Дисертації з теми "Bone marrow mesenchymal stromal cell"
Оформте джерело за APA, MLA, Chicago, Harvard та іншими стилями
Ознайомтеся з топ-50 дисертацій для дослідження на тему "Bone marrow mesenchymal stromal cell".
Біля кожної праці в переліку літератури доступна кнопка «Додати до бібліографії». Скористайтеся нею – і ми автоматично оформимо бібліографічне посилання на обрану працю в потрібному вам стилі цитування: APA, MLA, «Гарвард», «Чикаго», «Ванкувер» тощо.
Також ви можете завантажити повний текст наукової публікації у форматі «.pdf» та прочитати онлайн анотацію до роботи, якщо відповідні параметри наявні в метаданих.
Переглядайте дисертації для різних дисциплін та оформлюйте правильно вашу бібліографію.
François, Moïra. "Comprehensive study of the immunomodulatory properties of bone marrow-derived mesenchymal stromal cells." Thesis, McGill University, 2011. http://digitool.Library.McGill.CA:80/R/?func=dbin-jump-full&object_id=103683.
Повний текст джерелаAu cours de la dernière décennie, les cellules stromales mésenchymateuses (MSC) ont fait une entrée remarquée dans le domaine de l'immunothérapie cellulaire. In vitro, les MSC ont démontrées des propriétés immunomodulatrices, soit par leur action inhibitrice sur les fonctions des cellules du système immunitaire ou par leur capacité à présenter des antigènes aux lymphocytes T CD4+, à la suite d'une stimulation par IFN-. Malgré l'existence de nombreuses recherches in vivo chez les animaux et l'homme prouvant leurs propriétés immunologiques, les mécanismes par lesquels les MSC modulent le système immunitaire n'ont pas encore été élucidés. Dans le Chapitre 1, j'ai présenté une revue succincte de la littérature traitant des caractéristiques des MSC. Dans le Chapitre 2, j'ai adressé les mécanismes immunosuppressifs des MSC humaines sur les lymphocytes T. À l'aide d'un test de prolifération in vitro, j'ai démontré que les MSC humaines suppriment la prolifération des lymphocytes T par grâce à l'expression indoleamine 2,3-dioxygenase (IDO) induite par l'exposition à l'IFN-. De plus, les MSC isolées de différents donneurs inhibent la prolifération des lymphocytes T à différents degrés qui correspondent au le niveau d'expression d'IDO par chaque donneur. L'utilisation de cellules mononucléaires sanguines (PBMC) complet comparativement à l'utilisation de lymphocytes T purifiés a révélé le rôle joué par les monocytes dans la suppression de la prolifération des lymphocytes T par les MSC. L'activité enzymatique d'IDO en combinaison avec d'autres facteurs sécrétés par les MSC induisent la différentiation des monocytes en macrophages immunosuppressifs de type M2. En plus de déclencher les mécanismes immunosuppressifs des MSC, l'IFN-a aussi eu pour effet d'induire des propriétés typiques des cellules présentatrices d'antigène (CPA) chez les MSC. Dans le Chapitre 3, j'ai étudié les mécanismes moléculaires impliqués dans la modulation de l'expression des molécules MHC de type II et la présentation d'antigène par celles-ci dans les MSC. J'ai démontré que l'IFN- active la transcription du transactivateur de classe II (CIITA), ce qui a eu pour résultat d'uprégulation les molécules MHC de type II dans les MSC murines et humaines, et que l'ajout de TGF- contrecarre l'effet de l'IFN- en inhibant la transcription de CIITA. De plus, la densité cellulaire des MSC en culture module la présentation d'antigène en affectant l'expression des molécules MHC de type II différemment chez les MSC murines et humaines. Dans le Chapitre 4, j'ai examiné la capacité des MSC de souris à cross-présenter des antigènes exogènes, une autre propriété typique des CPA. J'ai démontré que l'IFN- induit la cross-présentation dans les MSC murines et que celle-ci dépend des molécules TAP et du protéasome. J'ai aussi prouvé à l'aide d'un modèle de reconstitution immunitaire in vivo, que les MSC murines peuvent induire l'activation des lymphocytes T CD8+ contre un antigène spécifique. Finalement, j'ai enquêté dans le Chapitre 5, l'impact immunologique de l'expression et de la signalisation par les TLR chez les MSC humaines et murines. J'ai illustré que l'activation des TLR induisait l'expression de chemokines et de cytokines par les MSC créant ainsi un milieu inflammatoire propice au recrutement des cellules immunitaires. J'ai conclue en démontrant que les MSC différaient des CPA classiques par l'absence de production IL-12p70, une cytokine essentielle à la réponse immunitaire innée et acquise, en réponse à la stimulation des TLR. Les résultats inclus dans cette thèse illustrent la complexité des mécanismes immunomodulatoires des MSC. Leurs réponses face aux signaux de leur environnement, tel que l'inflammation ou l'infection activent soit leurs propriétés immunosuppressives ou –stimulatrices dépendamment de la situation. Mes découvertes pourront optimiser l'utilisation des MSC dans le domaine de l'immunothérapie cellulaire.
