Peptides Derived from Insulin Granule Proteins Are Targeted by CD8+ T Cells

Total Page:16

File Type:pdf, Size:1020Kb

Peptides Derived from Insulin Granule Proteins Are Targeted by CD8+ T Cells Page 1 of 44 Diabetes Peptides derived from insulin granule proteins are targeted by CD8+ T cells across MHC Class I restrictions in humans and NOD mice. Marie Eliane Azoury1*, Mahmoud Tarayrah1*, Georgia Afonso1, Aurore Pais1, Maikel L. Colli2, Claire Maillard1, Cassandra Lavaud1, Laure Alexandre-Heymann1,3, Sergio Gonzalez- Duque1,4, Yann Verdier4, Joelle Vinh4, Sheena Pinto5, Soren Buus6, Danièle Dubois- Laforgue1,3, Etienne Larger1,3, Jean-Paul Beressi7, Graziella Bruno8, Decio L. Eizirik2, Sylvaine You1, Roberto Mallone1,3. 1Université de Paris, Institut Cochin, CNRS, INSERM, 75014 Paris, France. 2Université Libre de Bruxelles, Center for Diabetes Research and Welbio, Medical Faculty, Université Libre de Bruxelles, 1070 Brussels, Belgium. 3Assistance Publique Hôpitaux de Paris, Hôpitaux Universitaires de Paris Centre-Université de Paris, Cochin Hospital, Service de Diabétologie et Immunologie Clinique, 75014 Paris, France. 4ESPCI Paris, PSL University, Spectrométrie de Masse Biologique et Protéomique, CNRS USR3149, 75005 Paris, France. 5DKFZ, Division of Developmental Immunology, 69120 Heidelberg, Germany. 6University of Copenhagen, Department of Immunology and Microbiology, Laboratory of Experimental Immunology, DK-2200 N Copenhagen, Denmark. 7Centre Hospitalier de Versailles André Mignot, Service de Diabétologie, 78150 Le Chesnay, France. 8University of Turin, Department of Medical Sciences, 10126 Turin, Italy. *These authors contributed equally to this work. Running title: T-cell antigens from insulin granule proteins. Corresponding author: Roberto Mallone, [email protected] Manuscript length: 4,480 words, 1 table, 5 figures. Online Appendix: 3 tables, 3 figures. Tweet (204 characters): Proteins of the insulin granules in beta cells are key autoimmune targets in #type 1 diabetes. The list of antigens available to understand disease mechanisms and monitor its progression is growing longer! @InstitutCochin, @Inserm, @Innodiagroup 1 Diabetes Publish Ahead of Print, published online September 14, 2020 Diabetes Page 2 of 44 ABSTRACT The antigenic peptides processed by β cells and presented through surface HLA Class I molecules are poorly characterized. Each HLA variant, e.g. the most common HLA-A2 and HLA-A3, carries some peptide-binding specificity. Hence, features that, despite these specificities, remain shared across variants may reveal factors favoring β-cell immunogenicity. Building on our previous description of the HLA-A2/A3 peptidome of β cells, we analyzed the HLA-A3-restricted peptides targeted by circulating CD8+ T cells. Several peptides were recognized by CD8+ T cells within a narrow frequency (1-50/106), which was similar in donors with and without type 1 diabetes and harbored variable effector/memory fractions. These epitopes could be classified as conventional peptides or neo-epitopes, generated either via peptide cis-splicing or mRNA splicing, e.g. secretogranin-5 (SCG5)-009. As reported for HLA-A2-restricted peptides, several epitopes originated from β- cell granule proteins, e.g. SCG3, SCG5 and urocortin-3. Similarly, H-2Kd-restricted CD8+ T cells recognizing the murine orthologues of SCG5, urocortin-3, and proconvertase-2 infiltrated the islets of NOD mice and transferred diabetes into NOD/scid recipients. The finding of granule proteins targeted in both humans and NOD mice supports their disease relevance and identifies the insulin granule as a rich source of epitopes, possibly reflecting its impaired processing in type 1 diabetes. 2 Page 3 of 44 Diabetes INTRODUCTION Autoimmune CD8+ T cells are held as the final effectors of β-cell destruction in type 1 diabetes. Indirect support for this tenet comes from the observations that the pancreatic immune infiltrates of type 1 diabetic patients are dominated by CD8+ T cells (1); that at least some of these CD8+ T cells recognize islet epitopes (1-3); and that islet-reactive cytotoxic CD8+ T-cell clones can lyse β cells in vitro (4-7). However, clones with similar cytotoxic activity can be derived from healthy donors (2). More generally, recent reports from our laboratory (2, 3) and others (8) documented that the frequency of islet-reactive CD8+ T cells in peripheral blood is similar between donors with and without type 1 diabetes, while these T cells are enriched in the pancreas of diseased individuals (2, 3). We therefore