Control of CD4 Helper and CD8 Cytotoxic T Cell Differentiation

Total Page:16

File Type:pdf, Size:1020Kb

Control of CD4 Helper and CD8 Cytotoxic T Cell Differentiation https://www.scientificarchives.com/journal/journal-of-cellular-immunology Journal of Cellular Immunology Commentary Control of CD4 Helper and CD8 Cytotoxic T cell Differentiation Vibhuti P Dave* Département d’Immunologie-Oncologie, Centre de Recherche Hôpital Maisonneuve-Rosemont, Montreal, Quebec H1T 2M4, Canada *Correspondence should be addressed to Vibhuti P Dave; [email protected] Received date: June 29, 2020, Accepted date: July 15, 2020 Copyright: © 2020 Dave VP. This is an open-access article distributed under the terms of the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited. Engagement of mature T cell receptor (TCR), a culminating in shorter or weaker signal transduction multiprotein complex consisting of an αβ heterodimer in MHCI-signaled thymocytes, while continued CD4 associated with invariant CD3 signaling proteins, on expression at this stage, and therefore higher Lck activity, CD4+CD8+ double positive (DP) thymocytes by self- results in sustained or stronger signal in MHCII-signaled peptide/self-MHC complex on thymic stromal cells thymocytes. However, two observations suggest that results in negative selection of thymocytes expressing kinetic signal strength model alone cannot explain the strong affinity TCRs and positive selection of thymocytes CD4 helper versus CD8 cytotoxic lineage choice; (a) expressing weak affinity TCRs. Positively selected constitutive CD8 or chimeric CD8.4 (cytoplasmic domain thymocytes almost invariably differentiate into MHCII- of CD8 substituted by that of CD4) redirects only a fraction specific CD4+ helper and MHCI-specific CD8+ cytotoxic T of MHCI-signaled thymocytes into CD4 lineage [14] and cells endowed with effector functions crucial for effective (b) positively selected MHCI-restricted thymocytes cell-mediated immune responses [1,2]. Together, thymic expressing strong affinity TCR such as OTI- TCR and P14- selection and lineage choice ensures generation of mature TCR, which transduce stronger signal compared to several T cell repertoire that is capable of mounting effective MHCII-specific TCRs such as OTII-TCR, fail to develop responses to non-self-antigens but is tolerant to self- into the CD4 helper lineage [15]. It is proposed that signal antigens encountered in the periphery [3,4]. Although disruption renders MHCI-signaled thymocytes sensitive correlation between MHC specificity with helper and to cytokine signaling critical for Runx3 activation and the cytotoxic lineage choice remains unclear, it appears to CD8 cytotoxic lineage choice [16,17], while stronger TCR be influenced by duration and intensity of TCR signaling signal induces ThPOK (Zbtb7b, cKrox) in MHCII- signal that imprints transcriptional program underlying the thymocytes essential for the CD4 helper lineage choice two lineages [5]. Specificity of CD4 and CD8 coreceptor [18]. Identification of Helper Deficient (HD) mouse and for MHCII and MHCI, respectively, is important in TCR subsequent research showed that loss and gain of ThPOK signaling with CD4, compared to CD8, promoting stronger function redirects, respectively, MHCII-specific cells signaling due to stronger association of Src tyrosine kinase into the CD8 cytotoxic lineage and MHCI-specific cells Lck with cytoplasmic tail of CD4 than that of CD8 [6-10]. into the CD4 helper lineage [19-22]. In contrast, Runx3 Irrespective of MHC specificity positively selected DP deficiency or constitutive expression alters lineage choice thymocytes temporally downregulate Cd8 transcription of only a fraction of MHCI-signaled thymocytes [23-26]. and become CD4+CD8lo thymocytes that eventually Collectively, these observations suggest a dominant