Supporting Information

Total Page:16

File Type:pdf, Size:1020Kb

Supporting Information Supporting Information Cusimano et al. 10.1073/pnas.0810243106 SI Text slides (Nalge Nunc International). Two days after plating, cells Immunofluorescence and Antibodies. For immunofluorescence, were transiently transfected by the lipofectamine-Plus method 9 ϫ 104 primary rat myoblasts/cm2 were plated on 2% gelatin- (Invitrogen) and induced to differentiate in alpha-minimum ␣ coated (Sigma) glass coverslips and differentiated as described essential medium ( -MEM; Bio-Whittaker) containing 2 mM ␮ previously (1). Primary skeletal muscle cells were fixed at room L-glutamine (Bio-Whittaker), 100 g/mL streptomycin (Bio- ␮ temperature with 3% paraformaldehyde and 2% sucrose in PBS, Whittaker), 100 U/mL penicillin (Bio-Whittaker), 0,1 M dexa- ␮ permeabilized in Hepes Triton Buffer (20 mM Hepes, pH 7.4, methasone (Sigma), and 50 M hydrocortisone (Sigma), sup- plemented with 10% heat-inactivated FBS (Sigma) and 5% 300 mM sucrose, 50 mM NaCl, 3 mM MgCl2, and 0.5% Triton X-100) and blocked in PBS containing 0.2% BSA and 5% goat heat-inactivated horse serum (Biochrom). NIH 3T3 cells were ϫ 4 2 serum. Cells were incubated overnight with the primary anti- plated at 6 10 /cm per LabTek chamber slide and transiently bodies. Rabbit polyclonal antibodies included anti-ank1.5 (1), transfected the day after. FRAP experiments were performed by anti-RyR1 (2), and anti-triadin (trisk-95), kindly provided by I. using a Zeiss LSM 510 Meta confocal microscope. Cells were Marty (Universite´ Joseph Fouriere, Grenoble, France). Mouse imaged in LabTek chambers in medium containing 140 mM ␣ NaCl, 5 mM KCl, 10 mM glucose, 1 mM MgCl2, 0.1 mM CaCl2, monoclonal antibodies included anti -actinin (A-7811; clone ϫ EA-53; Sigma), anti-RyR (34C; Alexis), and anti-SERCA1 20 mM Hepes, and 0.4 mM EGTA. A 63 1.4 N.A. Plan- (CaF2–5D2; Developmental Studies Hybridoma Bank). Second- Apochromat oil immersion objective (Zeiss) was used with a pinhole aperture of 4.96 Airy units. GFP was excited at 488 nm ary goat anti-rabbit and goat anti-mouse IgG antibodies, con- with an argon laser with low laser power (0.5%) and emitted jugated with CyTM2orCyTM3, were purchased from Jackson fluorescence collected with a long-pass 505 emission filter. After ImmunoResearch Laboratories. Images were obtained with a acquisition of 10 prebleach images, photobleaching of GFP in a Zeiss LSM 510 Meta confocal microscope. circular area 1.08 ␮m in diameter was performed by using the argon laser lines 458, 477, and 488 nm at 50% laser power and GFP Fusion Proteins. Full-length cDNA of human ank1.5 (3) was 50% transmission. Recovery of fluorescence in the bleached inserted into the EcoRI/BamHI sites of the pEGFP-N1 vector ⌬ region was recorded every 50 ms over a period of 1–10 min until (Clontech). The ank1.5 -GFP mutant lacking the binding site fluorescence level reached a plateau. Fluorescence intensities for obscurin has been described previously (3). The construct were acquired for the bleached region (I ), the whole cell pSG5-SERCA2a was kindly provided by E. Carafoli (University frap (Iwhole), and a background region (Ibase). The data were low-pass of Padova, Padova, Italy) and subcloned into the EcoRI site of 3 ϫ 3 filtered with the Zeiss LSM 510 software for image noise the pEGFP-C3 expression vector. Full-length cDNA of murine reduction, and data analysis was performed by using macros InsP3R1 was kindly provided by K. Mikoshiba (University of designed in IgorPro software (WaveMetrics Inc.). The average Tokyo, Tokyo, Japan). The coding sequence of GFP was am- fluorescence intensity within the bleached region was normal- plified and inserted into the Nhe/AflII sites of pcDNA3.1- ized to prebleach intensity for each time point and corrected for ⌬ InsP3R1. The GFP-InsP3R1 14 