proposed a novel paradigm of “benign” islet autoimmunity, which is present in all individuals and imprinted in the thymus, due to a marginal impact on clonal deletion of the antigens presented by thymic medullary epithelial cells (mTECs) (2, 9, 10). The progression of this benign autoimmune state toward type 1 diabetes may rely on different, non-exclusive mechanisms. First, regulation may be at play, either T-cell-extrinsic (e.g. suppression by regulatory T cells, Tregs) or intrinsic (e.g. anergy, exhaustion). Indeed, conventional T cells from T1D patients are more resistant to Treg suppression (11, 12) and exhaustion (8, 13), and assays relying on T-cell activation consistently detected higher frequencies of some islet-reactive fractions in T1D patients (2, 4, 14-17). Second, a higher vulnerability of target β cells may favor type 1 diabetes progression (18). The lines of evidence for this ‘β-cell-centric’ hypothesis include observations that: i) some T1D susceptibility gene variants modulate islet inflammation (19-21), which makes β cells more ‘visible’ to autoimmune CD8+ T cells by upregulating HLA Class I (HLA-I) expression and peptide-HLA-I (pHLA-I) presentation (3); ii) the benefit of effective immunotherapies is limited in time, which may hint at β-cell-intrinsic components not impacted by treatment (22); iii) contrary to what found in the blood, islet-reactive CD8+ T cells are enriched in the type 1 3 Diabetes Page 4 of 44 diabetic pancreas (2, 3), pointing to local factors that promote their disease-specific homing to the target organ; and iv) diseased β cells display endoplasmic reticulum (ER) stress (23) and impaired proinsulin processing (24, 25), which may promote their immunogenicity by favoring HLA-I presentation of potentially modified proinsulin peptides. Overall, a unifying picture is emerging that prompts to consider type 1 diabetes as a disease of both the immune system and the β cell (18, 26). The identification of the peptides naturally processed by β cells that trigger CD8+ T-cell recognition through presentation by surface HLA-I molecules may provide novel and relevant information on how β cells exert an active role in disease pathogenesis. We recently provided a first description of the HLA-I peptidome of human β cells under basal and inflammatory conditions, and detailed novel antigenic peptides recognized by circulating CD8+ T cells in the frame of the most prevalent HLA- A*02:01 (HLA-A2) variant (3). As observed for known islet epitopes (2), the frequency of these CD8+ T cells was similar in the blood, but higher in the pancreas, of type 1 diabetic patients compared with healthy donors (3). Several other HLA-I-eluted peptides were predicted to bind the other common HLA-A*03:01 variant (HLA-A3 from hereon) (3), which presents peptides carrying different amino acid (aa) motifs compared to HLA-A2. Whether CD8+ T cells recognize these peptides was not assessed. We here studied this recognition in order to define whether HLA-A2 and HLA-A3, despite their different peptide binding preferences, present peptides with any shared feature. We report that peptides derived from insulin granule proteins are common for both variants, and are also targeted by CD8+ T cells that infiltrate the islets of prediabetic non-obese diabetic (NOD) mice and transfer disease into NOD/scid recipients. 4 Page 5 of 44 Diabetes RESEARCH DESIGN AND METHODS HLA-A3-restricted candidate epitopes identified by HLA-I peptidomics and in-silico mining of mRNA splice variants The first set of candidate HLA-A3-restricted epitopes was previously identified by HLA-I peptidomics (3) (Table 1, top). Potential HLA-A3 binding was assigned based on predicted affinity (NetMHC 4.0) and stability (NetMHCstab 1.0) scores. Candidates derived from mRNA splice variants were identified by a parallel in-silico mining of previous RNA-seq datasets from primary human islets exposed or not to interleukin (IL)-1 and interferon (IFN)- and from human mTECs, as described in (3) and summarized in Figure S1. Relevant mRNA splice variants were first selected based on a median reads per kilobase per million mapped reads (RPKM) >5 in islets, based on the median RPKM of known islet antigens (3, 27). Second, mRNA isoforms poorly expressed in mTECs, which might favor T-cell escape from clonal deletion, were selected based on an RPKM <0.1 in mTECs or with a fold decrease >100 versus islets (3). Third, only mRNA isoforms with >10-fold higher expression in islets versus other tissues were retained (3). We then analyzed the predicted