Lenz, Daniel. "Dissecting the heterogeneity of murine mesenchymal bone marrow stromal cells." Doctoral thesis, Humboldt-Universität zu Berlin, 2020. http://dx.doi.org/10.18452/21017.
Повний текст джерелаBone marrow stromal cells receive increasing amounts of attention lately. They have been shown to support survival of hematopoietic stem cells as well as memory lymphocytes which is of great importance when targeting the perseverance of autoimmune diseases. CD4+ memory T lymphocytes reside in the proximity of VCAM-1 expressing stromal cells which provide them with survival signals such as Interleukin-7. Herein, a protocol was developed to quantitatively obtain VCAM-1+ and VCAM-1+ IL-7+/- stromal cells via enzymatic/mechanic digestion and cytoskeleton-inhibition. Ex vivo gene expression analysis was performed from sorted, pure cells with good recovery. Candidate genes/markers were validated in (high-throughput) flow cytometry and histological analysis including subsequent semi-automated colocalization was performed. CD1d was found to be good surrogate marker for VCAM-1+PECAM-1- non-endothelial stroma while the population of CD200int/BP-1+/CD73+/CD105- stromal cells is greatly enriched in IL-7 producers which was equally true for the stromal transcription factor Prrx1. CD55, BP-1 and Cadherin-11 were found to be differentially expressed in differing IL-7 reporter mice haplotypes. The reporter mice haplotypes revealed monoallelic expression features of IL-7. All methodologies suggest that VCAM-1+ as well as IL-7+/- stromal cells are heterogeneous by marker expression yet don’t cluster extensively in flow cytometry co-stains. The functional relevance of the marker diversity described in this thesis remains to be tested but insinuates a broad repertoire for bone marrow stroma cells for new interaction pathways with lymphocyte subsets. Ultimately, this knowledge will hopefully feedback to clinical questions of autoimmunity for targeted treatment of stromal niches.
Tsui, Yat-ping, and 徐軼冰. "Derivation of oligodendrocyte precursor cells from adult bone marrow stromal cells." Thesis, The University of Hong Kong (Pokfulam, Hong Kong), 2013. http://hdl.handle.net/10722/197485.
Повний текст джерелаpublished_or_final_version
Biochemistry
Doctoral
Doctor of Philosophy
Yoshioka, Satoshi. "CCAAT/Enhancer-Binding Proteinβ Expressed by Bone Marrow Mesenchymal Stromal Cells Regulates Early B-Cell Lymphopoiesis". Kyoto University, 2014. http://hdl.handle.net/2433/185198.
Повний текст джерелаChandran, Priya. "Bone Marrow Microenvironment in Acute Myleoid Leukemia." Thèse, Université d'Ottawa / University of Ottawa, 2013. http://hdl.handle.net/10393/24301.
Повний текст джерелаLloyd, Brandon R. "Comparison of Bone Marrow Mesenchymal Stem Cells from Limb and Jaw Bones." The Ohio State University, 2016. http://rave.ohiolink.edu/etdc/view?acc_num=osu1458678153.
Повний текст джерелаAnastassiadis, Konstantinos, and Maria Rostovskaya. "Differential Expression of Surface Markers in Mouse Bone Marrow Mesenchymal Stromal Cell Subpopulations with Distinct Lineage Commitment." Saechsische Landesbibliothek- Staats- und Universitaetsbibliothek Dresden, 2016. http://nbn-resolving.de/urn:nbn:de:bsz:14-qucosa-191602.
Повний текст джерелаAnastassiadis, Konstantinos, and Maria Rostovskaya. "Differential Expression of Surface Markers in Mouse Bone Marrow Mesenchymal Stromal Cell Subpopulations with Distinct Lineage Commitment." Public Library of Science, 2012. https://tud.qucosa.de/id/qucosa%3A29135.
Повний текст джерелаEspig, Sandy [Verfasser]. "Isolation and characterization of rat bone-marrow derived mesenchymal stromal cells / Sandy Espig." Ulm : Universität Ulm. Medizinische Fakultät, 2016. http://d-nb.info/1082294284/34.
Повний текст джерелаClough, Sally. "IL7 as a marker of a subset of bone marrow mesenchymal stromal cells." Thesis, University of York, 2013. http://etheses.whiterose.ac.uk/4771/.
Повний текст джерелаMareddy, Shobha R. "Characterization of bone marrow stromal clonal populations derived from osteoarthritis patients." Thesis, Queensland University of Technology, 2008. https://eprints.qut.edu.au/17151/1/Shobha_Reddy_Mareddy_Thesis.pdf.
Повний текст джерелаMareddy, Shobha R. "Characterization of bone marrow stromal clonal populations derived from osteoarthritis patients." Queensland University of Technology, 2008. http://eprints.qut.edu.au/17151/.