mutant was prepared as de- loss of fluorescence during acquisition. Fluorescence loss during scribed previously (4). Rabbit RyR1 cDNA was kindly provided acquisition was basically no more than 10%. The normalized by P. D. Allen (Brigham and Women’s Hospital, Boston, MA) data were fitted by the exponential equation: I frap-est(t) ϭ A and cloned into the pEGFP-C3 vector. Full-length triadin (95- Ϫ␶ t (1-e 1 ) (1), where A represents the mobile fraction. Fitting kDa isoform) and junctin were amplified by PCR from a human quality was evaluated by the probability Q value and accepted cDNA library and cloned into the pEGFP-N3 vector. Human when Q Ͼ 0.01. The diffusion constant was calculated by using full-length JP1 was amplified by PCR and cloned in pEGFP-C2. the Soumpasis diffusion fitting (7). Statistical analyses were To obtain the triadin-junctin-GFP chimera, the fragment from performed by using Instat Software (GraphPad). Data were amino acids 1 to 73 of triadin and the fragment from amino acids initially evaluated for normal distribution with the Kolmogorov– 51 to 625 of junctin were amplified by PCR. The fragments Smirnov test. Not normally distributed data were compared by obtained were joined by PCR and cloned into the EcoRI/BamHI Mann–Whitney test. Comparisons between 2 groups with nor- sites of pEGFP-N3. The chimera junctin-triadin-GFP was ob- mally distributed variables were analyzed by Student’s unpaired tained by amplification of the fragments from amino acids 1 to t-test. Comparisons between 3 groups were analyzed by 1-way 50 of junctin and from amino acids 73 to 728 of triadin. The ANOVA followed by Bonferroni’s posthoc. Data are presented fragments were joined by PCR and cloned into the EcoRI/ as mean Ϯ SD. BamHI sites of pEGFP-N3. The constructs pEGFPC-␣1S (5) and pcDNA-␤1A (6), coding for subunits of rabbit DHPR, were Qualitative iFRAP Experiments. For iFRAP experiments, a 63 ϫ 1.4 kindly provided by M. Grabner and B. E. Flucher (Innsbruck N.A. Plan-Apochromat oil immersion objective and a pinhole Medical University, Innsbruck, Austria). To facilitate expression aperture of 4.96 Airy units were used. Considering that the and surface localization of heterologous DHPR GFP-␣1S, myo- myotubes were too long to be entirely visualized, a region of a blasts were cotransfected with the untagged ␤1A subunit of transfected myotube was selected for the bleaching. After ac- DHPR. All constructs were sequence-verified. quisition of 3 prebleach images, fluorescence was bleached by using full laser power, leaving unbleached a small area in the FRAP Experiments and Statistical Analysis. For FRAP analyses, 6 ϫ center of the myotube region. Postbleach images were acquired 104 cells/cm2 were plated in 2% gelatin-coated LabTek chamber- for up to 20 min at 300-ms intervals. 1. Armani A, et al. (2006) Molecular interactions with obscurin are involved in the 2. Giannini G, Conti A, Mammarella S, Scrobogna M, Sorrentino V (1995) The ryanodine localization of muscle-specific small ankyrin1 isoforms to subcompartments of the receptor/calcium channel genes are widely and differentially expressed in murine brain sarcoplasmic reticulum. Exp Cell Res 312:3546–3558. and peripheral tissues. J Cell Biol 128:893–904. Cusimano et al. www.pnas.org/cgi/content/short/0810243106 1of9 3. Bagnato P, Barone V, Giacomello E, Rossi D, Sorrentino V (2003) Binding of an 7. Soumpasis DM (1983) Theoretical analysis of fluorescence photobleaching recovery Ankyrin-1 isoform to Obscurin suggests a molecular link between the sarcoplasmic experiments. Biophys J 41:95–97. reticulum and myofibrils in striated muscles. J Cell Biol 160:245–253. 