Thymus Regeneration and Future Challenges

Valentin P Shichkin, Mariastefania Antica, Valentin P Shichkin, Mariastefania Antica

Abstract

Thymus regenerative therapy implementation is severely obstructed by the limited number and expansion capacity in vitro of tissue-specific thymic epithelial stem cells (TESC). Current solutions are mostly based on growth factors that can drive differentiation of pluripotent stem cells toward tissue-specific TESC. Target-specific small chemical compounds represent an alternative solution that could induce and support the clonal expansion of TESC and reversibly block their differentiation into mature cells. These compounds could be used both in the composition of culture media designed for TESC expansion in vitro, and in drugs development for thymic regeneration in vivo. It should allow reaching the ultimate objective - autologous thymic tissue regeneration in paediatric patients who had their thymus removed in the course of cardiac surgery.

Keywords: Small chemical compounds; Stem cells; Thymectomised patients; Thymic epithelial stem cells; Thymus; Thymus regeneration.

Figures

Fig. 1.
Fig. 1.
Human thymus cell architecture. The human thymus is located in the upper anterior part of the chest behind the sternum between lungs and lies on top of the heart along the trachea. The thymus reaches its maximum weight (about 28 gram) during puberty. This pinkish-gray organ consists of two lobes parted into lobules by connective tissue strands (trabeculae). Each thymic lobule has a cortex and medulla. Hematopoietic precursor cells (HPC) enters the thymus through postcapillary venules located at the corticomedullary junction (CMJ) and migrate to the capsule, committed CD4-CD8- T precursor cells (TPC) located in the subcapsular region, and immature CD4+CD8+ cortical thymocytes migrate through the cortex and CMJ to the medullar zone. The medulla contains CD4+ and CD8+ naïve thymocytes that will migrate to the periphery. The stromal-epithelial compartment of the thymus is represented by minor populations of EpCam+(CD326+)Foxn1+ bipotent thymic epithelial precursor cells/thymic epithelial stem cells (TEPC/TESC) and mesenchymal stem cells (MSC) located probably in the thymic parenchyma close to the CMJ region, as well as EpCam+CD205+ cortical thymic epithelial cells (cTEC) located in the cortex and EpCam+Air+ medullary thymic epithelial cells (mTEC) located in the medulla. Moreover, the cortex and the medulla contain also macrophages, fibroblasts and dendritic cells (DC) that together with cTEC and mTEC participate in the differentiation, maturation, positive and negative selection of thymocytes. HPC generate all thymocyte populations and alternatively may generate macrophages and DC; TEPC/TESC generate cTEC and mTEC lineages depending on local microenvironment and cross-talk with cortical or medullary thymocytes; MSC generate thymic fibroblasts and adipocytes. BV: Blood vessel; DT: Dead thymocyte; HC: Hassall’s corpuscle.
Fig. 2.
Fig. 2.
TESC/SCC-based strategy for thymus regenerative therapy in partially thymectomized infants. Thymic epithelial stem cell/Small chemical compound (TESC/SCC)-based strategy for autologous thymus regenerative therapy in infants could include the development of clinical grade protocols for collection, preparation and cryopreservation of primary infant thymic tissue and TESCenriched samples. These TESC could be used further to screen SCC for regulation, differentiation and proliferation of human TESC. The selected compounds would be tested for clonal expansion of TESC in vitro and for the reconstitution of thymic function in vivo in terms of maturation, differentiation and tolerance of autologous T cells as well as for supporting thymus tissue growth. Finally, full pharmacological evaluation of the properly selected and optimised compounds would be performed for high efficacy and low toxicity and further drug development. An actual challenge is the optimization of thymectomy procedure in infants to preserve a thymic fragment for consequent postsurgical thymus regenerative therapy. An additional impact on the efficacy of the post-surgical rehabilitation may provide the quality life monitoring of thymectomized patients in relation to their resistance to infections, allergies, autoimmune, oncological and other diseases associated with the impaired thymic function.