role develop into CD4+ and CD8+ single positive (SP) mature for ThPOK in the CD4 helper versus CD8 cytotoxic lineage thymocytes [3,11]. Indeed, ablating CD4 expression at choice of developing thymocytes. CD4+CD8lo stage redirects positively selected thymocytes into the cytotoxic lineage [12]. Thus, kinetics of coreceptor, Although MHC specificity of positively selected thymocytes particularly CD4, expression plays an important role in CD4 correlates with functionally distinct lineages, cellular vs CD8 lineage choice. Based on these observations kinetic components associated with proximal TCR signaling signal strength model has been proposed for CD4/CD8 are similar except for stronger Lck association with the lineage choice [13]. Accordingly, in CD4+CD8lo thymocytes cytoplasmic tail of CD4 than that of CD8. However, higher downregulation of CD8 results in reduced Lck activity Lck activity in MHCII-signaled cells alone cannot explain J Cell Immunol. 2020 Volume 2, Issue 5 220 Dave VP. Control of CD4 Helper and CD8 Cytotoxic T cell Differentiation. J Cell Immunol. 2020; 2(5): 220-226. CD4 lineage choice as constitutively active or dominant developing thymocytes [33,34]. Our subsequent analysis negative Lck alters lineage fate of only a small number showed that the same ThPOK transgene completely of positively selected thymocytes expressing monoclonal restores CD4 development of MHCII-specific OTII-TCR in TCR [8,9]. Similarly, temporal and differential kinetics of Thpok-/- mice providing first indication that, compared to ZAP70 expression, delayed and higher expression in the MHCII-signaled thymocytes, MHCI-signaled thymocytes CD8 compared to CD4 committed thymocytes, is proposed require significantly higher amount of ThPOK for efficient to play a role in lineage divergence although mechanism CD4 lineage choice. Further experimentations showed of its action remains undefined [5,27-29]. How differences that augmenting TCR signal strength via introduction of in kinetics and/or strength of proximal TCR signaling constitutively active Lck increases the efficiency of ThPOK- lead to activation of lineage specifying ThPOK and Runx3 induced CD4 lineage choice of MHCI-specific thymocytes expression in MHCII- and MHCI-signaled thymocytes, although it was still lower compared to the CD4 lineage respectively, is challenging and remains to be elucidated. choice of MHCII-signaled thymocytes expressing the same amount of ThPOK. Together these analyses suggest that Central role for ThPOK in defining the CD4 versus CD8 suppression of cytotoxic and induction of helper program lineage divergence has prompted intense investigation require different amount of ThPOK, and strength and of this BTB/POZ zinc finger transcription factor over the quality of TCR signaling largely determines the efficiency last decade. Previous studies showed that constitutive of ThPOK-induced CD4 lineage choice. ThPOK expression redirects MHCI-signaled thymocytes into the CD4 helper lineage [20,30]; however it remained How might TCR signal strength and MHC specificity unclear if a defined amount of ThPOK promoted the CD4 segregate CD4 versus CD8 lineage choice? Mechanistically lineage choice of MHCI- and MHCII-signaled thymocytes there are two mutually nonexclusive possibilities that may at comparable efficiency and whether TCR signal strength underlie differential impact of a defined amount of ThPOK or MHC specificity played any role in this process. In our on lineage choice. It is conceivable that during a temporal recently published article, we evaluated role of ThPOK window of lineage choice [35], accessibility of genes dose and TCR signal strength on the CD4 lineage choice regulated by ThPOK or Runx3 is influenced by strength of MHCI- and MHCII-signaled thymocytes [31]. Analysis and duration of TCR signaling; stronger