8. Phimister AJ, et al. (2007) Conformation-dependent stability of junctophilin 1 (JP1) and 4. Zhang S, et al. (2003) Protein 4.1N is required for translocation of inositol 1,4,5- ryanodine receptor type 1 (RyR1) channel complex is mediated by their hyper-reactive triphosphate receptor type 1 to the basolateral membrane domain in polarized thiols. J Biol Chem 282:8667–8677. Madin-Darby canine kidney cells. J Biol Chem 278:4048–4056. 9. Kontrogianni-Konstantopoulos A, et al. (2006) Obscurin modulates the assembly and 5. Grabner M, Dirkens RT, Beam KG (1998) Tagging with green fluorescent protein reveals organization of sarcomeres and the sarcoplasmic reticulum. FASEB J 20:2102–2111. a distinct subcellular distribution of L-type and non-L-type Ca2ϩ channels expressed in 10. Fukatsu K, et al. (2004) Lateral diffusion of inositol 1,4,5-trisphosphate receptor type dysgenic myotubes. Proc Natl Acad Sci USA 95:1903–1908. 1 is regulated by actin filaments and 4.1N in neuronal dendrites. J Biol Chem 6. Schredelseker J, et al. (2005) The beta 1a subunit is essential for the assembly of 279:48976–48982. dihydropyridine-receptor arrays in skeletal muscle. Proc Natl Acad Sci USA 102:17219– 11. Fukatsu K, Bannai H, Inoue T, Mikoshiba K (2006) 4.1N binding of inositol 1,4,5- 17224. triphosphate receptor typ1 1. Biochem Biophys Res Commun 342:573–576. Cusimano et al. www.pnas.org/cgi/content/short/0810243106 2of9 Fig. S1. Localization of ank1.5, SERCA and RyR in differentiating myotubes. In a fraction of 4-day differentiating myotubes, in addition to the ank1.5-signal at the level of the M-band, a less intense signal could be observed at the level of the Z-disk (A, C). Staining with ␣-actinin is shown to identify the Z-disk, (B, C). In a fraction of 6-day differentiating myotubes, ank1.5 could still be observed only at the level of the M-band (D, F), as shown by the alternating pattern with ␣-actinin (E, F). In 6-day differentiated myotubes, RyR signal was organized at triads (G, I) only in cells where SERCA staining (H, I) was organized at the Z-disk. (Scale bar, 5␮m.) Cusimano et al.
Recommended publications
  • Human Periprostatic Adipose Tissue: Secretome from Patients With
    CANCER GENOMICS & PROTEOMICS 16 : 29-58 (2019) doi:10.21873/cgp.20110 Human Periprostatic Adipose Tissue: Secretome from Patients With Prostate Cancer or Benign Prostate Hyperplasia PAULA ALEJANDRA SACCA 1, OSVALDO NÉSTOR MAZZA 2, CARLOS SCORTICATI 2, GONZALO VITAGLIANO 3, GABRIEL CASAS 4 and JUAN CARLOS CALVO 1,5 1Institute of Biology and Experimental Medicine (IBYME), CONICET, Buenos Aires, Argentina; 2Department of Urology, School of Medicine, University of Buenos Aires, Clínical Hospital “José de San Martín”, Buenos Aires, Argentina; 3Department of Urology, Deutsches Hospital, Buenos Aires, Argentina; 4Department of Pathology, Deutsches Hospital, Buenos Aires, Argentina; 5Department of Biological Chemistry, School of Exact and Natural Sciences, University of Buenos Aires, Buenos Aires, Argentina Abstract. Background/Aim: Periprostatic adipose tissue Prostate cancer (PCa) is the second most common cancer in (PPAT) directs tumour behaviour. Microenvironment secretome men worldwide. While most men have indolent disease, provides information related to its biology. This study was which can be treated properly, the problem consists in performed to identify secreted proteins by PPAT, from both reliably distinguishing between indolent and aggressive prostate cancer and benign prostate hyperplasia (BPH) disease. Evidence shows that the microenvironment affects patients. Patients and Methods: Liquid chromatography-mass tumour behavior. spectrometry-based proteomic analysis was performed in Adipose tissue microenvironment is now known to direct PPAT-conditioned media (CM) from patients with prostate tumour growth, invasion and metastases (1, 2). Adipose cancer (CMs-T) (stage T3: CM-T3, stage T2: CM-T2) or tissue is adjacent to the prostate gland and the site of benign disease (CM-BPH). Results: The highest number and invasion of PCa.