References

    1. Miller JFAP. Immunological function of the thymus. The Lancet. 1961;278:748–749.
    1. Prelog M, Keller M, Geiger R, Brandstätter A, Würzner R, Schweigmann U, Zlamy M, Zimmerhackl LB, Grubeck-Loebenstein B. Thymectomy in early childhood: Significant alterations of the CD4+CD45RA+CD62L+ T cell compartment in later life. Clinical Immunology. 2009;130:123–132.
    1. Afifi A, Raja SG, Pennington DJ, Tsang VT. For neonates undergoing cardiac surgery does thymectomy as opposed to thymic preservation have any adverse immunological consequences? Interactive Cardiovascular and Thoracic Surgery. 2010;11:287–291.
    1. Kurobe H, Tominaga T, Sugano M, Hayabuchi Y, Egawa Y, Takahama Y, Kitagawa T. Complete but not partial thymectomy in early infancy reduces T-cell–mediated immune response: Three-year tracing study after pediatric cardiac surgery. The Journal of Thoracic and Cardiovascular Surgery. 2013;145:656–662.e652.
    1. van den Broek T, Delemarre EM, Janssen WJM, Nievelstein RAJ, Broen JC, Tesselaar K, Borghans JAM, Nieuwenhuis EES, Prakken BJ, Mokry M, et al. Neonatal thymectomy reveals differentiation and plasticity within human naive T cells. The Journal of Clinical Investigation. 2016;126:1126–1136.
    1. Stosio M, Ruszkowski J, Mikosik-Roczynska A, Haponiuk I, Witkowski JM. The significance of neonatal thymectomy for shaping the immune system in children with congenital heart defects. Kardiochir Torakochirurgia Pol. 2017;14:258–262.
    1. Gudmundsdottir J, Söderling J, Berggren H, Óskarsdóttir S, Neovius M, Stephansson O, Ekwall O. Long-term clinical effects of early thymectomy: Associations with autoimmune diseases, cancer, infections, and atopic diseases. Journal of Allergy and Clinical Immunology. 2018;141:2294–2297.
    1. Roosen J, Oosterlinck W, Meyns B. Routine thymectomy in congenital cardiac surgery changes adaptive immunity without clinical relevance. Interactive Cardiovascular and Thoracic Surgery. 2014;20:101–106.
    1. Martín-Gayo E, Sierra-Filardi E, Corbí AL, Toribio ML. Plasmacytoid dendritic cells resident in human thymus drive natural Treg cell development. Blood. 2010;115:5366–5375.
    1. Manley, N. R., Richie, E. R., Blackburn, C. C., Condie, B. G., & Sage, J. (2011). Structure and function of the thymic microenvironment. Frontiers in bioscience (Landmark edition). 10.2741/3866.
    1. Stoeckle, C., Rota, I.A., Tolosa, E., Haller, C., Melms, A. and Adamopoulou, E. (2013) Isolation of myeloid dendritic cells and epithelial cells from human thymus. Journal of visualized experiments : JoVE, e50951-e50951.
    1. Martín-Gayo E, González-García S, García-León MJ, Murcia-Ceballos A, Alcain J, García-Peydró M, Allende L, de Andrés B, Gaspar ML, Toribio ML. Spatially restricted JAG1-notch signaling in human thymus provides suitable DC developmental niches. The Journal of Experimental Medicine. 2017;214:3361–3379.
    1. García-León MJ, Fuentes P, de la Pompa JL, Toribio ML. Dynamic regulation of NOTCH1 activation and NOTCH ligand expression in human thymus development. Development. 2018;145:dev165597.
    1. Matsumoto, M., Rodrigues, P., Sousa, L., Tsuneyama, K., Matsumoto, M. and Alves, N. (2019), The ins and outs of Thymic epithelial cell differentiation and function. From: Thymus Transcriptome and cell biology, 10.1007/978-3-030-12040-5_3 pp. 35-65.
    1. Ucar A, Ucar O, Klug P, Matt S, Brunk F, Hofmann TG, Kyewski B. Adult thymus contains FoxN1(−) epithelial stem cells that are bipotent for medullary and cortical thymic epithelial lineages. Immunity. 2014;41:257–269.