aa neo- sequences encoded by these mRNA variants, taking the longest and/or most prevalent islet mRNA isoform as reference. This neo-sequence filter yielded 88/166 mRNA variants (53%) and 340 peptide neo-sequences identified as a potential source of neo-epitopes. Thirty-seven peptides predicted to bind HLA-A3 were thus identified. We further restricted this list by focusing on 9- to 10-aa-long peptides (29/37, 78%) carrying a neo-sequence stretch ≥3 aa, with 24/29 (83%) candidates finally retained (Table 1, bottom). Peptides and HLA-A3 binding assays Peptides (>90% pure, Synpeptides) were tested for binding to HLA-A3 by flow cytometry using biotin-tagged HLA-A3 monomers (immunAware), per the manufacturer’s protocol. Briefly, biotinylated monomers (final concentration 1.2 nM) were folded as described (3) and 5 Diabetes Page 6 of 44 captured on 6-8 µm streptavidin-coated beads (Spherotech). Beads were subsequently incubated with an anti-β2-microglobulin BBM.1 monoclonal antibody (mAb; Santa Cruz), followed by an AlexaFluor (AF)488-labeled goat polyclonal anti-mouse IgG (RRID:AB_2728715). The HLA-A3-binding peptide Flu NP265-273 (ILRGSVAHK), and a non-binding peptide CHGA382-390 (HPVGEADYF) were included as positive and negative controls, respectively.
Recommended publications
  • Resolving Transcriptional States and Predicting Lineages in the Annelid Capitella Teleta Using 1 Single-Cell Rnaseq 2 3 Abhinav
    bioRxiv preprint doi: https://doi.org/10.1101/2020.10.16.342709; this version posted October 16, 2020. 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. 1 Resolving transcriptional states and predicting lineages in the annelid Capitella teleta using 2 single-cell RNAseq 3 4 Abhinav Sur1 and Néva P. Meyer2* 5 6 1Unit on Cell Specification and Differentiation, National Institute of Child Health and Human 7 Development (NICHD), Bethesda, Maryland, USA, 20814 8 9 2Department of Biology, Clark University, 950 Main Street, Worcester, Massachusetts, USA, 10 01610. 11 12 13 [email protected] 14 [email protected] 15 16 *Corresponding author 17 18 19 20 21 Keywords: neurogenesis, single-cell RNAseq, annelid, cell type, differentiation trajectory, 22 pseudotime, RNA velocity, gene regulatory network. 23 bioRxiv preprint doi: https://doi.org/10.1101/2020.10.16.342709; this version posted October 16, 2020. 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. 24 Abstract 25 Evolution and diversification of cell types has contributed to animal evolution. However, gene 26 regulatory mechanisms underlying cell fate acquisition during development remains largely 27 uncharacterized in spiralians. Here we use a whole-organism, single-cell transcriptomic approach 28 to map larval cell types in the annelid Capitella teleta at 24- and 48-hours post gastrulation 29 (stages 4 and 5).
    [Show full text]
  • HNF6 Antibody (R31338)
    HNF6 Antibody (R31338) Catalog No. Formulation Size R31338 0.5mg/ml if reconstituted with 0.2ml sterile DI water 100 ug Bulk quote request Availability 1-3 business days Species Reactivity Human, Mouse, Rat Format Antigen affinity purified Clonality Polyclonal (rabbit origin) Isotype Rabbit IgG Purity Antigen affinity Buffer Lyophilized from 1X PBS with 2.5% BSA and 0.025% sodium azide/thimerosal UniProt Q9UBC0 Applications Western blot : 0.5-1ug/ml IHC (FFPE) : 0.5-1ug/ml IHC (Frozen) : 0.5-1ug/ml Immunocytochemistry : 0.5-1ug/ml Limitations This HNF6 antibody is available for research use only. Western blot testing of HNF6 antibody and Lane 1: rat liver; 2: mouse liver; 3: human HeLa cell lysate. Expected/observed size ~51KD IHC-P: HNF6 antibody testing of human liver cancer tissue ICC testing of HNF6 antibody and HCT116 cells IHC-F testing of rat liver tissue IHC-P testing of rat liver tissue Description One cut homeobox 1 (ONECUT1), also called Hepatocyte nuclear factor 6 (HNF6) is found strong expression in liver and lower expression in testis and skin. The gene encodes a member of the Cut homeobox family of transcription factors. Expression of the encoded protein is enriched in the liver, where it stimulates transcription of liver-expressed genes, and antagonizes glucocorticoid-stimulated gene transcription. This gene may influence a variety of cellular processes including glucose metabolism, cell cycle regulation, and it may also be associated with cancer. Application Notes The stated application concentrations are suggested starting amounts. Titration of the HNF6 antibody may be required due to differences in protocols and secondary/substrate sensitivity.