    [Show full text]
  • Heterotrimeric Go Protein Links Wnt-Frizzled Signaling with Ankyrins to Regulate the Neuronal Microtubule Cytoskeleton Anne-Marie Lüchtenborg1,2, Gonzalo P
    © 2014. Published by The Company of Biologists Ltd | Development (2014) 141, 3399-3409 doi:10.1242/dev.106773 RESEARCH ARTICLE Heterotrimeric Go protein links Wnt-Frizzled signaling with ankyrins to regulate the neuronal microtubule cytoskeleton Anne-Marie Lüchtenborg1,2, Gonzalo P. Solis1, Diane Egger-Adam2, Alexey Koval1, Chen Lin1,2, Maxime G. Blanchard1, Stephan Kellenberger1 and Vladimir L. Katanaev1,2,* ABSTRACT The evolutionarily conserved Wg pathway is important for Drosophila neuromuscular junctions (NMJs) represent a powerful numerous developmental programs and cellular processes (Logan model system with which to study glutamatergic synapse formation and Nusse, 2004). In the nervous system of Drosophila,Wg and remodeling. Several proteins have been implicated in these signaling is involved in the formation of neuromuscular junctions processes, including components of canonical Wingless (Drosophila (NMJs) (Packard et al., 2002; Miech et al., 2008). Being a Wnt1) signaling and the giant isoforms of the membrane-cytoskeleton glutamatergic synapse, the Drosophila NMJ provides a useful linker Ankyrin 2, but possible interconnections and cooperation experimental model with which to study mammalian central between these proteins were unknown. Here, we demonstrate that nervous system synapses, their formation and remodeling (Collins the heterotrimeric G protein Go functions as a transducer of Wingless- and DiAntonio, 2007). The Drosophila NMJ is a beads-on-a-string- Frizzled 2 signaling in the synapse. We identify Ankyrin 2 as a target like structure that is formed at the axon terminus and is composed of – – of Go signaling required for NMJ formation. Moreover, the Go-ankyrin distinct circular structures the synaptic boutons which contain interaction is conserved in the mammalian neurite outgrowth pathway.
    [Show full text]
  • A De Novo ANK1 Mutation Associated to Hereditary Spherocytosis: a Case Report
    Huang et al. BMC Pediatrics (2019) 19:62 https://doi.org/10.1186/s12887-019-1436-4 CASEREPORT Open Access A de novo ANK1 mutation associated to hereditary spherocytosis: a case report Ti-Long Huang1, Bao-Hua Sang1, Qing-Ling Lei1, Chun-Yan Song1, Yun-Bi Lin1,YuLv1, Chun-Hui Yang1,NaLi1, Yue-Huang Yang1, Xian-Wen Zhang2* and Xin Tian1* Abstract Background: Hereditary spherocytosis (HS) is a type of hemolytic anemia caused by abnormal red cell membrane skeletal proteins with few unique clinical manifestations in the neonate and infant. An ANK1 gene mutation is the most common cause of HS. Case presentation: The patient was a 11-month-old boy who suffered from anemia and needed a regular transfusion therapy at an interval of 2–3 months. Hematological investigations showed moderate anemia (Hb80 g/L). Red cells displayed microcytosis (MCV76.4 fl, MCH25.6 pg, MCHC335 g/L). The reticulocytes were elevated (4.8%) and the spherocytes were increased (10%). Direct antiglobulin test was negative. Biochemical test indicated a slight elevation of bilirubin, mainly indirect reacting (TBIL32.5 μmol/L, IBIL24 μmol/L). The neonatal HS ratio is 4.38, obviously up the threshold. Meanwhile, adenovoANK1 mutation (exon 25:c.2693dupC:p.A899Sfs*11) was identified by next-generation sequencing (NGS). Thus, hereditary spherocytosis was finally diagnosed. Conclusions: Gene detection should be considered in some hemolytic anemia which is difficult to diagnose by routine means. We identified a novel de novo ANK1 heterozygous frameshift mutation in a Yi nationality patient while neither of his parents carried this mutation. Keywords: Hereditary spherocytosis- ANK1- frameshift mutation Background (erythrocyte membrane protein band 4.2) are associated Hereditary spherocytosis (HS) results from defects in with HS [5].
