12
views
0
recommends
+1 Recommend
0 collections
    0
    shares
      • Record: found
      • Abstract: found
      • Article: found
      Is Open Access

      Establishment and Characterization of a Reliable Xenograft Model of Hodgkin Lymphoma Suitable for the Study of Tumor Origin and the Design of New Therapies

      research-article

      Read this article at

      Bookmark
          There is no author summary for this article yet. Authors can add summaries to their articles on ScienceOpen to make them more accessible to a non-specialist audience.

          Abstract

          To identify the cells responsible for the initiation and maintenance of Hodgkin lymphoma (HL) cells, we have characterized a subpopulation of HL cells grown in vitro and in vivo with the aim of establishing a reliable and robust animal model for HL. To validate our model, we challenged the tumor cells in vivo by injecting the alkylating histone-deacetylase inhibitor, EDO-S101, a salvage regimen for HL patients, into xenografted mice. Methodology: Blood lymphocytes from 50 HL patients and seven HL cell lines were used. Immunohistochemistry, flow cytometry, and cytogenetics analyses were performed. The in vitro and in vivo effects of EDO-S101 were assessed. Results: We have successfully determined conditions for in vitro amplification and characterization of the HL L428-c subline, containing a higher proportion of CD30−/CD15− cells than the parental L428 cell line. This subline displayed excellent clonogenic potential and reliable reproducibility upon xenografting into immunodeficient NOD-SCID-gamma (−/−)(NSG) mice. Using cell sorting, we demonstrate that CD30−/CD15− subpopulations can gain the phenotype of the L428-c cell line in vitro. Moreover, the human cells recovered from the seventh week after injection of L428-c cells into NSG mice were small cells characterized by a high frequency of CD30−/CD15− cells. Cytogenetic analysis demonstrated that they were diploid and showed high telomere instability and telomerase activity. Accordingly, chromosomal instability emerged, as shown by the formation of dicentric chromosomes, ring chromosomes, and breakage/fusion/bridge cycles. Similarly, high telomerase activity and telomere instability were detected in circulating lymphocytes from HL patients. The beneficial effect of the histone-deacetylase inhibitor EDO-S101 as an anti-tumor drug validated our animal model. Conclusion: Our HL animal model requires only 10 3 cells and is characterized by a high survival/toxicity ratio and high reproducibility. Moreover, the cells that engraft in mice are characterized by a high frequency of small CD30−/CD15− cells exhibiting high telomerase activity and telomere dysfunction.

          Related collections

          Most cited references38

          • Record: found
          • Abstract: found
          • Article: not found

          Telomerase maintains telomere structure in normal human cells.

          In normal human cells, telomeres shorten with successive rounds of cell division, and immortalization correlates with stabilization of telomere length. These observations suggest that human cancer cells achieve immortalization in large part through the illegitimate activation of telomerase expression. Here, we demonstrate that the rate-limiting telomerase catalytic subunit hTERT is expressed in cycling primary presenescent human fibroblasts, previously believed to lack hTERT expression and telomerase activity. Disruption of telomerase activity in normal human cells slows cell proliferation, restricts cell lifespan, and alters the maintenance of the 3' single-stranded telomeric overhang without changing the rate of overall telomere shortening. Together, these observations support the view that telomerase and telomere structure are dynamically regulated in normal human cells and that telomere length alone is unlikely to trigger entry into replicative senescence.
            Bookmark
            • Record: found
            • Abstract: found
            • Article: not found

            Circulating clonotypic B cells in classic Hodgkin lymphoma.

            Although Hodgkin and Reed-Sternberg (HRS) cells are B lymphoid cells, they are unlike any normal cells of that lineage. Moreover, the limited proliferative potential of HRS cells belies the clinical aggressiveness of Hodgkin lymphoma (HL). More than 20 years ago, the L428 HL cell line was reported to contain a small population of phenotypic B cells that appeared responsible for the continued generation of HRS cells. This observation, however, has never been corroborated, and such clonotypic B cells have never been documented in HL patients. We found that both the L428 and KM-H2 HL cell lines contained rare B-cell subpopulations responsible for the generation and maintenance of the predominant HRS cell population. The B cells within the HL cell lines expressed immunoglobulin light chain, the memory B-cell antigen CD27, and the stem cell marker aldehyde dehydrogenase (ALDH). Clonal CD27(+)ALDH(high) B cells, sharing immunoglobulin gene rearrangements with lymph node HRS cells, were also detected in the blood of most newly diagnosed HL patients regardless of stage. Although the clinical significance of circulating clonotypic B cells in HL remains unclear, these data suggest they may be the initiating cells for HL.
              Bookmark
              • Record: found
              • Abstract: found
              • Article: not found

              The loss of a single telomere can result in instability of multiple chromosomes in a human tumor cell line.