Повний текст джерелаVillaggio, Giusy. "Relationship between extracellular matrix (ECM) components and mineralization in bone marrow stromal cells." Doctoral thesis, Università di Catania, 2014. http://hdl.handle.net/10761/1492.
Повний текст джерелаMa, Ming, and 馬明. "Human mesenchymal stromal cells enhance bone marrow metastases of neuroblastoma via SDF-1 related pathways." Thesis, The University of Hong Kong (Pokfulam, Hong Kong), 2010. http://hub.hku.hk/bib/B45790486.
Повний текст джерелаMunshi, Afnan M. N. Alam. "Comprehensive Proteomic Analysis and Characterization of Human Bone Marrow Mesenchymal Stem/Stromal Derived Extracellular Vesicles." Thesis, Université d'Ottawa / University of Ottawa, 2019. http://hdl.handle.net/10393/39538.
Повний текст джерелаSugino, Noriko. "Early osteoinductive human bone marrow mesenchymal stromal/stem cells support an enhanced hematopoietic cell expansion with altered chemotaxis- and adhesion-related gene expression profiles." Kyoto University, 2016. http://hdl.handle.net/2433/215424.
Повний текст джерелаKyoto University (京都大学)
0048
新制・課程博士
博士(医学)
甲第19598号
医博第4105号
新制||医||1014(附属図書館)
32634
京都大学大学院医学研究科医学専攻
(主査)教授 三森 経世, 教授 開 祐司, 教授 妻木 範行
学位規則第4条第1項該当
Carlton, Morgan M. "Proteomic characterisation of rat bone marrow-derived mesenchymal stromal cells cultured in 'stemness' promoting conditions." Thesis, Queensland University of Technology, 2018. https://eprints.qut.edu.au/117288/1/Morgan_Carlton_Thesis.pdf.
Повний текст джерелаBradhurst, Christopher John. "Monitoring mesenchymal stem cell cultures using image processing and pattern recognition techniques." Thesis, Queensland University of Technology, 2010. https://eprints.qut.edu.au/43623/1/Christopher_Bradhurst_Thesis.pdf.
Повний текст джерелаWojtowicz, Abigail M. "Genetically-engineered bone marrow stromal cells and collagen mimetic scaffold modification for healing critically-sized bone defects." Diss., Georgia Institute of Technology, 2009. http://hdl.handle.net/1853/34705.
Повний текст джерелаGoulard, Marie. "The bone marrow microenvironment in myelodysplastic syndromes : functional and molecular study." Thesis, Sorbonne Paris Cité, 2017. http://www.theses.fr/2017USPCC302.
Повний текст джерелаMyelodysplastic syndromes (MDS) are a heterogeneous group of clonal myeloid pathologies characterized by an impaired hematopoiesis. The role of the bone marrow microenvironment (BMM) remains unclear in the natural history of these diseases. Abnormalities of the BMM have been observed in myelodysplasia and a recent published murine model implies that alterations of the BMM could play a role in the trigger/progression of these diseases.Firstly, we tried to develop an in vivo model of MDS in NSG and NSG-S mice. The low rate of engraftment pushed us to develop a 2D co-culture model in vitro. This model is a good alternative to test new therapeutic strategies for MDS patients.In this study, we analysed mesenchymal stromal cells (MSCs) from the bone marrow of pretreated MDS patients in order to identify the functional and molecular abnormalities in those cells of the BMM, central for their interactions with the hematopoietic stem cells (HSCs).MDS MSCs have an impaired clonogenic capacity. We didn’t observed modifications of their differentiation toward osteogenic, adipogenic and chondrogenic pathways and capacity to support of a normal hematopoiesis. MDS MSCs display epigenetic and transcriptomic modifications that could explain the alteration of the relationships between these cells and HSCs observed in imagery in a 3D co-culture model.These results showed that MDS MSCs have functional and molecular abnormalities and that these alterations could impair their relationship with HSCs
Fujii, Sumie. "GVHD amelioration by human bone marrow mesenchymal stromal/stem cell-derived extracellular vesicles is associated with peripheral preservation of naive T cell populations." Kyoto University, 2018. http://hdl.handle.net/2433/232136.
Повний текст джерелаWoolston, Caroline. "The role of bone marrow mesenchymal stromal cells in the protection of B-cell chronic lymphocytic leukaemia from spontaneous and drug induced apoptosis." Thesis, University of Leicester, 2004. http://hdl.handle.net/2381/29873.
Повний текст джерелаRostovskaya, Maria. "Lineage Commitment of Conditionally Immortalized Bone Marrow Mesenchymal Stromal Cells from Tetracycline-Regulated SV40 Large T-antigen Transgenic Mice." Doctoral thesis, Saechsische Landesbibliothek- Staats- und Universitaetsbibliothek Dresden, 2010. http://nbn-resolving.de/urn:nbn:de:bsz:14-qucosa-63435.
