be applied for molecular distinction of different phenotypes within the same disorder’s macro-area. We discuss about the main criticisms regarding molecular bases of cerebral cavernous malformations (CCM) and brain arteriovenous malformations (AVM), highlighting both common pathogenic aspects and genetic differences leading to lesion devel- opment. Many recent studies performed on human CCM and AVM tissues aim to detect genetic markers to better understand molecular bases and pathogenic mechanism, particularly for sporadic cases. Several genes involved in angiogenesis show different expression patterns between CCM and AVM, and these could represent a valid starting point to project a NGS panel to apply for differential cerebrovascular malformation diagnosis. Keywords Cerebral cavernous malformations . Arteriovenous malformations . Genetics . Differential molecular diagnosis Introduction #116860) and arteriovenous malformations (AVM, OMIM #108010) are the most frequent, affecting more Cerebrovascular malformations include a wide spectrum than 3% of the population [1]. Venous developmental of intracranial blood vessel disorders, involving arterial anomalies (VDAs) are rare conditions usually associated wall, capillary bed, and venous and lymphatic systems.
    [Show full text]
  • A Genome-Wide Association Study of Idiopathic Dilated Cardiomyopathy in African Americans
    Journal of Personalized Medicine Article A Genome-Wide Association Study of Idiopathic Dilated Cardiomyopathy in African Americans Huichun Xu 1,* ID , Gerald W. Dorn II 2, Amol Shetty 3, Ankita Parihar 1, Tushar Dave 1, Shawn W. Robinson 4, Stephen S. Gottlieb 4 ID , Mark P. Donahue 5, Gordon F. Tomaselli 6, William E. Kraus 5,7 ID , Braxton D. Mitchell 1,8 and Stephen B. Liggett 9,* 1 Division of Endocrinology, Diabetes and Nutrition, Department of Medicine, University of Maryland School of Medicine, Baltimore, MD 21201, USA; [email protected] (A.P.); [email protected] (T.D.); [email protected] (B.D.M.) 2 Center for Pharmacogenomics, Department of Internal Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA; [email protected] 3 Institute for Genome Sciences, University of Maryland School of Medicine, Baltimore, MD 21201, USA; [email protected] 4 Division of Cardiovascular Medicine, University of Maryland School of Medicine, Baltimore, MD 21201, USA; [email protected] (S.W.R.); [email protected] (S.S.G.) 5 Division of Cardiology, Department of Medicine, Duke University Medical Center, Durham, NC 27708, USA; [email protected] (M.P.D.); [email protected] (W.E.K.) 6 Department of Medicine, Division of Cardiology, Johns Hopkins University, Baltimore, MD 21218, USA; [email protected] 7 Duke Molecular Physiology Institute, Duke University Medical Center, Durham, NC 27701, USA 8 Geriatrics Research and Education Clinical Center, Baltimore Veterans Administration
    [Show full text]
  • Global Analysis of O-Glcnac Glycoproteins in Activated Human T Cells Peder J
    Global Analysis of O-GlcNAc Glycoproteins in Activated Human T Cells Peder J. Lund, Joshua E. Elias and Mark M. Davis This information is current as J Immunol 2016; 197:3086-3098; Prepublished online 21 of October 2, 2021. September 2016; doi: 10.4049/jimmunol.1502031 http://www.jimmunol.org/content/197/8/3086 Downloaded from Supplementary http://www.jimmunol.org/content/suppl/2016/09/20/jimmunol.150203 Material 1.DCSupplemental References This article cites 89 articles, 32 of which you can access for free at: http://www.jimmunol.org/content/197/8/3086.full#ref-list-1 http://www.jimmunol.org/ Why The JI? Submit online. • Rapid Reviews! 30 days* from submission to initial decision • No Triage! Every submission reviewed by practicing scientists • Fast Publication! 4 weeks from acceptance to publication by guest on October 2, 2021 *average Subscription Information about subscribing to The Journal of Immunology is online at: http://jimmunol.org/subscription Permissions Submit copyright permission requests at: http://www.aai.org/About/Publications/JI/copyright.html Author Choice Freely available online through The Journal of Immunology Author Choice option Email Alerts Receive free email-alerts when new articles cite this article. Sign up at: http://jimmunol.org/alerts The Journal of Immunology is published twice each month by The American Association of Immunologists, Inc., 1451 Rockville Pike, Suite 650, Rockville, MD 20852 Copyright © 2016 by The American Association of Immunologists, Inc. All rights reserved. Print ISSN: 0022-1767 Online ISSN: 1550-6606. The Journal of Immunology Global Analysis of O-GlcNAc Glycoproteins in Activated Human T Cells Peder J.
    [Show full text]