    [Show full text]
  • Downloaded from the National Database for Autism Research (NDAR)
    International Journal of Molecular Sciences Article Phenotypic Subtyping and Re-Analysis of Existing Methylation Data from Autistic Probands in Simplex Families Reveal ASD Subtype-Associated Differentially Methylated Genes and Biological Functions Elizabeth C. Lee y and Valerie W. Hu * Department of Biochemistry and Molecular Medicine, The George Washington University, School of Medicine and Health Sciences, Washington, DC 20037, USA; [email protected] * Correspondence: [email protected]; Tel.: +1-202-994-8431 Current address: W. Harry Feinstone Department of Molecular Microbiology and Immunology, y Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205, USA. Received: 25 August 2020; Accepted: 17 September 2020; Published: 19 September 2020 Abstract: Autism spectrum disorder (ASD) describes a group of neurodevelopmental disorders with core deficits in social communication and manifestation of restricted, repetitive, and stereotyped behaviors. Despite the core symptomatology, ASD is extremely heterogeneous with respect to the severity of symptoms and behaviors. This heterogeneity presents an inherent challenge to all large-scale genome-wide omics analyses. In the present study, we address this heterogeneity by stratifying ASD probands from simplex families according to the severity of behavioral scores on the Autism Diagnostic Interview-Revised diagnostic instrument, followed by re-analysis of existing DNA methylation data from individuals in three ASD subphenotypes in comparison to that of their respective unaffected siblings. We demonstrate that subphenotyping of cases enables the identification of over 1.6 times the number of statistically significant differentially methylated regions (DMR) and DMR-associated genes (DAGs) between cases and controls, compared to that identified when all cases are combined. Our analyses also reveal ASD-related neurological functions and comorbidities that are enriched among DAGs in each phenotypic subgroup but not in the combined case group.
    [Show full text]
  • Genome-Wide Association Study Identifies Eight Novel Loci
    cells Article Genome-Wide Association Study Identifies Eight Novel Loci for Susceptibility of Scrub Typhus and Highlights Immune-Related Signaling Pathways in Its Pathogenesis Yong-Chan Kim 1,2, Soriul Kim 3, Hee-Kwon Kim 4, Yi Lee 5 , Chol Shin 3,6, Chang-Seop Lee 7,8,* and Byung-Hoon Jeong 1,2,* 1 Korea Zoonosis Research Institute, Jeonbuk National University, Iksan, Jeonbuk 54531, Korea; [email protected] 2 Department of Bioactive Material Sciences, Jeonbuk National University, Jeonju, Jeonbuk 54896, Korea 3 Institute for Human Genomic Study, College of Medicine, Korea University, Seoul 02841, Korea; [email protected] (S.K.); [email protected] (C.S.) 4 Molecular Imaging & Therapeutic Medicine Research Center, Department of Nuclear Medicine, Biomedical Research Institute, Jeonbuk National University Medical School and Hospital, Jeonju, Jeonbuk 54907, Korea; [email protected] 5 Department of Industrial Plant Science & Technology, Chungbuk National University, Chungju, Chungbuk 28644, Korea; [email protected] 6 Department of Internal Medicine, Division of Pulmonary Sleep and Critical Care Medicine, Korea University Ansan Hospital, Ansan 15355, Korea 7 Department of Internal Medicine, Research Institute of Clinical Medicine, Jeonbuk National University Medical School, Jeonju, Jeonbuk 54907, Korea Citation: Kim, Y.-C.; Kim, S.; Kim, 8 Biomedical Research Institute, Jeonbuk National University Hospital, Jeonju, Jeonbuk 54907, Korea H.-K.; Lee, Y.; Shin, C.; Lee, C.-S.; * Correspondence: [email protected] (C.-S.L.); [email protected] (B.-H.J.); Jeong, B.-H. Genome-Wide Tel.: +82-63-250-2391 (C.-S.L.); +82-63-900-4040 (B.-H.J.); Fax: +82-63-254-1609 (C.-S.L.); +82-63-900-4012 (B.-H.J.) Association Study Identifies Eight Novel Loci for Susceptibility of Scrub Abstract: Scrub typhus is a fatal zoonotic disease caused by Orientia tsutsugamushi.