Повний текст джерелаMukohira, Hisa. "Mesenchymal stromal cells in bone marrow express adiponectin and are efficiently targeted by an adiponectin promoter-driven Cre transgene." Kyoto University, 2020. http://hdl.handle.net/2433/253155.
Повний текст джерелаFutrega, Katarzyna. "Device and application development for haematopoietic stem and progenitor cell (HSPC) and mesenchymal stromal cell (MSC) 3D spheroid cultures." Thesis, Queensland University of Technology, 2016. https://eprints.qut.edu.au/92605/1/Katarzyna_Futrega_Thesis.pdf.
Повний текст джерелаDing, Ximing [Verfasser], and Judith [Akademischer Betreuer] Zaugg. "Aging of human bone marrow – functional and epigenetic changes in senescent mesenchymal stromal cells / Ximing Ding ; Betreuer: Judith Zaugg." Heidelberg : Universitätsbibliothek Heidelberg, 2018. http://d-nb.info/1177691140/34.
Повний текст джерелаLenz, Daniel [Verfasser], Andreas [Gutachter] Radbruch, Andreas [Gutachter] Thiel, and Enrico [Gutachter] Klotzsch. "Dissecting the heterogeneity of murine mesenchymal bone marrow stromal cells / Daniel Lenz ; Gutachter: Andreas Radbruch, Andreas Thiel, Enrico Klotzsch." Berlin : Humboldt-Universität zu Berlin, 2020. http://d-nb.info/1203623933/34.
Повний текст джерелаPlatzbecker, Uwe, Ruben A. Ferrer, Manja Wobus, Catrin List, Rebekka Wehner, Claudia Schönefeldt, Barbara Brocard, et al. "Mesenchymal stromal cells from patients with myelodyplastic syndrome display distinct functional alterations that are modulated by lenalidomide." Saechsische Landesbibliothek- Staats- und Universitaetsbibliothek Dresden, 2015. http://nbn-resolving.de/urn:nbn:de:bsz:14-qucosa-178822.
Повний текст джерелаKylmäoja, E. (Elina). "Osteoclastogenesis from bone marrow and peripheral blood monocytes:the role of gap junctional communication and mesenchymal stromal cells in the differentiation." Doctoral thesis, Oulun yliopisto, 2018. http://urn.fi/urn:isbn:9789526221045.
Повний текст джерелаTiivistelmä Osteoklastit ovat monitumaisia luuta hajottavia soluja, jotka ovat erilaistuneet monosyyteistä. Monosyyttejä voidaan eristää luuytimestä tai perifeerisestä verestä. Erilaistumisen aikana osteoklastien esiastesolujen sekä muiden luusolujen, kuten mesenkymaalisten stroomasolujen (MSC) välillä tapahtuu monimutkaista signalointia. Aukkoliitoskommunikointi (GJC) on eräs solufuusiossa tapahtuvista mekanismeista. GJC:tä voidaan muunnella useilla aineilla, esimerkiksi spesifisillä stimulaattoreilla, antiarytmisillä peptideillä (AAP). AAP-yhdisteiden vaikutuksia on tutkittu laajalti sydänkudoksessa johtuen niiden lupaavista kliinisistä ominaisuuksista sydänperäisten oireiden hoidossa. Tämän tutkimuksen tarkoituksena oli selvittää GJC:n ja AAP-yhdisteiden roolia luusoluviljelmissä. Lisäksi tutkittiin MSC-solujen osallistumista AAP-yhdisteiden vaikutuksiin sekä vertailtiin kahta erilaista osteoklastogeneesiviljelmää, joissa oli eri määrä MSC-soluja. GJC:tä osteoklastogeneesissä tutkittiin sekä sitä estävillä että stimuloivilla yhdisteillä hiiren monosyyttilinjan RAW 264.7 -soluissa sekä luuytimen hematopoieettisten solujen primääriviljelmissä. Seuraavat tutkimukset tehtiin ihmisen luuytimen ja perifeerisen veren monosyyteillä, ja niissä selvitettiin AAP10-yhdisteen vaikutuksia fysiologisissa sekä happamissa olosuhteissa. Lisäksi vertailtiin luuytimen ja perifeerisen veren monosyyttien osteoklastogeneesiä. In vitro -soluviljelmät tehtiin naudan tai ihmisen luulastujen päällä, ja soluista analysoitiin monitumaisuus, luun resorptio sekä useiden osteoklastimarkkereiden ilmentyminen. Tulokset osoittavat, että GJC:tä hyödynnetään osteoklastogeneesissä, mutta se ei ole korvaamaton mekanismi. GJC:tä voidaan stimuloida AAP-yhdisteillä osteoklastogeneesin aikana, mutta vaikutukset riippuvat viljelyolosuhteista sekä MSC-solujen läsnäolosta. AAP-yhdisteet voivat aktivoida myös MSC-soluja johtaen osteoklastogeneesin epäsuoraan säätelyyn, kun MSC-solut tuottavat useita erilaistumiseen vaikuttavia molekyylejä. Lisäksi perifeerisen veren monosyyteillä havaittiin korkeampi osteoklastogeeninen erilaistumispotentiaali verrattuna luuytimen monosyytteihin. Tulokset voidaan selittää osteoklastogeneesiä säätelevien MSC-solujen läsnäololla luuydinviljelmissä, kun taas perifeerisen veren monosyyttiviljelmissä näitä soluja on vain vähän, jolloin myös niiden säätelyominaisuudet puuttuvat
Brohlin, Maria. "Mesenchymal stem cells for repair of the peripheral and central nervous system." Doctoral thesis, Umeå universitet, Anatomi, 2011. http://urn.kb.se/resolve?urn=urn:nbn:se:umu:diva-47746.