and longer of three independent transgenic mouse lines expressing duration of TCR signal in MHCII-signaled thymocytes different amounts of ThPOK in developing thymocytes may permit target gene loci access for an extended period showed that the efficiency of CD4 lineage choice of MHCI- requiring a smaller amount of ThPOK for modulating their signaled thymocytes expressing monoclonal (OTI- and expression leading to suppression of cytotoxic program P14-TCR) or polyclonal (MHCII-/- mice) TCR specificities and induction of helper program. Conversely, TCR was directly proportional to ThPOK levels in the signal disruption in MHCI-signaled thymocytes reduced preselection DP thymocytes. These data suggest the extent accessibility of target genes would require significantly of transgenic ThPOK occupancy at the target gene loci more ThPOK (than the amount required for CD4 lineage may prime positively selected MHCI-signaled thymocytes choice of MHCII-signaled thymocytes) for the CD4 for the CD4 lineage development. The ability of retrogenic helper lineage choice. Increased efficiency of CD4 lineage ThPOK expression to only partially reprogram peripheral choice of MHCI-specific thymocytes due to augmented CD8 cytotoxic T cells into the CD4 helper T cell supports TCR signaling in ThPOK transgenic mice supports this this possibility [30]. Interestingly, all three transgenic notion [31]. Such a temporal target gene accessibility lines expressed higher amount of ThPOK compared to model may explain incomplete modulation of helper endogenous ThPOK levels observed in CD4+ T cells from and cytotoxic lineage genes in peripheral CD8 cytotoxic WT mice, and yet the lowest ThPOK expressing transgene T cells following enforced ThPOK expression [30] or in (ThPOK-H transgene) caused partial CD8 to CD4 lineage developing thymocytes with impaired Runx function redirection. As expected Runx3 was suppressed in the due to compound deficiency of Tel/Groucho proteins
Recommended publications
  • 2007-05 TLE1 Synovial Sarcoma
    TLE1 Immunostains in the Diagnosis of Synovial Sarcoma May 2007 by Rodney T. Miller, M.D., Director of Immunohistochemistry The diagnosis of synovial sarcoma can be a chal- lenging task, particularly on small biopsy specimens, as the morphologic features of this tumor can be mimicked by a variety of other neoplasms. This month we call attention to a paper published in the February 2007 edition of the American Journal of Surgical Pathology describing the utility of immu- nostains for TLE1 in the diagnosis of this tumor. TLE immunostains are now available at ProPath. Synovial sarcoma occurs in three morphologic varie- ties: monophasic, biphasic, and poorly differentiated. It has been known for some time that synovial sar- coma is associated with a specific chromosomal H&E (left) and TLE1 immunostain (right) on a monophasic translocation, t(X;18), that results in the fusion of the synovial sarcoma. Note the numerous strongly positive nuclei SYT gene on chromosome 18 to either the SSX1 or on the TLE1 immunostain, a typical feature of this tumor. SSX2 gene on the X chromosome, resulting in the production of a SYT-SSX fusion protein. Identifica- In their study, the authors performed TLE1 immu- tion of this translocation in the appropriate setting is nostains on multiple tissue microarrays using two dif- regarded by many to be diagnostic of synovial sar- ferent antibodies (monoclonal and polyclonal), both coma. However, the methodologies used for this performing in a similar fashion. A total of 693 adult purpose (cytogenetics, fluorescent in situ hybridiza- soft tissue tumors were examined, including 94 cases tion, and reverse-transcriptase polymerase chain re- of synovial sarcoma that had documentation of the t action) are not readily available in many diagnostic (X;18) translocation.