    [Show full text]
  • Review Article Mouse Homologues of Human Hereditary Disease
    I Med Genet 1994;31:1-19 I Review article J Med Genet: first published as 10.1136/jmg.31.1.1 on 1 January 1994. Downloaded from Mouse homologues of human hereditary disease A G Searle, J H Edwards, J G Hall Abstract involve homologous loci. In this respect our Details are given of 214 loci known to be genetic knowledge of the laboratory mouse associated with human hereditary dis- outstrips that for all other non-human mam- ease, which have been mapped on both mals. The 829 loci recently assigned to both human and mouse chromosomes. Forty human and mouse chromosomes3 has now two of these have pathological variants in risen to 900, well above comparable figures for both species; in general the mouse vari- other laboratory or farm animals. In a previous ants are similar in their effects to the publication,4 102 loci were listed which were corresponding human ones, but excep- associated with specific human disease, had tions include the Dmd/DMD and Hprt/ mouse homologues, and had been located in HPRT mutations which cause little, if both species. The number has now more than any, harm in mice. Possible reasons for doubled (table 1A). Of particular interest are phenotypic differences are discussed. In those which have pathological variants in both most pathological variants the gene pro- the mouse and humans: these are listed in table duct seems to be absent or greatly 2. Many other pathological mutations have reduced in both species. The extensive been detected and located in the mouse; about data on conserved segments between half these appear to lie in conserved chromo- human and mouse chromosomes are somal segments.
    [Show full text]
  • A Knowledge-Guided Mechanistic Model of Synthetic Lethality in The
    bioRxiv preprint doi: https://doi.org/10.1101/2021.06.22.449530; this version posted June 23, 2021. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY-NC-ND 4.0 International license. A Knowledge-guided Mechanistic Model of Synthetic Lethality in the HCT116 Vorinostat-resistant Colon Cancer Xenograft Model Cell-line Paul Aiyetan June 22, 2021 Abstract With an overall lifetime risk of about 4.3% and 4.0%, in men and women respectively, colorectal cancer remains the third leading cause of cancer-related deaths in the United States. In persons aged 55 and below, its rate increased at 1% per year in the years 2008 to 2017 despite the steady decline associated with improved screening, early diagnosis and treatment in the general population. Besides standardized therapeutic regimen, many trials continue to evaluate the potential benefits of vorinostat, mostly in combination with other anti-neoplastic agents for its treatment. Vorinostat, an FDA approved anti-cancer drug known as suberoylanilide hydroxamic acid (SAHA), an histone deacylase (HDAC) inhibitor, through many mechanisms, causes cancer cell arrest and death. However, like many other anti-neoplastic agents, resistance and or failures have been observed. In the HCT116 colon cancer cell line xenograft model, exploiting potential lethal molecular interactions by additional gene knockouts restored vorinotat sensitivity. This phenomenon, known as synthetic lethality, offers a promise to selectively target cancer cells. Although without clearly delineated understanding of underlying molecular processes, it has been demonstrated as an effective cancer-killing mechanism.
    [Show full text]
  • Genetic Cardiomyopathies: the Lesson Learned from Hipscs
    Journal of Clinical Medicine Review Genetic Cardiomyopathies: The Lesson Learned from hiPSCs Ilaria My 1,2 and Elisa Di Pasquale 2,3,* 1 Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, 20090 Milan, Italy; [email protected] 2 Humanitas Clinical and Research Center—IRCCS, Rozzano, 20089 Milan, Italy 3 Institute of Genetic and Biomedical Research (IRGB)—UOS of Milan, National Research Council (CNR), 20138 Milan, Italy * Correspondence: [email protected] Abstract: Genetic cardiomyopathies represent a wide spectrum of inherited diseases and constitute an important cause of morbidity and mortality among young people, which can manifest with heart failure, arrhythmias, and/or sudden cardiac death. Multiple underlying genetic variants and molecular pathways have been discovered in recent years; however, assessing the pathogenicity of new variants often needs in-depth characterization in order to ascertain a causal role in the disease. The application of human induced pluripotent stem cells has greatly helped to advance our knowledge in this field and enabled to obtain numerous in vitro patient-specific cellular models useful to study the underlying molecular mechanisms and test new therapeutic strategies. A milestone in the research of genetically determined heart disease was the introduction of genomic technologies that provided unparalleled opportunities to explore the genetic architecture of cardiomyopathies, thanks to the generation of isogenic pairs. The aim of this review is to provide an overview of the main research that helped elucidate the pathophysiology of the most common genetic cardiomyopathies: hypertrophic, dilated, arrhythmogenic, and left ventricular noncompaction cardiomyopathies. A special focus is provided on the application of gene-editing techniques in understanding key disease characteristics and on the therapeutic approaches that have been tested.