Повний текст джерелаFung, Kwong-lam, and 馮廣林. "The effect of microtubule targeting chemotherapeutic agents on bone marrow derived mesenchymal stromal cells and its interaction withacute lymphoblastic leukemia blasts." Thesis, The University of Hong Kong (Pokfulam, Hong Kong), 2009. http://hub.hku.hk/bib/B43085660.
Повний текст джерелаFung, Kwong-lam. "The effect of microtubule targeting chemotherapeutic agents on bone marrow derived mesenchymal stromal cells and its interaction with acute lymphoblastic leukemia blasts." Click to view the E-thesis via HKUTO, 2009. http://sunzi.lib.hku.hk/hkuto/record/B43085660.
Повний текст джерелаPlatzbecker, Uwe, Ruben A. Ferrer, Manja Wobus, Catrin List, Rebekka Wehner, Claudia Schönefeldt, Barbara Brocard, et al. "Mesenchymal stromal cells from patients with myelodyplastic syndrome display distinct functional alterations that are modulated by lenalidomide." Ferrata Storti Foundation, 2013. https://tud.qucosa.de/id/qucosa%3A28910.
Повний текст джерелаHe, Q. "Peripheral blood derived multi-potent mesenchymal stromal cells (MSCs) in rats : their differentiation and characteristic comparison with bone marrow derived MSCs." Thesis, Queen's University Belfast, 2006. http://ethos.bl.uk/OrderDetails.do?uin=uk.bl.ethos.431597.
Повний текст джерелаBurk, Janina, Claudia Gittel, Sandra Heller, Bastian Pfeiffer, Felicitas Paebst, Annette B. Ahrberg, and Walter Brehm. "Gene expression of tendon markers in mesenchymal stromal cells derived from different sources." Universitätsbibliothek Leipzig, 2014. http://nbn-resolving.de/urn:nbn:de:bsz:15-qucosa-157823.
Повний текст джерелаAzizivarzaneh, Zahra [Verfasser], Kathrin [Akademischer Betreuer] Maedler, and Elke [Akademischer Betreuer] Oetjen. "beta-MSCs: Successful fusion of bone marrow mesenchymal stromal cells with beta-cells results in a beta-cell like phenotype / Zahra Azizivarzaneh. Gutachter: Kathrin Maedler ; Elke Oetjen. Betreuer: Kathrin Maedler." Bremen : Staats- und Universitätsbibliothek Bremen, 2015. http://d-nb.info/1075609410/34.
Повний текст джерелаOhno, Satoshi. "Implantation of an Atelocollagen Sponge With Autologous Bone Marrow-Derived Mesenchymal Stromal Cells for Treatment of Vocal Fold Scarring in a Canine Model." Kyoto University, 2012. http://hdl.handle.net/2433/157415.
Повний текст джерелаBENEFORTI, LINDA. "Role of Bone Marrow-Mesenchymal Stromal Cells and inflammation in the pre-leukemic phase of ETV6-RUNX1-positive childhood Acute Lymphoblastic Leukemia (ALL)." Doctoral thesis, Università degli Studi di Milano-Bicocca, 2019. http://hdl.handle.net/10281/241325.