    [Show full text]
  • Characterizing Novel Interactions of Transcriptional Repressor Proteins BCL6 & BCL6B
    Characterizing Novel Interactions of Transcriptional Repressor Proteins BCL6 & BCL6B by Geoffrey Graham Lundell-Smith A thesis submitted in conformity with the requirements for the degree of Master of Science Department of Biochemistry University of Toronto © Copyright by Geoffrey Lundell-Smith, 2017 Characterizing Novel Interactions of Transcriptional Repression Proteins BCL6 and BCL6B Geoffrey Graham Lundell-Smith Masters of Science Department of Biochemistry University of Toronto 2016 Abstract B-cell Lymphoma 6 (BCL6) and its close homolog BCL6B encode proteins that are members of the BTB-Zinc Finger family of transcription factors. BCL6 plays an important role in regulating the differentiation and proliferation of B-cells during the adaptive immune response, and is also involved in T cell development and inflammation. BCL6 acts by repressing genes involved in DNA damage response during the affinity maturation of immunoglobulins, and the mis- expression of BCL6 can lead to diffuse large B-cell lymphoma. Although BCL6B shares high sequence similarity with BCL6, the functions of BCL6B are not well-characterized. I used BioID, an in vivo proximity-dependent labeling method, to identify novel BCL6 and BCL6B protein interactors and validated a number of these interactions with co-purification experiments. I also examined the evolutionary relationship between BCL6 and BCL6B and identified conserved residues in an important interaction interface that mediates corepressor binding and gene repression. ii Acknowledgments Thank you to my supervisor, Gil Privé for his mentorship, guidance, and advice, and for giving me the opportunity to work in his lab. Thanks to my committee members, Dr. John Rubinstein and Dr. Jeff Lee for their ideas, thoughts, and feedback during my Masters.
    [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]
  • Hes6 Inhibits Astrocyte Differentiation and Promotes Neurogenesis Through Different Mechanisms
    The Journal of Neuroscience, October 25, 2006 • 26(43):11061–11071 • 11061 Cellular/Molecular Hes6 Inhibits Astrocyte Differentiation and Promotes Neurogenesis through Different Mechanisms Sumit Jhas, Sorana Ciura,* Stephanie Belanger-Jasmin,* Zhifeng Dong, Estelle Llamosas, Francesca M. Theriault, Kerline Joachim, Yeman Tang, Lauren Liu, Jisheng Liu, and Stefano Stifani Center for Neuronal Survival, Montreal Neurological Institute, McGill University, Montreal, Quebec, Canada H3A 2B4 The mechanisms regulating the generation of cell diversity in the mammalian cerebral cortex are beginning to be elucidated. In that regard, Hairy/Enhancer of split (Hes) 1 and 5 are basic helix-loop-helix (bHLH) factors that inhibit the differentiation of pluripotent cortical progenitors into neurons. In contrast, a related Hes family member termed Hes6 promotes neurogenesis. It is shown here that knockdown of endogenous Hes6 causes supernumerary cortical progenitors to differentiate into cells that exhibit an astrocytic morphol- ogy and express the astrocyte marker protein GFAP. Conversely, exogenous Hes6 expression in cortical progenitors inhibits astrocyte differentiation. The negative effect of Hes6 on astrocyte differentiation is independent of its ability to promote neuronal differentiation. We also show that neither its proneuronal nor its anti-gliogenic functions appear to depend on Hes6 ability to bind to DNA via the basic arm of its bHLH domain. Both of these activities require Hes6 to be localized to nuclei, but only its anti-gliogenic function depends on two short peptides, LNHLL and WRPW, that are conserved in all Hes6 proteins. These findings suggest that Hes6 is an important regulator of the neurogenic phase of cortical development by promoting the neuronal fate while suppressing astrocyte differentiation.