    1. Bredenkamp N, Jin X, Liu D, O'Neill KE, Manley NR, Blackburn CC. Construction of a functional thymic microenvironment from pluripotent stem cells for the induction of central tolerance. Regen Med. 2015;10:317–329.
    1. Siepe M, Thomsen AR, Duerkopp N, Krause U, Forster K, Hezel P, Beyersdorf F, Schlensak C, Sudkamp NP, Bosse R, et al. Human neonatal thymus-derived mesenchymal stromal cells: Characterization, differentiation, and immunomodulatory properties. Tissue Engineering. Part A. 2009;15:1787–1796.
    1. Iacobazzi D, Swim MM, Albertario A, Caputo M, Ghorbel MT. Thymus-derived Mesenchymal stem cells for tissue engineering clinical-grade cardiovascular grafts. Tissue Engineering. Part A. 2018;24:794–808.
    1. Shichkin V. Properties of intrathymic T-lymphocyte precursors--targets of thymocyte growth factor (THGF) Biomedical Science. 1990;1:279–287.
    1. Wu L, Antica M, Johnson GR, Scollay R, Shortman K. Developmental potential of the earliest precursor cells from the adult mouse thymus. The Journal of Experimental Medicine. 1991;174:1617–1627.
    1. Antica M, Wu L, Shortman K, Scollay R. Intrathymic lymphoid precursor cells during fetal thymus development. Journal of Immunology. 1993;151:5887–5895.
    1. Márquez, C., Trigueros, C.s., Franco, J. M., Ramiro, A. R., Carrasco, Y. R., López-Botet, M., & Toribio, M. L. (1998). Identification of a common developmental pathway for Thymic natural killer cells and dendritic cells. Blood, 91, 2760–2771.
    1. Weerkamp F, Baert MRM, Brugman MH, Dik WA, de Haas EFE, Visser TP, de Groot CJM, Wagemaker G, van Dongen JJM, Staal FJT. Human thymus contains multipotent progenitors with T/B lymphoid, myeloid, and erythroid lineage potential. Blood. 2006;107:3131–3137.
    1. Tang Y, Yang YG, Bai O, Xia J, Hu Z. Long-term survival and differentiation of human thymocytes in human thymus-grafted immunodeficient mice. Immunotherapy. 2019;11:881–888.
    1. Lepletier A, Chidgey AP, Savino W. Perspectives for improvement of the Thymic microenvironment through manipulation of Thymic epithelial cells: A mini-review. Gerontology. 2015;61:504–514.
    1. O'Neill, K.E., Bredenkamp, N., Tischner, C., Vaidya, H.J., Stenhouse, F.H., Peddie, C.D., Nowell, C.S., Gaskell, T. and Blackburn, C.C. (2016) Foxn1 is dynamically regulated in Thymic epithelial cells during embryogenesis and at the onset of Thymic involution. PLoS One, 11.
    1. Koch U, Fiorini E, Benedito R, Besseyrias V, Schuster-Gossler K, Pierres M, Manley NR, Duarte A, Macdonald HR, Radtke F. Delta-like 4 is the essential, nonredundant ligand for Notch1 during thymic T cell lineage commitment. The Journal of Experimental Medicine. 2008;205:2515–2523.
    1. Shortman K. Cellular aspects of early T-cell development. Curr Opin Immunol. 1992;4:140–146.
    1. Anderson G, Takahama Y. Thymic epithelial cells: Working class heroes for T cell development and repertoire selection. Trends in Immunology. 2012;33:256–263.
    1. Lei Y, Ripen AM, Ishimaru N, Ohigashi I, Nagasawa T, Jeker LT, Bosl MR, Hollander GA, Hayashi Y, Malefyt Rde W, et al. Aire-dependent production of XCL1 mediates medullary accumulation of thymic dendritic cells and contributes to regulatory T cell development. The Journal of Experimental Medicine. 2011;208:383–394.