    [Show full text]
  • Identification of Potential Key Genes and Pathway Linked with Sporadic Creutzfeldt-Jakob Disease Based on Integrated Bioinformatics Analyses
    medRxiv preprint doi: https://doi.org/10.1101/2020.12.21.20248688; this version posted December 24, 2020. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted medRxiv a license to display the preprint in perpetuity. All rights reserved. No reuse allowed without permission. Identification of potential key genes and pathway linked with sporadic Creutzfeldt-Jakob disease based on integrated bioinformatics analyses Basavaraj Vastrad1, Chanabasayya Vastrad*2 , Iranna Kotturshetti 1. Department of Biochemistry, Basaveshwar College of Pharmacy, Gadag, Karnataka 582103, India. 2. Biostatistics and Bioinformatics, Chanabasava Nilaya, Bharthinagar, Dharwad 580001, Karanataka, India. 3. Department of Ayurveda, Rajiv Gandhi Education Society`s Ayurvedic Medical College, Ron, Karnataka 562209, India. * Chanabasayya Vastrad [email protected] Ph: +919480073398 Chanabasava Nilaya, Bharthinagar, Dharwad 580001 , Karanataka, India NOTE: This preprint reports new research that has not been certified by peer review and should not be used to guide clinical practice. medRxiv preprint doi: https://doi.org/10.1101/2020.12.21.20248688; this version posted December 24, 2020. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted medRxiv a license to display the preprint in perpetuity. All rights reserved. No reuse allowed without permission. Abstract Sporadic Creutzfeldt-Jakob disease (sCJD) is neurodegenerative disease also called prion disease linked with poor prognosis. The aim of the current study was to illuminate the underlying molecular mechanisms of sCJD. The mRNA microarray dataset GSE124571 was downloaded from the Gene Expression Omnibus database. Differentially expressed genes (DEGs) were screened.
    [Show full text]
  • Supplementary Materials
    Supplementary materials Supplementary Table S1: MGNC compound library Ingredien Molecule Caco- Mol ID MW AlogP OB (%) BBB DL FASA- HL t Name Name 2 shengdi MOL012254 campesterol 400.8 7.63 37.58 1.34 0.98 0.7 0.21 20.2 shengdi MOL000519 coniferin 314.4 3.16 31.11 0.42 -0.2 0.3 0.27 74.6 beta- shengdi MOL000359 414.8 8.08 36.91 1.32 0.99 0.8 0.23 20.2 sitosterol pachymic shengdi MOL000289 528.9 6.54 33.63 0.1 -0.6 0.8 0 9.27 acid Poricoic acid shengdi MOL000291 484.7 5.64 30.52 -0.08 -0.9 0.8 0 8.67 B Chrysanthem shengdi MOL004492 585 8.24 38.72 0.51 -1 0.6 0.3 17.5 axanthin 20- shengdi MOL011455 Hexadecano 418.6 1.91 32.7 -0.24 -0.4 0.7 0.29 104 ylingenol huanglian MOL001454 berberine 336.4 3.45 36.86 1.24 0.57 0.8 0.19 6.57 huanglian MOL013352 Obacunone 454.6 2.68 43.29 0.01 -0.4 0.8 0.31 -13 huanglian MOL002894 berberrubine 322.4 3.2 35.74 1.07 0.17 0.7 0.24 6.46 huanglian MOL002897 epiberberine 336.4 3.45 43.09 1.17 0.4 0.8 0.19 6.1 huanglian MOL002903 (R)-Canadine 339.4 3.4 55.37 1.04 0.57 0.8 0.2 6.41 huanglian MOL002904 Berlambine 351.4 2.49 36.68 0.97 0.17 0.8 0.28 7.33 Corchorosid huanglian MOL002907 404.6 1.34 105 -0.91 -1.3 0.8 0.29 6.68 e A_qt Magnogrand huanglian MOL000622 266.4 1.18 63.71 0.02 -0.2 0.2 0.3 3.17 iolide huanglian MOL000762 Palmidin A 510.5 4.52 35.36 -0.38 -1.5 0.7 0.39 33.2 huanglian MOL000785 palmatine 352.4 3.65 64.6 1.33 0.37 0.7 0.13 2.25 huanglian MOL000098 quercetin 302.3 1.5 46.43 0.05 -0.8 0.3 0.38 14.4 huanglian MOL001458 coptisine 320.3 3.25 30.67 1.21 0.32 0.9 0.26 9.33 huanglian MOL002668 Worenine
    [Show full text]
  • Analysis of the Indacaterol-Regulated Transcriptome in Human Airway