    [Show full text]
  • Variability in the Degree of Expression of Phosphorylated I B in Chronic
    6796 Vol. 10, 6796–6806, October 15, 2004 Clinical Cancer Research Featured Article Variability in the Degree of Expression of Phosphorylated I␬B␣ in Chronic Lymphocytic Leukemia Cases With Nodal Involvement Antonia Rodrı´guez,1 Nerea Martı´nez,1 changes in the expression profile (mRNA and protein ex- Francisca I. Camacho,1 Elena Ruı´z-Ballesteros,2 pression) and clinical outcome in a series of CLL cases with 2 1 lymph node involvement. Activation of NF-␬B, as deter- Patrocinio Algara, Juan-Fernando Garcı´a, ␬ ␣ 3 5 mined by the expression of p-I B , was associated with the Javier Mena´rguez, Toma´s Alvaro, expression of a set of genes comprising key genes involved in 6 4 Manuel F. Fresno, Fernando Solano, the control of B-cell receptor signaling, signal transduction, Manuela Mollejo,2 Carmen Martin,1 and and apoptosis, including SYK, LYN, BCL2, CCR7, BTK, Miguel A. Piris1 PIK3CD, and others. Cases with increased expression of ␬ ␣ 1Molecular Pathology Program, Centro Nacional de Investigaciones p-I B showed longer overall survival than cases with lower Oncolo´gicas, Madrid, Spain; 2Department of Genetics and Pathology, expression. A Cox regression model was derived to estimate 3 Hospital Virgen de la Salud, Toledo, Spain; Department of some parameters of prognostic interest: IgVH mutational Pathology, Hospital General Universitario Gregorio Maran˜o´n, ␬ ␣ 4 status, ZAP-70, and p-I B expression. The multivariate Madrid, Spain; Department of Hematology, Hospital Nuestra Sen˜ora analysis disclosed p-I␬B␣ and ZAP-70 expression as inde- del Prado, Talavera de la Reina, Toledo, Spain; 5Department of Pathology, Hospital Verge de la Cinta, Tortosa, Spain; pendent prognostic factors of survival.
    [Show full text]
  • RUNX3 Is Multifunctional in Carcinogenesis of Multiple Solid Tumors
    Oncogene (2010) 29, 2605–2615 & 2010 Macmillan Publishers Limited All rights reserved 0950-9232/10 $32.00 www.nature.com/onc REVIEW RUNX3 is multifunctional in carcinogenesis of multiple solid tumors LSH Chuang1 and Y Ito1,2,3 1Cancer Biology Program, Cancer Science Institute of Singapore, National University of Singapore, Singapore; 2Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore and 3Institute of Molecular and Cell Biology, Singapore The study of RUNX3 in tumor pathogenesis is a rapidly Sgpp1, Ncam1, p21WAF1) (Wotton et al., 2008), the expanding area of cancer research. Functional inactiva- RUNX proteins have been shown to perform distinct tion of RUNX3—through mutation, epigenetic silencing, functions that are dependent on tissue type (Brady and or cytoplasmic mislocalization—is frequently observed in Farrell, 2009). This review explores the dualistic solid tumors of diverse origins. This alone indicates that associations of RUNX3 with cancer with emphasis on RUNX3 inactivation is a major risk factor in tumorigen- the emerging concept of RUNX3 as a multifunctional esis and that it occurs early during progression to suppressor of solid tumors. malignancy. Conversely, RUNX3 has also been described to have an oncogenic function in a subset of tumors. Although the mechanism of how RUNX3 switches from RUNX3 inactivation is prevalent in solid tumors tumor suppressive to oncogenic activity is unclear, this is of clinical relevance with implications for cancer detection Hemizygous deletion of RUNX3 gene and prognosis. Recent developments have significantly The location of RUNX3 at chromosome 1p36, a deletion contributed to our understanding of the pleiotropic tumor hotspot in diverse cancers of epithelial, hematopoietic, suppressive properties of RUNX3 that regulate major and neural origins (Bagchi and Mills, 2008), prompts signaling pathways.