    1. Proietto AI, van Dommelen S, Zhou P, Rizzitelli A, D'Amico A, Steptoe RJ, Naik SH, Lahoud MH, Liu Y, Zheng P, et al. Dendritic cells in the thymus contribute to T-regulatory cell induction. Proceedings of the National Academy of Sciences of the United States of America. 2008;105:19869–19874.
    1. Shichkin VP. Radioresistant cells of thymus - producers and targets of thymocyte growth factor and their possible role in postradiation restoration of thymus. Immunology Letters. 1992;33:247–254.
    1. Këpuska Z, Sempowski G. Mechanisms of thymic recovery and T cell reconstitution following sublethal ionizing radiation (104.21) The Journal of Immunology. 2011;186:104.121–104.121.
    1. Dudakov JA, Hanash AM, Jenq RR, Young LF, Ghosh A, Singer NV, West ML, Smith OM, Holland AM, Tsai JJ, Boyd RL, van den Brink M. Interleukin-22 drives endogenous thymic regeneration in mice. Science. 2012;336:91–95.
    1. Kadish JL, Basch RS. Thymic regeneration after lethal irradiation evidence for an intra-thymic radioresistant T cell precursor. Journal of Immunology. 1975;114:452–458.
    1. Rossi SW, Jenkinson WE, Anderson G, Jenkinson EJ. Clonal analysis reveals a common progenitor for thymic cortical and medullary epithelium. Nature. 2006;441:988–991.
    1. Wong K, Lister NL, Barsanti M, Lim JMC, Hammett MV, Khong DM, Siatskas C, Gray DHD, Boyd RL, Chidgey AP. Multilineage potential and self-renewal define an epithelial progenitor cell population in the adult Thymus. Cell Reports. 2014;8:1198–1209.
    1. Ulyanchenko S, O'Neill KE, Medley T, Farley AM, Vaidya HJ, Cook AM, Blair NF, Blackburn CC. Identification of a Bipotent epithelial progenitor population in the adult Thymus. Cell Rep. 2016;14:2819–2832.
    1. Shichkin V, Gorbach O, Zuieva O, Martsenyuk O. Optimization of quality parameters for human thymic cell samples stored in liquid nitrogen. Trends in Transplantation. 2018;10:1–11.
    1. Sheridan JM, Keown A, Policheni A, Roesley SNA, Rivlin N, Kadouri N, Ritchie ME, Jain R, Abramson J, Heng TSP, et al. Thymospheres are formed by Mesenchymal cells with the potential to generate adipocytes, but not epithelial cells. Cell Rep. 2017;21:934–942.
    1. Markert ML, Devlin BH, Chinn IK, McCarthy EA, Li YJ. Factors affecting success of thymus transplantation for complete DiGeorge anomaly. American Journal of Transplantation. 2008;8:1729–1736.
    1. Davies EG, Cheung M, Gilmour K, Maimaris J, Curry J, Furmanski A, Sebire N, Halliday N, Mengrelis K, Adams S, et al. Thymus transplantation for complete DiGeorge syndrome: European experience. The Journal of Allergy and Clinical Immunology. 2017;140:1660–1670.
    1. Parent AV, Russ HA, Khan IS, LaFlam TN, Metzger TC, Anderson MS, Hebrok M. Generation of functional thymic epithelium from human embryonic stem cells that supports host T cell development. Cell Stem Cell. 2013;13:219–229.
    1. Sun X, Xu J, Lu H, Liu W, Miao Z, Sui X, Liu H, Su L, Du W, He Q, et al. Directed differentiation of human embryonic stem cells into thymic epithelial progenitor-like cells reconstitutes the thymic microenvironment in vivo. Cell Stem Cell. 2013;13:230–236.
    1. Bredenkamp N, Ulyanchenko S, O'Neill KE, Manley NR, Vaidya HJ, Blackburn CC. An organized and functional thymus generated from FOXN1-reprogrammed fibroblasts. Nature Cell Biology. 2014;16:902–908.
    1. Deng, Z., Liu, H., Rui, J. and Liu, X. (2016) Reconstituted Thymus organ culture. Methods Mol Biol, 2809-2805_2813.
    1. Tajima A, Pradhan I, Trucco M, Fan Y. Restoration of Thymus function with bioengineered Thymus Organoids. Current stem cell reports. 2016;2:128–139.