    Supplemental material to this article can be found at: http://jpet.aspetjournals.org/content/suppl/2018/04/13/jpet.118.249292.DC1 1521-0103/366/1/220–236$35.00 https://doi.org/10.1124/jpet.118.249292 THE JOURNAL OF PHARMACOLOGY AND EXPERIMENTAL THERAPEUTICS J Pharmacol Exp Ther 366:220–236, July 2018 Copyright ª 2018 by The American Society for Pharmacology and Experimental Therapeutics Analysis of the Indacaterol-Regulated Transcriptome in Human Airway Epithelial Cells Implicates Gene Expression Changes in the s Adverse and Therapeutic Effects of b2-Adrenoceptor Agonists Dong Yan, Omar Hamed, Taruna Joshi,1 Mahmoud M. Mostafa, Kyla C. Jamieson, Radhika Joshi, Robert Newton, and Mark A. Giembycz Departments of Physiology and Pharmacology (D.Y., O.H., T.J., K.C.J., R.J., M.A.G.) and Cell Biology and Anatomy (M.M.M., R.N.), Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada Received March 22, 2018; accepted April 11, 2018 Downloaded from ABSTRACT The contribution of gene expression changes to the adverse and activity, and positive regulation of neutrophil chemotaxis. The therapeutic effects of b2-adrenoceptor agonists in asthma was general enriched GO term extracellular space was also associ- investigated using human airway epithelial cells as a therapeu- ated with indacaterol-induced genes, and many of those, in- tically relevant target. Operational model-fitting established that cluding CRISPLD2, DMBT1, GAS1, and SOCS3, have putative jpet.aspetjournals.org the long-acting b2-adrenoceptor agonists (LABA) indacaterol, anti-inflammatory, antibacterial, and/or antiviral activity. Numer- salmeterol, formoterol, and picumeterol were full agonists on ous indacaterol-regulated genes were also induced or repressed BEAS-2B cells transfected with a cAMP-response element in BEAS-2B cells and human primary bronchial epithelial cells by reporter but differed in efficacy (indacaterol $ formoterol .
    [Show full text]
  • Neuroscience Recombinant Proteins & Monoclonal Antibodies
    www.atgenglobal.com Neuroscience Recombinant Proteins & Monoclonal Antibodies Global Bio Frontier Global Bio Frontier Neurodegenerative Disease Neurodegeneration is the progressive loss of Prion disease structure or function of neurons, including death of Frontotemporal dementia Alzheimer's disease neurons. Many neurodegenerative diseases – including amyotrophic lateral sclerosis, Parkinson's disease, Alzheimer's disease, and Huntington's disease – occur as a result of neurodegenerative processes. Such diseases are incurable, resulting in progres- Lewy body dementia sive degeneration and/or death of neuron cells. As Parkinson's disease research progresses, many similarities appear that relate these diseases to one another on a sub-cellular Huntington's disease level. Neurodegeneration can be found in many different levels of neuronal circuitry ranging from molecular to systemic. Amyotrophic Lateral Sclerosis Related Monoclonal Antibodies Product name Clone No. Applications Host Cat No. AT1E10 ELISA, WB, ICC/IF M ATGA0244 alpha-Synuclein 5C2 ELISA, WB, ICC/IF M ANC0603 alpha/beta-Synuclein 3B6 ELISA, WB, FACS, ICC/IF M ASC0632 HMGB1/HMG-1 J2E1 ELISA, WB, IHC M AHM0915 AT1E12 ELISA, WB, FACS, ICC/IF M ATGA0292 Park7/DJ-1 1B11 ELISA, WB M ADJ0618 UCH-L1/PGP9.5 3D9 ELISA, WB, ICC/IF, IHC M APG0714 AT3F9 ELISA, WB, ICC/IF M ATGA0342 HTRA2/Omi 1B3 ELISA, WB M AOH0607 APP/Protease Nexin II J4H9 ELISA, WB, FACS, ICC/IF M AAP0836 GSK-3 beta 1F7 ELISA, WB, FACS, ICC/IF M AGK0601 SOD1/Cu-Zn SOD AT6F1 ELISA, WB, FACS, ICC/IF M ATGA0405 14-3-3 gamma AT4B9 ELISA, WB, ICC/IF M ATGA0312 14-3-3 epsilon AT4F8 ELISA, WB, ICC/IF M ATGA0124 14-3-3 theta AT1A1 ELISA, WB, ICC/IF M ATGA0132 CAPNS1 AT1D11 ELISA, WB, ICC/IF M ATGA0367 TDP-43/TARDBP k1B8 ELISA, WB, IHC M ATA0834 Alpha-Synuclein antibody (AT1E10) UCH-L1/PGP9.5 antibody (3D9) HMGB1/HMG-1 antibody (J2E1) (Cat No.