    [Show full text]
  • TLE1 Corepressor Complex Mediates a Camkinase II␦-Dependent Neurogenic Gene Activation Pathway
    View metadata, citation and similar papers at core.ac.uk brought to you by CORE provided by Elsevier - Publisher Connector Cell, Vol. 119, 815–829, December 17, 2004, Copyright ©2004 by Cell Press ActivatingthePARP-1SensorComponentoftheGroucho/ TLE1 Corepressor Complex Mediates a CaMKinase II␦-Dependent Neurogenic Gene Activation Pathway Bong-Gun Ju,1 Derek Solum,1 Eun Joo Song,4 entiate into neurons in response to both extrinsic and Kong-Joo Lee,4 David W. Rose,2 intrinsic cues (reviewed in Bally-Cuif and Hammer- Christopher K. Glass,3 and Michael G. Rosenfeld1,* schmidt [2003]). The commitment of neural stem cells 1Howard Hughes Medical Institute to the neuronal fate and their subsequent differentiation 2 Department of Medicine into neurons are controlled, in part, by mechanisms in- Division of Endocrinology volving the opposing activities of transcription factor 3 Department of Cellular and Molecular Medicine families containing the basic helix-loop-helix (bHLH) do- University of California, San Diego main, the Hairy/Enhancer of split (HES) family proteins Department and School of Medicine that negatively regulate neural differentiation, and other La Jolla, California 92093 bHLH family proteins belonging to the Neurogenin, Ash, 4 Center for Cell Signaling Research and NeuroD subgroups, collectively referred to as Division of Molecular Life Sciences and College proneural proteins (reviewed in Bertrand et al. [2002]; of Pharmacy Ross et al. [2003]). Ewha Womans University The inhibitory bHLH protein, HES1, interacts with Seoul 120-750 groucho (Gro)/transducin like enhancer of split (TLE) Korea corepressor in a manner dependent on its C-terminal WRPW motif and serves in a well-described repressor of MASH1 transcription preventing neuronal differentiation Summary (reviewed in Chen and Courey [2000]).
    [Show full text]
  • Genomic Anatomy of the Tyrp1 (Brown) Deletion Complex
    Genomic anatomy of the Tyrp1 (brown) deletion complex Ian M. Smyth*, Laurens Wilming†, Angela W. Lee*, Martin S. Taylor*, Phillipe Gautier*, Karen Barlow†, Justine Wallis†, Sancha Martin†, Rebecca Glithero†, Ben Phillimore†, Sarah Pelan†, Rob Andrew†, Karen Holt†, Ruth Taylor†, Stuart McLaren†, John Burton†, Jonathon Bailey†, Sarah Sims†, Jan Squares†, Bob Plumb†, Ann Joy†, Richard Gibson†, James Gilbert†, Elizabeth Hart†, Gavin Laird†, Jane Loveland†, Jonathan Mudge†, Charlie Steward†, David Swarbreck†, Jennifer Harrow†, Philip North‡, Nicholas Leaves‡, John Greystrong‡, Maria Coppola‡, Shilpa Manjunath‡, Mark Campbell‡, Mark Smith‡, Gregory Strachan‡, Calli Tofts‡, Esther Boal‡, Victoria Cobley‡, Giselle Hunter‡, Christopher Kimberley‡, Daniel Thomas‡, Lee Cave-Berry‡, Paul Weston‡, Marc R. M. Botcherby‡, Sharon White*, Ruth Edgar*, Sally H. Cross*, Marjan Irvani¶, Holger Hummerich¶, Eleanor H. Simpson*, Dabney Johnson§, Patricia R. Hunsicker§, Peter F. R. Little¶, Tim Hubbard†, R. Duncan Campbell‡, Jane Rogers†, and Ian J. Jackson*ʈ *Medical Research Council Human Genetics Unit, Edinburgh EH4 2XU, United Kingdom; †Wellcome Trust Sanger Institute, and ‡Medical Research Council Rosalind Franklin Centre for Genome Research, Hinxton CB10 1SA, United Kingdom; §Life Sciences Division, Oak Ridge National Laboratory, Oak Ridge, TN 37831; and ¶Department of Biochemistry, Imperial College, London SW7 2AZ, United Kingdom Communicated by Liane B. Russell, Oak Ridge National Laboratory, Oak Ridge, TN, January 9, 2006 (received for review September 15, 2005) Chromosome deletions in the mouse have proven invaluable in the deletions also provided the means to produce physical maps of dissection of gene function. The brown deletion complex com- genetic markers. Studies of this kind have been published for prises >28 independent genome rearrangements, which have several loci, including albino (Tyr), piebald (Ednrb), pink-eyed been used to identify several functional loci on chromosome 4 dilution (p), and the brown deletion complex (2–6).