Повний текст джерелаTranslocation t(12;21) is the most frequent chromosomal rearrangement in pediatric cancers, exclusively leading to B-cell Precursors Acute Lymphoblastic Leukemia (BCP-ALL). Translocation occurs in utero in stem-progenitor cells (HSPC) but it is insufficient for leukemogenesis, since the consequent ETV6-RUNX1 (E/R) fusion gene only generates a silent B-progenitor pre-leukemic clone; additional mutations are thus required for transformation. These latter occur in the post-natal period likely due to dysregulated immune response to common infections/inflammation. Our published data demonstrated that TGFβ, a cytokine produced during inflammation, limited the proliferation of normal pro-B and cells while favoring the insensitive E/R+ clone; moreover, TGFβ selected putative pre-leukemic stem cells (preLSC) in umbilical cord blood (UCB) CD34+ progenitors transduced with the oncogene. On the other hand, we previously showed that ETV6-RUNX1+ murine B-progenitors were altered in adhesion molecules expression and CXCL12-directed migration, suggesting possible dysregulated interactions within the bone marrow (BM) niche. Mesenchymal Stromal Cells (MSC) are key regulators of both HSPC and inflammation in the niche. Importantly, it has been shown that mesenchymal inflammation promotes secondary myeloid leukemia in predisposing syndromes by increasing DNA damage in HSPC, while MSC/BCP-ALL blasts cross-talk profoundly modifies cytokine and chemokine signalling within the niche excluding normal hematopoiesis in favor of leukemia. Taking advantages from two ETV6-RUNX1-expressing cell models (Ba/F3, a murine pro-B cell line, and ETV6-RUNX1-expressing human UCB-CD34+ progenitors) the present PhD study demonstrates that ETV6-RUNX1-expressing cells take advantage from mesenchymal inflammation in terms of migration, persistence and potential progression. In particular, we have found that pre-leukemic Ba/F3 show a peculiar pro-inflammatory gene expression profile characterized by a marked migratory and myeloid signature. Concordantly, ETV6-RUNX1+ Ba/F3 and CD34+ cells preferentially migrate toward inflamed compared to unstimulated BM-MSC supernatants; in case of the first, migration is CXCR2-dependent. Moreover, ETV6-RUNX1+ Ba/F3 are favored compared to controls in presence of BM-MSC and inflammatory cytokines, as they decrease normal cells proliferation and survival while minimally affecting pre-leukemic cells. The effect is mediated by soluble factors, but neither TGFβ nor CXCR2 axis are implicated. Importantly, the inflamed mesenchymal niche increases genotoxic stress in both control and E/R+ Ba/F3, as indicated by high levels of H2AX phoshorilation, as well as transcription of the activation-induced cytidine deaminase (AID) enzyme (which is implicated in ETV6-RUNX1+ pre-leukemia to leukemia transition). However, while control cells go through apoptosis, pre-leukemic Ba/F3 are resistant to this fail-safe mechanism, increasing chance to accumulate secondary mutations and malignantly transform. Finally, an inflamed MSC favor the emergence of CD34+ILR7+ compartment within ETV6-RUNX1+ UCB-CD34+ population while decreasing its frequency in the normal counterpart; of note, such differential effect doesn’t occur in case of unstimulated MSC. This observation is particularly important as the CD34+ILR+ compartment seems to represent the critical developmental stage during early fetal hematopoiesis for ETV6-RUNX1 pre-leukemic activity. Concluding, our work demonstrated that BM-MSC and inflammation cooperate in favoring the persistence and transformation of ETV6-RUNX1+ pre-leukemic clone. Elucidating mechanisms that underlay such promoting action could provide novel strategies for the pre-leukemic clone eradication.
Montzka, Katrin [Verfasser]. "An investigation into the characteristics and potential therapeutic application of human bone marrow-derived mesenchymal stromal cells in experimental spinal cord injury / Katrin Montzka." Aachen : Hochschulbibliothek der Rheinisch-Westfälischen Technischen Hochschule Aachen, 2011. http://d-nb.info/1018200118/34.
Повний текст джерелаGanier, Clarisse. "Potentiel thérapeutique des cellules stromales mésenchymateuses dans l'épidermolyse bulleuse dystrophique récessive Intradermal injection of bone marrow-MSCs corrects recessive dystrophic epidermolysis bullosa in a xenograft model Intradermal injection of human umbilical cord-MSCs shows less efficacy than bone marrow-MSCs to correct recessive dystrophic epidermolysis bullosa in a xenograft model." Thesis, Sorbonne Paris Cité, 2018. https://wo.app.u-paris.fr/cgi-bin/WebObjects/TheseWeb.woa/wa/show?t=2117&f=15515.