    1. Wertheimer T, Velardi E, Tsai J, Cooper K, Xiao S, Kloss CC, Ottmüller KJ, Mokhtari Z, Brede C, de Roos P, et al. Production of BMP4 by endothelial cells is crucial for endogenous thymic regeneration. Science immunology. 2018;3:eaal2736.
    1. Tuckett AZ, Thornton RH, Shono Y, Smith OM, Levy ER, Kreines FM, van den Brink MR, Zakrzewski JL. Image-guided intrathymic injection of multipotent stem cells supports lifelong T-cell immunity and facilitates targeted immunotherapy. Blood. 2014;123:2797–2805.
    1. Velardi E, Tsai JJ, Radtke S, Cooper K, Argyropoulos KV, Jae-Hung S, Young LF, Lazrak A, Smith OM, Lieberman S, Kreines F, Shono Y, Wertheimer T, Jenq RR, Hanash AM, Narayan P, Lei Z, Moore MA, Kiem HP, van den Brink M, Dudakov JA. Suppression of luteinizing hormone enhances HSC recovery after hematopoietic injury. Nature Medicine. 2018;24:239–246.
    1. Calderon L, Boehm T. Synergistic, context-dependent, and hierarchical functions of epithelial components in thymic microenvironments. Cell. 2012;149:159–172.
    1. Bonfanti P, Claudinot S, Amici AW, Farley A, Blackburn CC, Barrandon Y. Microenvironmental reprogramming of thymic epithelial cells to skin multipotent stem cells. Nature. 2010;466:978–982.
    1. Hynds RE, Bonfanti P, Janes SM. Regenerating human epithelia with cultured stem cells: Feeder cells, organoids and beyond. EMBO Molecular Medicine. 2018;10:139–150.
    1. Rama P, Matuska S, Paganoni G, Spinelli A, De Luca M, Pellegrini G. Limbal stem-cell therapy and long-term corneal regeneration. New England Journal of Medicine. 2010;363:147–155.
    1. Sato T, van Es JH, Snippert HJ, Stange DE, Vries RG, van den Born M, Barker N, Shroyer NF, van de Wetering M, Clevers H. Paneth cells constitute the niche for Lgr5 stem cells in intestinal crypts. Nature. 2011;469:415–418.
    1. Conti L, Pollard SM, Gorba T, Reitano E, Toselli M, Biella G, Sun Y, Sanzone S, Ying QL, Cattaneo E, et al. Niche-independent symmetrical self-renewal of a mammalian tissue stem cell. PLoS Biology. 2005;3:16.
    1. Yasuda S-Y, Ikeda T, Shahsavarani H, Yoshida N, Nayer B, Hino M, Vartak-Sharma N, Suemori H, Hasegawa K. Chemically defined and growth-factor-free culture system for the expansion and derivation of human pluripotent stem cells. Nature Biomedical Engineering. 2018;2:173–182.
    1. Brizzi MF, Tarone G, Defilippi P. Extracellular matrix, integrins, and growth factors as tailors of the stem cell niche. Current Opinion in Cell Biology. 2012;24:645–651.
    1. Chen S, Lewallen M, Xie T. Adhesion in the stem cell niche: Biological roles and regulation. Development. 2013;140:255–265.
    1. Simons BD, Clevers H. Strategies for homeostatic stem cell self-renewal in adult tissues. Cell. 2011;145:851–862.
    1. Ying Q-L, Wray J, Nichols J, Batlle-Morera L, Doble B, Woodgett J, Cohen P, Smith A. The ground state of embryonic stem cell self-renewal. Nature. 2008;453:519–523.
    1. Li VC, Kirschner MW. Molecular ties between the cell cycle and differentiation in embryonic stem cells. Proceedings of the National Academy of Sciences of the United States of America. 2014;111:9503–9508.