    [Show full text]
  • 1 an Update on Ancillary Techniques in the Diagnosis of Soft Tissue Tumors
    1 An update on ancillary techniques in the diagnosis of soft tissue tumors Andrew Horvai MD PhD Clinical Professor, Pathology INTRODUCTION Mesenchymal neoplasms represent unique diagnostic challenges to the pathologist because of the rarity, large number of discrete entities and need for highly specific diagnoses on ever decreasing sample sizes. Bone and soft tissue tumors represent less than 1% of neoplasia with over 100 unique diagnoses in soft tissue alone.[1] Although routine H&E diagnosis, combined with gross, clinical and radiographic correlation, remains the mainstay of bone and soft tissue pathology, in an effort to improve sensitivity on small specimens and specificity within the myriad of entities with overlapping histologic features, ancillary techniques are often necessary. In the middle of the last century, tissue culture and electron microscopy became available to supplement routine histologic sections for diagnosis. However, the application of immunohistochemistry (IHC) toward the end of the 20th century quickly became the standard ancillary technique for the evaluation of sarcomas. Especially with the introduction of monoclonal antibodies and automated staining platforms, IHC became routinely used in most histology laboratories. Useful antibodies for mesenchymal tumors target (1) lineage specific proteins and (2) proteins that result from tumor-specific genetic or molecular abnormalities. IHC- LINEAGE SPECIFIC PROTEINS Two approaches exist to identify putative lineage specific proteins for IHC. 1) tumors that recapitulate normal mesenchymal cells or their precursors and express proteins, detectible by IHC, specific to that lineage. 2) profiling a tumor for highly expressed gene(s) that are not expressed in morphologic mimics. IHC to detect the protein products of these genes, even if the functions are unknown, can be diagnostically useful.
    [Show full text]
  • Proteomics Analysis Reveals a Th17-Prone Cell Population in Pre-Symptomatic Graft- Versus-Host Disease
    Supplemental Data for: Proteomics analysis reveals a Th17-prone cell population in pre-symptomatic graft- versus-host disease Authors: Wei Li,1* Liangyi Liu,1* Aurelie Gomez,2* Jilu Zhang,1 Abdulraouf Ramadan,1 Qing Zhang,3 Sung W. Choi,2 Peng Zhang,2 Joel K. Greenson,2 Chen Liu,4 Di Jiang,5 Elizabeth Virts,1 Stephanie L. Kelich,1 Hong Wei Chu,5 Ryan Flynn,6 Bruce R. Blazar,6 Helmut Hanenberg,1 Samir Hanash,7 and Sophie Paczesny1‡ Affiliations: 1Indiana University School of Medicine, Indianapolis, IN, USA; 2University of Michigan, Ann Arbor, MI, USA; 3Fred Hutchinson Cancer Research Center, Seattle, WA, USA; 4Rutgers-Robert Wood Johnson Medical School, New Brunswick, NJ, USA; 5National Jewish Health, Denver, CO, USA; 6University of Minnesota, Minneapolis, MN, USA; 7MD Anderson Cancer Center, Houston, TX, USA. *These authors contributed equally to this work. ‡Corresponding author. E-mail: [email protected] This PDF file includes: Supplemental Figure 1. Conventional T cells (Tcons) and Tregs gating strategy. Supplemental Figure 2. Absolute lymphocyte counts and correlation of CD4+CD146+CCR5+ T-cell counts and frequencies of CD4+CD146+CCR5+ T cells in PB of HCT patients. Supplemental Figure 3. Frequencies of CD4+CD146+ T cells and CD4+CCR5+ T cells in PB of HCT patients. Supplemental Figure 4. CD146 expression on CD4 and CD8 T cells in PB of healthy donors and GI-GVHD patients. Supplemental Figure 5. Receiver operating characteristic (ROC) curve comparing GI-GVHD versus non-GVHD enteritis HCT patients. Supplemental Figure 6. CD4+CD146+CCR5+ T-cell frequency stratified by maximum GI- GVHD stage.