Повний текст джерелаRecessive dystrophic epidermolysis bullosa (RDEB) is a severe skin disease caused by loss-of-function mutations in COL7A1 encoding type VII collagen. Type VII collagen forms anchoring fibrils which are essential structures for dermal-epidermal adherence. Patients with RDEB suffer since birth from skin and mucosal blistering and develop severe complications. The development of aggressive squamous cell carcinomas is the first cause of demise of these young patients. To date, there is no treatment. Mesenchymal stromal cells (MSC) are multipotent cells, isolated from adult tissue (bone marrow, adipose tissue) or perinatal tissue (umbilical cord). Previous works have shown that local and systemic injections of allogeneic bone marrow-derived MSC (BM-MSC) have a potential to reduce skin inflammation and to improve wound healing in RDEB patients. However, clinical improvement was transient and the mechanisms of action of BM-MSC in RDEB and also their survival after injection are still poorly understood. BM-MSC could act through immunomodulation, anti-fibrotic and angiogenic proprieties, paracrine effects leading to type VII collagen production in the host tissues and/or type VII collagen secretion by injected BM-MSC. The aim of our work was to study the therapeutic potential of MSC for RDEB in preclinical models. We first showed that BM-MSC produce COL7A1 mRNA and type VII collagen levels comparable to healthy dermal fibroblasts in culture. We then assessed the long-term capacity of human BM-MSC to survive, produce and deposit type VII collagen at the dermal-epidermal junction (DEJ) after local injection in human RDEB skin equivalents transplanted onto nude mice. In vivo intradermal (ID) injection of a single dose of human BM-MSC led to the production and deposition of human type VII collagen at the DEJ and allowed anchoring fibrils formation for at least six months post-injection. Injected human BM-MSC were found in the skin at least four months post-injection. These data show that intradermally injected human BM-MSC have the potential to improve dermal-epidermal adhesion of RDEB skin equivalents through sustained deposit of type VII collagen molecules and subsequent anchoring fibrils formation. We then compared the efficacy of human Umbilical Cord Wharton's Jelly-MSC (UC-MSC) with BM-MSC using the same methodology as previously described. UC-MSC showed in vitro a significantly higher amount of COL7A1 mRNA and type VII collagen compared to BM-MSC and healthy dermal fibroblasts in culture. ID injection of a single dose of UC-MSC in vivo led to the production and deposition of low levels of human type VII collagen at the DEJ for four months post-injection. Injected human UC-MSC were found in the skin two months post-injection. These data disclosed a lower efficacy of UC-MSC to restore collagen VII at the DEJ compared to BM-MSC injected in the same xenograft RDEB model. These data open the perspective of using gene-corrected BM-MSC from a Col7a1-/- RDEB murine model to restore normal dermal-epidermal adhesion. Col7a1-/- mice reproduce cutaneous and mucosal lesions observed in RDEB patients. The life expectancy of these animals is very short. We could show that transduction of Col7a1-/- murine BM-MSC in culture using a COL7A1-expressing SIN retroviral vector led to type VII collagen expression levels which were 30-fold higher on average than in BM-MSC from WT mice. In vivo data are required to determine whether the injection of gene-corrected BM-MSC has the potential to treat skin and mucosal lesions in RDEB mice and to define the optimal dose and duration of the effect in vivo. Restoration of type VII collagen expression and anchoring fibrils formation in Col7a1-/- mice would represent an important step towards clinical translation
Burk, Janina. "Klinische Anwendung und vergleichende Charakterisierung equiner mesenchymaler Stromazellen." Doctoral thesis, Universitätsbibliothek Leipzig, 2012. http://nbn-resolving.de/urn:nbn:de:bsz:15-qucosa-99657.
Повний текст джерелаIn horses, mesenchymal stromal cells (MSCs) are used for the treatment of musculoskeletal diseases, especially tendon injuries, with promising results. Previous clinical studies mainly focused on the treatment of superficial digital flexor tendon injuries in racehorses, which, however, represent only a relatively small percentage of the overall equine case load in Germany. Average outcome to be expected following MSC treatment of suspensory ligament injuries was not yet determined. Moreover, basic knowledge on equine MSC biology is still deficient, hampering the understanding and thus the optimisation of the existing treatment regime. The use of alternative MSC sources is frequently discussed, yet to date, only few data comparing the cellular properties of equine MSCs from different sources have been published. The aim of this study was, on the one hand, to gain more knowledge concerning the expected outcome after MSC treatment of tendon injuries, including injuries to the suspensory ligament. On the other hand, it was aimed at expanding the knowledge on equine MSC characterisation in vitro, thereby focusing on the comparison of clinically relevant properties of MSCs derived from different sources. In the clinical study, 98 horses were included, all of which had received MSC treatment for tendon or ligament injuries. In 58 of these horses, long term results after a follow-up period of at least one year could be collected. These data were analysed with respect to treatment outcome and potential influencing factors. Treatment was considered successful when horses were back to full training or competition after the follow-up period, without having suffered a re-injury. The overall success rate was 84.5 %. Success rates in horses suffering from superficial digital flexor tendon injuries and in horses suffering from suspensory ligament injuries were comparably good (84.2 % and 83.3 %, respectively). Similar to conventional therapies, the sports discipline in which the horses performed, age and disease stage tended to influence the outcome. Overall, re-injury rates after MSC treatment were considerably lower than those described in the literature following conventional treatment. For the comparative characterisation of MSCs from different sources in vitro, samples of bone marrow, adipose and tendon tissue, as well as umbilical cord blood and –tissue were collected. Plastic-adherent MSCs were isolated out of these samples and