              Spontaneous telomere loss has been proposed as an important mechanism for initiating the chromosome instability commonly found in cancer cells. We have previously shown that spontaneous telomere loss in a human cancer cell line initiates breakage/fusion/bridge (B/F/B) cycles that continue for many cell generations, resulting in DNA amplification and translocations on the chromosome that lost its telomere. We have now extended these studies to determine the effect of the loss of a single telomere on the stability of other chromosomes. Our study showed that telomere acquisition during B/F/B cycles occurred mainly through translocations involving either the nonreciprocal transfer or duplication of the arms of other chromosomes. Telomere acquisition also occurred through small duplications involving the subtelomeric region of the other end of the same chromosome. Although all of these mechanisms stabilized the chromosome that lost its telomere, they differed in their consequences for the stability of the genome as a whole. Telomere acquisition involving nonreciprocal translocations resulted in the loss of a telomere on the donor chromosome, which consequently underwent additional translocations, isochromosome formation, or complete loss. In contrast, telomere acquisition involving duplications stabilized the genome, although the large duplications created substantial allelic imbalances. Thus, the loss of a single telomere can generate a variety of chromosome alterations commonly associated with human cancer, not only on a chromosome that loses its telomere but also on other chromosomes. Factors promoting telomere loss are therefore likely to have an important role in generating the karyotype evolution associated with human cancer.
                Bookmark

                Author and article information

                Journal
                Cancers (Basel)
                Cancers (Basel)
                cancers
                Cancers
                MDPI
                2072-6694
                31 October 2018
                November 2018
                : 10
                : 11
                : 414
                Affiliations
                [1 ]Radiobiology and Oncology Laboratory, CEA, iRCM, University Paris-Saclay, 92 265 Fontenay aux Roses, France; monika.frenzel@ 123456hotmail.com (M.F.); mustafa.aljawhari@ 123456hotmail.fr (M.A.J.); cuceu_corina@ 123456yahoo.com (C.C.); luc.morat@ 123456cea.fr (L.M.); audelenain@ 123456yahoo.fr (A.L.); williamhempel824@ 123456gmail.com (W.M.H.)
                [2 ]Cell Environment, Oncology Section, 75020 Paris, France
                [3 ]Institute of Biomedicine, University of Aarhus, DK-8000 Aarhus C, Denmark; sjunker@ 123456biomed.au.dk
                [4 ]Platform for Experimental Pathology PathEX/CRC MIRCen/CEA-INSERM, University Paris-Saclay, 92265 Fontenay aux Rroses, France; anne-laure.bauchet@ 123456sanofi.com (A.-L.B.); lev.stimmer@ 123456cea.fr (L.S.)
                [5 ]Platform for Cell Sorting, CEA, iRCM, 92265 Fontenay aux Roses, France; nathalie.dechamps@ 123456cea.fr
                [6 ]Laboratoire d’Imagerie Moléculaire Expérimentale Groupe d’Imagerie du Petit Animal CEA/DSV/I2BM/SHFJ/U1023, University Paris-Saclay, 91400 Orsay, France; geraldine.pottier@ 123456cea.fr (G.P.); raphael.boisgard@ 123456cea.fr (R.B.)
                [7 ]MetaSystems GmbH, Robert-Bosch-Str. 6D, 68804 Altlussheim, Germany; lheidingsfelder@ 123456metasystems.de
                [8 ]Pole Concept, 75016 Paris, France; eric.laplagne@ 123456gmail.com
                [9 ]APHP-Hopital Paul Brousse Université Paris Sud/ESteam Paris Inserm UMR 935, 94800 Villejuif, France; claire.borie@ 123456aphp.fr (C.B.); noufissa.oudrhiri@ 123456aphp.fr (N.O.); dimajouni@ 123456yahoo.fr (D.J.); annelise.bennaceur@ 123456pbr.aphp.fr (A.B.-G.)
                [10 ]IRIMAS, Institut de Recherche en Informatique, Mathématiques, Automatique et Signal, Université de Haute-Alsace, 68093 Mulhouse, France; bruno.colicchio@ 123456uha.fr (B.C.); alain.dieterlen@ 123456uha.fr (A.D.)
                [11 ]Department of Radiation Oncology, Gustave Roussy Cancer Campus, University Paris-Saclay, 94805 Villejuif, France; theogirinsky@ 123456me.com (T.G.); Jean.Bourhis@ 123456unil.ch (J.B.)
                [12 ]Departement of Anapathology, Gustave Roussy Cancer Campus, University Paris-Saclay, 94805 Vilejuif, France; jacques.bosq@ 123456gustaveroussy.fr
                [13 ]Mundipharma-EDO GmbH, CH-4020 Basel, Switzerland; thomas.mehrling@ 123456mundipharma-edo.com
                [14 ]Department of Genetic, Groupe Hospitalier de la Région de Mulhouse Sud-Alsace, 68093 Mulhouse, France; jeandidiere@ 123456ghrmsa.fr
                [15 ]Department of Medicine, Gustave Roussy Cancer Campus, University Paris-Saclay, 94805 Villejuif, France; dr.pcarde@ 123456gmail.com
                Author notes
                [†]

                These authors contributed equally to this work.

                Author information
                https://orcid.org/0000-0003-0639-4760
                https://orcid.org/0000-0002-3557-4499
                https://orcid.org/0000-0002-2757-0770
                Article
                cancers-10-00414
                10.3390/cancers10110414
                6265845
                30384446
                030cd787-a610-4b4c-88cb-837076ab967d
                © 2018 by the authors.

                Licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license ( http://creativecommons.org/licenses/by/4.0/).

                History
                : 05 July 2018
                : 26 October 2018
                Categories
                Article

                hodgkin lymphoma,animal model,cd30−/cd15−,telomere dysfunction,telomerase,edo-s101

                Comments

                Comment on this article