    [Show full text]
  • Cadherins As Targets for Genetic Diseases
    Downloaded from http://cshperspectives.cshlp.org/ on September 28, 2021 - Published by Cold Spring Harbor Laboratory Press Cadherins as Targets for Genetic Diseases Aziz El-Amraoui1,2,3 and Christine Petit1,2,3,4 1Institut Pasteur, Unite´ de Ge´ne´tique et Physiologie de l’Audition, 25 rue du Dr Roux, 75015 Paris, France 2INSERM UMRS587, 75015 Paris, France 3UPMC, F75015 Paris, France 4Colle`ge de France, 75005 Paris, France Correspondence: [email protected], [email protected] The 6-billion human population provides a vast reservoir of mutations, which, in addition to the opportunity of detecting very subtle defects, including specific cognitive dysfunctions as well as late appearing disorders, offers a unique background in which to investigate the roles of cell–cell adhesion proteins. Here we focus on inherited human disorders involving members of the cadherin superfamily. Most of the advances concern monogenic disorders. Yet,with the development of single nucleotide polymorphism (SNP) association studies, cad- herin genes are emerging as susceptibility genes in multifactorial disorders. Various skin and heart disorders revealed the critical role played by desmosomal cadherins in epidermis, hairs, and myocardium, which experience high mechanical stress. Of particular interest in that respect is the study of Usher syndrome type 1 (USH1), a hereditary syndromic form of deafness. Studies of USH1 brought to light the crucial role of transient fibrous links formed by cadherin 23 and protocadherin 15 in the cohesion of the developing hair bundle, the mechanoreceptive structure of the auditory sensory cells, as well as the involvement of these cadherins in the formation of the tip-link, a key component of the mechano-electrical transduction machinery.
    [Show full text]
  • Methylomic Profiling Implicates Cortical Deregulation of ANK1 in Alzheimer’S Disease
    ART ic LE S Methylomic profiling implicates cortical deregulation of ANK1 in Alzheimer’s disease Katie Lunnon1, Rebecca Smith2, Eilis Hannon1, Philip L De Jager3–5, Gyan Srivastava3,5, Manuela Volta2, Claire Troakes2, Safa Al-Sarraj2, Joe Burrage1, Ruby Macdonald1, Daniel Condliffe2, Lorna W Harries1, Pavel Katsel6, Vahram Haroutunian6–8, Zachary Kaminsky9,10, Catharine Joachim11, John Powell2, Simon Lovestone2,12, David A Bennett13, Leonard C Schalkwyk2,14 & Jonathan Mill1,2,14 Alzheimer’s disease (AD) is a chronic neurodegenerative disorder that is characterized by progressive neuropathology and cognitive decline. We performed a cross-tissue analysis of methylomic variation in AD using samples from four independent human post-mortem brain cohorts. We identified a differentially methylated region in the ankyrin 1 (ANK1) gene that was associated with neuropathology in the entorhinal cortex, a primary site of AD manifestation. This region was confirmed as being substantially hypermethylated in two other cortical regions (superior temporal gyrus and prefrontal cortex), but not in the cerebellum, a region largely protected from neurodegeneration in AD, or whole blood obtained pre-mortem from the same individuals. Neuropathology-associated ANK1 hypermethylation was subsequently confirmed in cortical samples from three independent brain cohorts. This study represents, to the best of our knowledge, the first epigenome-wide association study of AD employing a sequential replication design across multiple tissues and highlights the power of this approach for identifying methylomic variation associated with complex disease. AD contributes substantially to the global burden of disease, affecting is a relatively new endeavor, however, and there are important in excess of 26 million people worldwide1,2.