    [Show full text]
  • Proteome Dynamics During Homeostatic Scaling in Cultured
    RESEARCH ADVANCE Proteome dynamics during homeostatic scaling in cultured neurons Aline Ricarda Do¨ rrbaum1,2, Beatriz Alvarez-Castelao1, Belquis Nassim-Assir1, Julian D Langer1,3*, Erin M Schuman1* 1Max Planck Institute for Brain Research, Frankfurt, Germany; 2Goethe University Frankfurt, Faculty of Biological Sciences, Frankfurt, Germany; 3Max Planck Institute of Biophysics, Frankfurt, Germany Abstract Protein turnover, the net result of protein synthesis and degradation, enables cells to remodel their proteomes in response to internal and external cues. Previously, we analyzed protein turnover rates in cultured brain cells under basal neuronal activity and found that protein turnover is influenced by subcellular localization, protein function, complex association, cell type of origin, and by the cellular environment (Do¨ rrbaum et al., 2018). Here, we advanced our experimental approach to quantify changes in protein synthesis and degradation, as well as the resulting changes in protein turnover or abundance in rat primary hippocampal cultures during homeostatic scaling. Our data demonstrate that a large fraction of the neuronal proteome shows changes in protein synthesis and/or degradation during homeostatic up- and down-scaling. More than half of the quantified synaptic proteins were regulated, including pre- as well as postsynaptic proteins with diverse molecular functions. Introduction *For correspondence: Long-lasting changes in synaptic strength, which are a basis for learning and memory formation, [email protected] (JDL); require de-novo protein synthesis as well as the degradation of existing proteins (Cajigas et al., [email protected] (EMS) 2010; Hegde, 2017; Jarome and Helmstetter, 2013; Tai and Schuman, 2008). For example, injec- tions of the protein synthesis inhibitor puromycin (Flexner et al., 1963) or of the proteasome inhibi- Competing interests: The tor lactacystin (Lopez-Salon et al., 2001) into rodent brains during specific time windows after authors declare that no training blocked long-term memory formation.
    [Show full text]
  • Strand Breaks for P53 Exon 6 and 8 Among Different Time Course of Folate Depletion Or Repletion in the Rectosigmoid Mucosa
    SUPPLEMENTAL FIGURE COLON p53 EXONIC STRAND BREAKS DURING FOLATE DEPLETION-REPLETION INTERVENTION Supplemental Figure Legend Strand breaks for p53 exon 6 and 8 among different time course of folate depletion or repletion in the rectosigmoid mucosa. The input of DNA was controlled by GAPDH. The data is shown as ΔCt after normalized to GAPDH. The higher ΔCt the more strand breaks. The P value is shown in the figure. SUPPLEMENT S1 Genes that were significantly UPREGULATED after folate intervention (by unadjusted paired t-test), list is sorted by P value Gene Symbol Nucleotide P VALUE Description OLFM4 NM_006418 0.0000 Homo sapiens differentially expressed in hematopoietic lineages (GW112) mRNA. FMR1NB NM_152578 0.0000 Homo sapiens hypothetical protein FLJ25736 (FLJ25736) mRNA. IFI6 NM_002038 0.0001 Homo sapiens interferon alpha-inducible protein (clone IFI-6-16) (G1P3) transcript variant 1 mRNA. Homo sapiens UDP-N-acetyl-alpha-D-galactosamine:polypeptide N-acetylgalactosaminyltransferase 15 GALNTL5 NM_145292 0.0001 (GALNT15) mRNA. STIM2 NM_020860 0.0001 Homo sapiens stromal interaction molecule 2 (STIM2) mRNA. ZNF645 NM_152577 0.0002 Homo sapiens hypothetical protein FLJ25735 (FLJ25735) mRNA. ATP12A NM_001676 0.0002 Homo sapiens ATPase H+/K+ transporting nongastric alpha polypeptide (ATP12A) mRNA. U1SNRNPBP NM_007020 0.0003 Homo sapiens U1-snRNP binding protein homolog (U1SNRNPBP) transcript variant 1 mRNA. RNF125 NM_017831 0.0004 Homo sapiens ring finger protein 125 (RNF125) mRNA. FMNL1 NM_005892 0.0004 Homo sapiens formin-like (FMNL) mRNA. ISG15 NM_005101 0.0005 Homo sapiens interferon alpha-inducible protein (clone IFI-15K) (G1P2) mRNA. SLC6A14 NM_007231 0.0005 Homo sapiens solute carrier family 6 (neurotransmitter transporter) member 14 (SLC6A14) mRNA.