    [Show full text]
  • A SARS-Cov-2 Protein Interaction Map Reveals Targets for Drug Repurposing
    Article A SARS-CoV-2 protein interaction map reveals targets for drug repurposing https://doi.org/10.1038/s41586-020-2286-9 A list of authors and affiliations appears at the end of the paper Received: 23 March 2020 Accepted: 22 April 2020 A newly described coronavirus named severe acute respiratory syndrome Published online: 30 April 2020 coronavirus 2 (SARS-CoV-2), which is the causative agent of coronavirus disease 2019 (COVID-19), has infected over 2.3 million people, led to the death of more than Check for updates 160,000 individuals and caused worldwide social and economic disruption1,2. There are no antiviral drugs with proven clinical efcacy for the treatment of COVID-19, nor are there any vaccines that prevent infection with SARS-CoV-2, and eforts to develop drugs and vaccines are hampered by the limited knowledge of the molecular details of how SARS-CoV-2 infects cells. Here we cloned, tagged and expressed 26 of the 29 SARS-CoV-2 proteins in human cells and identifed the human proteins that physically associated with each of the SARS-CoV-2 proteins using afnity-purifcation mass spectrometry, identifying 332 high-confdence protein–protein interactions between SARS-CoV-2 and human proteins. Among these, we identify 66 druggable human proteins or host factors targeted by 69 compounds (of which, 29 drugs are approved by the US Food and Drug Administration, 12 are in clinical trials and 28 are preclinical compounds). We screened a subset of these in multiple viral assays and found two sets of pharmacological agents that displayed antiviral activity: inhibitors of mRNA translation and predicted regulators of the sigma-1 and sigma-2 receptors.
    [Show full text]
  • Immunohistochemical Validation of TLE1, a Novel Marker, for Synovial Sarcomas
    Indian J Med Res 136, November 2012, pp 766-775 Immunohistochemical validation of TLE1, a novel marker, for synovial sarcomas Bharat Rekhi*, Ranjan Basak**, Sangeeta B. Desai*,** & Nirmala A. Jambhekar* *Department of Pathology, Tata Memorial Hospital, Mumbai & **Department of Molecular Pathology, Advanced Centre for Treatment, Research & Education in Cancer (ACTREC), Navi Mumbai, India Received July 26, 2011 Background & objectives: Logistic and financial constraints limit application of several available immunohistochemical (IHC) markers and molecular analysis in every case of synovial sarcoma, diagnosed in our settings. Recently, TLE1 has been recognized as a robust IHC marker for diagnosing a synovial sarcoma. Here, we present IHC features of synovial sarcomas, including TLE1 expression in these cases and in some other tumours. Methods: Conventional sections from 42 synovial sarcomas (30 retrospective & 12 prospectively diagnosed) were subjected to TLE1 IHC staining, including 21 tumours confirmed with molecular testing. TLE1 immunostaining was graded from 0, 1+, 2+, 3+, with 2+ or 3+ grades interpreted as positive staining. Results: Of the 42 tumours, 26 (61.9%) were of monophasic spindle cell type, 13 biphasic type (30.9%), two (4.7%) calcifying type and remaining one (2.3%) was a poorly differentiated synovial sarcoma. On immunohistochemistry (IHC), tumours were positive for epithelial membrane antigen (EMA) (26/34, 76.4%), cytokeratin (CK)7 (6/10, 60%), CK/MNF116 (6/21, 28.6%), B cell lymphoma 2 (BCL2) (36/37, 97.3%), cluster of differentiation molecule 99 (MIC2) (23/31, 74.1%) and transducin-like enhancer of split 1 (TLE1) (40/42, 95.2%), while negative for CD34 in all 21 tumours, wherever performed.
    [Show full text]