    [Show full text]
  • Human CD Marker Chart Reviewed by HLDA1 Bdbiosciences.Com/Cdmarkers
    BD Biosciences Human CD Marker Chart Reviewed by HLDA1 bdbiosciences.com/cdmarkers 23-12399-01 CD Alternative Name Ligands & Associated Molecules T Cell B Cell Dendritic Cell NK Cell Stem Cell/Precursor Macrophage/Monocyte Granulocyte Platelet Erythrocyte Endothelial Cell Epithelial Cell CD Alternative Name Ligands & Associated Molecules T Cell B Cell Dendritic Cell NK Cell Stem Cell/Precursor Macrophage/Monocyte Granulocyte Platelet Erythrocyte Endothelial Cell Epithelial Cell CD Alternative Name Ligands & Associated Molecules T Cell B Cell Dendritic Cell NK Cell Stem Cell/Precursor Macrophage/Monocyte Granulocyte Platelet Erythrocyte Endothelial Cell Epithelial Cell CD1a R4, T6, Leu6, HTA1 b-2-Microglobulin, CD74 + + + – + – – – CD93 C1QR1,C1qRP, MXRA4, C1qR(P), Dj737e23.1, GR11 – – – – – + + – – + – CD220 Insulin receptor (INSR), IR Insulin, IGF-2 + + + + + + + + + Insulin-like growth factor 1 receptor (IGF1R), IGF-1R, type I IGF receptor (IGF-IR), CD1b R1, T6m Leu6 b-2-Microglobulin + + + – + – – – CD94 KLRD1, Kp43 HLA class I, NKG2-A, p39 + – + – – – – – – CD221 Insulin-like growth factor 1 (IGF-I), IGF-II, Insulin JTK13 + + + + + + + + + CD1c M241, R7, T6, Leu6, BDCA1 b-2-Microglobulin + + + – + – – – CD178, FASLG, APO-1, FAS, TNFRSF6, CD95L, APT1LG1, APT1, FAS1, FASTM, CD95 CD178 (Fas ligand) + + + + + – – IGF-II, TGF-b latency-associated peptide (LAP), Proliferin, Prorenin, Plasminogen, ALPS1A, TNFSF6, FASL Cation-independent mannose-6-phosphate receptor (M6P-R, CIM6PR, CIMPR, CI- CD1d R3G1, R3 b-2-Microglobulin, MHC II CD222 Leukemia
    [Show full text]
  • Alzheimer's Disease Risk Gene BIN1 Induces Tau-Dependent Network
    RESEARCH ARTICLE Alzheimer’s disease risk gene BIN1 induces Tau-dependent network hyperexcitability Yuliya Voskobiynyk1†, Jonathan R Roth1†, J Nicholas Cochran1, Travis Rush1, Nancy VN Carullo2, Jacob S Mesina1, Mohammad Waqas1, Rachael M Vollmer1, Jeremy J Day2, Lori L McMahon3, Erik D Roberson1* 1Center for Neurodegeneration and Experimental Therapeutics, Alzheimer’s Disease Center, and Evelyn F. McKnight Brain Institute, Departments of Neurology and Neurobiology, University of Alabama at Birmingham, Birmingham, United States; 2Department of Neurobiology, University of Alabama at Birmingham, Birmingham, United States; 3Department of Cell, Developmental and Integrative Biology, University of Alabama at Birmingham, Birmingham, United States Abstract Genome-wide association studies identified the BIN1 locus as a leading modulator of genetic risk in Alzheimer’s disease (AD). One limitation in understanding BIN1’s contribution to AD is its unknown function in the brain. AD-associated BIN1 variants are generally noncoding and likely change expression. Here, we determined the effects of increasing expression of the major neuronal isoform of human BIN1 in cultured rat hippocampal neurons. Higher BIN1 induced network hyperexcitability on multielectrode arrays, increased frequency of synaptic transmission, and elevated calcium transients, indicating that increasing BIN1 drives greater neuronal activity. In exploring the mechanism of these effects on neuronal physiology, we found that BIN1 interacted with L-type voltage-gated calcium channels (LVGCCs) and that BIN1–LVGCC interactions were modulated by Tau in rat hippocampal neurons and mouse brain. Finally, Tau reduction prevented BIN1-induced network hyperexcitability. These data shed light on BIN1’s neuronal function and *For correspondence: suggest that it may contribute to Tau-dependent hyperexcitability in AD.