    1. Screpanti I, Meco D, Scarpa S, Morrone S, Frati L, Gulino A, Modesti A. Neuromodulatory loop mediated by nerve growth factor and interleukin 6 in thymic stromal cell cultures. Proceedings of the National Academy of Sciences of the United States of America. 1992;89:3209–3212.
    1. Fernandez E, Vicente A, Zapata A, Brera B, Lozano JJ, Martinez C, Toribio ML. Establishment and characterization of cloned human thymic epithelial cell lines. Analysis of adhesion molecule expression and cytokine production. Blood. 1994;83:3245–3254.
    1. Shichkin VP, Gorbach OI, Zuieva OA, Grabchenko NI, Aksyonova IA, Todurov BM. Effect of cryopreservation on viability and growth efficiency of stromal-epithelial cells derived from neonatal human thymus. Cryobiology. 2017;78:70–79.
    1. Villegas JA, Gradolatto A, Truffault F, Roussin R, Berrih-Aknin S, Le Panse R, Dragin N. Cultured human Thymic-derived cells display medullary Thymic epithelial cell phenotype and functionality. Frontiers in Immunology. 2018;9:1663–1663.
    1. Burdick JA, Vunjak-Novakovic G. Engineered microenvironments for controlled stem cell differentiation. Tissue Engineering. Part A. 2009;15:205–219.
    1. Liu K, Yu C, Xie M, Li K, Ding S. Chemical modulation of cell fate in stem cell therapeutics and regenerative medicine. Cell Chemical Biology. 2016;23:893–916.
    1. •• Clarke, K., Christie VB, Whiting A and SA, P. (2018) Using small molecules to control stem cell growth and differentiation. Tocris Scientific Review Series 1-16, . Provides justification for searching for small chemical compounds that may control the clonal expansion of human thymic epithelial stem cells.
    1. Xu Y, Zhu X, Hahm HS, Wei W, Hao E, Hayek A, Ding S. Revealing a core signaling regulatory mechanism for pluripotent stem cell survival and self-renewal by small molecules. Proceedings of the National Academy of Sciences of the United States of America. 2010;107:8129–8134.
    1. Motamedi YK, Peymani M, Baharvand H, Nasr-Esfahani MH, Bender A. Systematic selection of small molecules to promote differentiation of embryonic stem cells and experimental validation for generating cardiomyocytes. Cell Death Discovery. 2016;2:16007.
    1. Kell DB, Goodacre R. Metabolomics and systems pharmacology: Why and how to model the human metabolic network for drug discovery. Drug Discovery Today. 2014;19:171–182.
    1. Wang Y, Xing J, Xu Y, Zhou N, Peng J, Xiong Z, Liu X, Luo X, Luo C, Chen K, Zheng M, Jiang H. In silico ADME/T modelling for rational drug design. Quarterly Reviews of Biophysics. 2015;48:488–515.
    1. Yan X, Liao C, Liu Z, Hagler AT, Gu Q, Xu J. Chemical structure similarity search for ligand-based virtual screening: Methods and computational resources. Current Drug Targets. 2016;17:1580–1585.
    1. Zhou W, Wang Y, Lu A, Zhang G. Systems pharmacology in small molecular drug discovery. International Journal of Molecular Sciences. 2016;17:246–246.
    1. Al-Awadhi FH, Salvador LA, Luesch H. Screening strategies for drug discovery and target identification. Mar. Biomed. Beach Bedside. 2015;1:135–166.
    1. Li, P., Fu, Y. and Wang, Y. (2015) Network based Approach to Drug Discovery: A Mini Review. Mini reviews in medicinal chemistry, 15.
    1. Conklin, J.F., Baker, J. and Sage, J. (2012) The RB family is required for the self-renewal and survival of human embryonic stem cells. Nat Commun, 3.
    1. Garfin PM, Min D, Bryson JL, Serwold T, Edris B, Blackburn CC, Richie ER, Weinberg KI, Manley NR, Sage J, Viatour P. Inactivation of the RB family prevents thymus involution and promotes thymic function by direct control of Foxn1 expression. The Journal of Experimental Medicine. 2013;210:1087–1097.