    [Show full text]
  • Cis-Regulatory Analysis of Onecut1 Expression in Fate
    bioRxiv preprint doi: https://doi.org/10.1101/854778; this version posted November 26, 2019. 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 4.0 International license. 1 Cis-regulatory analysis of Onecut1 expression in fate-restricted retinal progenitor cells 2 3 Sruti Patoori1,2, Nathalie Jean-Charles2, Ariana Gopal2, Sacha Sulaiman2, Sneha Gopal2,3, Brian 4 Wang2, Benjamin Souferi2,4, Mark M. Emerson1,2,5 5 6 1 Biology PhD Program, The Graduate Center, The City University of New York, New York, NY, 7 10016 8 2 Department of Biology, The City College of New York, The City University of New York, New York, 9 NY, 10031 10 3 Present Address: Doctoral program in Department of Chemical and Biological Engineering, 11 Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, Troy , 12 NY 12180 13 4 Present Address: Touro College of Osteopathic Medicine, New York, NY 10027 14 5 Biochemistry PhD Program, Graduate Center, City University of New York, NY 10016 15 16 *Corresponding author: [email protected] 17 18 Email addresses: 19 20 Sruti Patoori: [email protected] 21 Ariana Gopal: [email protected] 22 Sacha Sulaiman: [email protected] 23 Nathalie Jean-Charles: [email protected] 24 Sneha Gopal: [email protected] 25 Brian Wang: [email protected] 26 Benjamin Souferi: [email protected] 27 Mark Emerson: [email protected] 28 29 30 31 32 33 Running title: Retinal fate-restricted cis-regulatory elements 34 35 36 37 Keywords: retinal progenitor cells, multipotent, fate-restricted, cone photoreceptors, basic 38 helix-loop-helix, chicken, electroporation 39 40 41 42 43 1 bioRxiv preprint doi: https://doi.org/10.1101/854778; this version posted November 26, 2019.
    [Show full text]
  • 1 1 2 3 Cell Type-Specific Transcriptomics of Hypothalamic
    1 2 3 4 Cell type-specific transcriptomics of hypothalamic energy-sensing neuron responses to 5 weight-loss 6 7 Fredrick E. Henry1,†, Ken Sugino1,†, Adam Tozer2, Tiago Branco2, Scott M. Sternson1,* 8 9 1Janelia Research Campus, Howard Hughes Medical Institute, 19700 Helix Drive, Ashburn, VA 10 20147, USA. 11 2Division of Neurobiology, Medical Research Council Laboratory of Molecular Biology, 12 Cambridge CB2 0QH, UK 13 14 †Co-first author 15 *Correspondence to: [email protected] 16 Phone: 571-209-4103 17 18 Authors have no competing interests 19 1 20 Abstract 21 Molecular and cellular processes in neurons are critical for sensing and responding to energy 22 deficit states, such as during weight-loss. AGRP neurons are a key hypothalamic population 23 that is activated during energy deficit and increases appetite and weight-gain. Cell type-specific 24 transcriptomics can be used to identify pathways that counteract weight-loss, and here we 25 report high-quality gene expression profiles of AGRP neurons from well-fed and food-deprived 26 young adult mice. For comparison, we also analyzed POMC neurons, an intermingled 27 population that suppresses appetite and body weight. We find that AGRP neurons are 28 considerably more sensitive to energy deficit than POMC neurons. Furthermore, we identify cell 29 type-specific pathways involving endoplasmic reticulum-stress, circadian signaling, ion 30 channels, neuropeptides, and receptors. Combined with methods to validate and manipulate 31 these pathways, this resource greatly expands molecular insight into neuronal regulation of 32 body weight, and may be useful for devising therapeutic strategies for obesity and eating 33 disorders.
    [Show full text]