    [Show full text]
  • PDF Full Text
    From www.bloodjournal.org by guest on October 10, 2018. For personal use only. Regular Article LYMPHOID NEOPLASIA North American ATLL has a distinct mutational and transcriptional profile and responds to epigenetic therapies Urvi A. Shah,1-3 Elaine Y. Chung,2 Orsi Giricz,3 Kith Pradhan,3 Keisuke Kataoka,4 Shanisha Gordon-Mitchell,3 Tushar D. Bhagat,3 Yun Mai,2 Yongqiang Wei,2,5 Elise Ishida,2 Gaurav S. Choudhary,3 Ancy Joseph,6 Ronald Rice,7 Nadege Gitego,3 Crystall Parrish,3 Matthias Bartenstein,3 Swati Goel,1 Ioannis Mantzaris,1 Aditi Shastri,1,3 Olga Derman,1 Adam Binder,1 Kira Gritsman,1,2 Noah Kornblum,1 Ira Braunschweig,1 Chirag Bhagat,8 Jeff Hall,8 Armin Graber,8 Lee Ratner,6 Yanhua Wang,9 Seishi Ogawa,4 Amit Verma,1,3 B. Hilda Ye,2 and Murali Janakiram1 1Department of Oncology, Montefiore Medical Center, Bronx, NY; 2Department of Cell Biology and 3Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, Bronx, NY; 4Department of Pathology and Tumor Biology, Graduate School of Medicine, Kyoto University, Kyoto, Japan; 5Department of Hematology, Nanfang Hospital, Southern Medical University, Guangzhou, China; 6Division of Oncology, Washington University School of Medicine, St Louis, MO; 7Department of Pathology, Phelps Hospital, Northwell Health, Sleepy Hollow, NY; 8Genoptix, Carlsbad, CA; and 9Department of Pathology, Montefiore Medical Center, Bronx, NY KEY POINTS Adult T-cell leukemia lymphoma (ATLL) is a rare T cell neoplasm that is endemic in Japanese, Caribbean, and Latin American populations. Most North American ATLL patients are of l North American ATLL has a distinct genomic Caribbean descent and are characterized by high rates of chemo-refractory disease and landscape with a worse prognosis compared with Japanese ATLL.
    [Show full text]
  • The Use of Genetic Analyses and Functional Assays for the Interpretation of Rare Variants in Pediatric Heart Disease
    The use of genetic analyses and functional assays for the interpretation of rare variants in pediatric heart disease A dissertation submitted to the Division of Graduate Studies and Research, University of Cincinnati in partial fulfillment of the requirements for the degree of Doctor of Philosophy in Molecular Genetics by Jeffrey A. Schubert Bachelor of Science, Mount St. Joseph University, 2012 Committee Chair: Stephanie M. Ware, M.D., Ph.D. Edmund Choi, Ph.D. Benjamin Landis, M.D. Anil Menon, Ph.D. David Wieczorek, Ph.D. Molecular Genetics, Biochemistry, and Microbiology Graduate Program College of Medicine, University of Cincinnati Cincinnati, Ohio, USA, 2018 ABSTRACT The use of next generation technologies such as whole exome sequencing (WES) has paved the way for discovering novel causes of Mendelian diseases. This has been demonstrated in pediatric heart diseases, including cardiomyopathy (CM) and familial thoracic aortic aneurysm (TAA). Each of these conditions carries a high risk of a serious cardiac event, including sudden heart failure or aortic rupture, which are often fatal. Patients with either disease can be asymptomatic before presenting with these events, which necessitates early diagnosis. Though there are many known genetic causes of disease for both conditions, there is still room for discovery of novel pathogenic genes and variants, as many patients have an undefined genetic diagnosis. WES covers the protein-coding portion of the genome, which yields a massive amount of data, though it comprises only 1% of the genome. Sorting and filtering sequencing information to identify (sometimes) a single base pair change responsible for the patient phenotype is challenging. Further, interpreting identified candidate variants must be done according to strict standards, which makes it difficult to definitively say whether a coding change is pathogenic or benign.
    [Show full text]