    1. Vaidya HJ, Briones Leon A, Blackburn CC. FOXN1 in thymus organogenesis and development. European Journal of Immunology. 2016;46:1826–1837.
    1. Choudhry Z, Rikani AA, Choudhry AM, Tariq S, Zakaria F, Asghar MW, Sarfraz MK, Haider K, Shafiq AA, Mobassarah NJ. Sonic hedgehog signalling pathway: a complex network. Annals of Neurosciences. 2014;21:28–31.
    1. Yamamoto S, Ikeda H, Toyama D, Hayashi M, Akiyama K, Suzuki M, Tanaka Y, Watanabe T, Fujimoto Y, Hosaki I, Nishihira H, Isoyama K. Quality of long-term cryopreserved umbilical cord blood units for hematopoietic cell transplantation. International Journal of Hematology. 2011;93:99–105.
    1. Badowski M, Muise A, Harris DT. Mixed effects of long-term frozen storage on cord tissue stem cells. Cytotherapy. 2014;16:1313–1321.
    1. Marquez-Curtis LA, Janowska-Wieczorek A, McGann LE, Elliott JA. Mesenchymal stromal cells derived from various tissues: Biological, clinical and cryopreservation aspects. Cryobiology. 2015;71:181–197.
    1. Harris D. Long-term frozen storage of stem cells: Challenges and solutions. Journal of Biorepository Science for Applied Medicine. 2016;4:9–20.
    1. Kulikov AV, Arkhipova LV, Smirnova GN, Novoselova EG, Shpurova NA, Shishova NV, Sukhikh GT. Slowing down the rate of irreversible age-related atrophy of the thymus gland by atopic autotransplantation of its tissue, subjected to long-term cryoconservation. Advances in Gerontology. 2010;23:76–80.
    1. Brown ME, Zhou Y, McIntosh BE, Norman IG, Lou HE, Biermann M, Sullivan JA, Kamp TJ, Thomson JA, Anagnostopoulos PV, et al. A humanized mouse model generated using surplus neonatal tissue. Stem Cell Reports. 2018;10:1175–1183.
    1. Watanabe K, Ueno M, Kamiya D, Nishiyama A, Matsumura M, Wataya T, Takahashi JB, Nishikawa S, Muguruma K, Sasai Y. A ROCK inhibitor permits survival of dissociated human embryonic stem cells. Nature Biotechnology. 2007;25:681–686.
    1. Tannenbaum C, Day D. Age and sex in drug development and testing for adults. Pharmacological Research. 2017;121:83–93.
    1. Ritz SA, Antle DM, Cote J, Deroy K, Fraleigh N, Messing K, Parent L, St-Pierre J, Vaillancourt C, Mergler D. First steps for integrating sex and gender considerations into basic experimental biomedical research. The FASEB Journal. 2014;28:4–13.
    1. Clayton JA. Studying both sexes: a guiding principle for biomedicine. FASEB Journal : Official Publication of the Federation of American Societies for Experimental Biology. 2016;30:519–524.
    1. Tannenbaum C, Schwarz JM, Clayton JA, de Vries GJ, Sullivan C. Evaluating sex as a biological variable in preclinical research: The devil in the details. Biology of Sex Differences. 2016;7:13–13.
    1. Cornelison TL, Clayton JA. Considering sex as a biological variable in biomedical research. Gender and the Genome. 2017;1:89–93.
    1. Shah K, McCormack CE, Bradbury NA. Do you know the sex of your cells? American Journal of Physiology. Cell Physiology. 2014;306:6.
    1. US Food and Drug Administration. .
    1. European Medicines Agency.
    1. Campbell LD, Betsou F, Garcia DL, Giri JG, Pitt KE, Pugh RS, Sexton KC, Skubitz AP, Somiari SB. Development of the ISBER best practices for repositories: Collection, storage, retrieval and distribution of biological materials for research. Biopreservation and Biobanking. 2012;10:232–233.
    1. Final Report Summary - THYMISTEM Development of Stem Cell Based Therapy for Thymic Regeneration, (2018) .

Source: PubMed

3
購読する