comparatively characterised focusing on cell yields, proliferation and migration properties, trilineage differentiation potential and the expression of the tendon markers collagen 1A2 and scleraxis. MSC yields were significantly higher in all solid tissues (adipose, tendon and umbilical cord tissue) (p < 0.001). Further, MSCs from adipose and tendon tissue proliferated significantly faster than MSCs from bone marrow or umbilical cord blood (p < 0.01). Moreover, approximately three quarters of the samples derived from the latter sources underwent senescence before reaching passage eight. The highest migration potential was found in MSCs derived from tendon and adipose tissue again, while MSCs from umbilical cord tissue showed the least (p < 0.01). The adipogenic differentiation potential was comparably good in MSCs from all different sources. The osteogenic differentiation was most distinct in MSCs from bone marrow, while MSCs from umbilical cord blood and tissue showed only weak evidence of differentiation (day 21: p < 0.01; day 35: p < 0.05). In contrast, following chondrogenic differentiation, MSCs from umbilical cord blood scored highest and MSCs from bone marrow scored lowest (p < 0.05). Collagen 1A2 was most highly expressed in MSCs from adipose tissue, highest scleraxis expression levels were found in MSCs from umbilical cord blood. MSCs from tendon tissue, however, expressed both markers at almost evenly high levels. Contrastingly, lowest expression levels of both markers were found in MSCs derived from bone marrow (p < 0.05 for collagen 1A2). Based on the results of the clinical study, MSC therapy can still be considered a very promising treatment option for tendon diseases and is also a suitable treatment for suspensory ligament injuries. In the future, controlled clinical studies will have to further confirm the efficacy of this treatment regime. The in-vitro-study showed significant differences between equine MSCs derived from different sources, which should be considered when choosing a MSC source for clinical applications. For autologous therapies, MSCs derived from adipose tissue appear to be a good alternative to MSCs derived from bone marrow, due to their remarkable proliferation and reliable differentiation capacities. Furthermore, according to this study, MSCs derived from tendon tissue are especially suitable for treating tendon injuries. Prior to routine clinical applicability of these MSCs, however, their properties should be further investigated
Gittel, Claudia. "Einfluss von Ursprungsquelle und Isolationsmethode auf zellbiologische Charakteristika equiner mesenchymaler Stromazellen." Doctoral thesis, Universitätsbibliothek Leipzig, 2014. http://nbn-resolving.de/urn:nbn:de:bsz:15-qucosa-153119.
Повний текст джерелаNot only in humans but also in veterinary medicine, multipotent mesenchymal stromal cells (MSCs) are a promising treatment option in the therapy of injured musculoskeletal tissues. This is due to the improved tissue regeneration instead of the insufficient reparation following conventional therapies. With regard to an application of MSCs for treatment of tendinopathies in horses, lower rates of reinjury have been reported. However, further investigations to optimize the MSC treatment are still outstanding. Differences in MSCs from different origins have been already reported, but there are still remaining questions about the influence of origin and isolation procedures of MSCs. Fundamental research on equine MSCs derived from different sources and their potential impact due to the isolation process has not been published so far. The aim of this study was to isolate equine MSCs from different sources and to demonstrate potential differences in vitro. Furthermore, differences in cell features following different isolation methods were investigated. In the present study, MSCs from horses were isolated from adipose tissue, tendon tissue, bone marrow, umbilical cord blood and umbilical cord tissue and subsequently subjected to comparative characterization. In case of the solid tissues, two different isolation methods, digestion and explantation, were performed in order to analyze influences on obtained cells. Investigated cell features included cell yield, proliferation, differentiation and migration potential. Furthermore, expression of tendon markers was evaluated with regard to an application of MSCs in tendinopathies. In the present study it was shown that MSCs derived from different sources differ distinctly in cell yield and proliferation potential. In comparison to body fluids, significantly more MSCs could be isolated from solid tissues when using the digestion method (p < 0.001). Furthermore, the cell yield at first cell harvest was distinctly higher when performing the isolation by digestion in comparison to isolation by explantation (p < 0.05). With regard to further cultivation, MSCs derived from tendon tissue and adipose tissue displayed a significantly better proliferation potential compared to MSCs derived from other sources. Considering the differentiation potential, significant differences were obvious between the MSCs derived from different sources. Bone marrow-MSCs showed an excellent osteogenic differentiation capacity in comparison to MSCs derived from umbilical cord blood and tissue (p < 0.05). In contrast, the birth-associated MSCs displayed a distinctly better chondrogenic differentiation than MSCs derived from bone marrow (p < 0.05). No difference in the differentiation potential was noticeable following the different isolation procedures. Furthermore, differences in the gene expression of tendon markers were evident with regard to the cell source. MSCs derived from adipose tissue and tendon tissue expressed collagen 1A2 on the highest level. On the other hand, scleraxis was expressed highest in MSCs derived from umbilical cord blood and tendon tissue. In these cells, MSCs isolated by the digestion method showed a significantly higher expression level of scleraxis in comparison to MSCs isolated by explantation (p < 0.05). Based on the results obtained so far, a relevant impact of the source of MSCs on cell features was evident. MSCs derived from adipose tissue are a promising alternative to bone marrow-MSCs. However, with regard to a clinical application of MSCs, a selection of the MSC source depending on the respective intended use seems to be advantageous. For routine isolation of MSCs from solid tissues, the digestion method could be recommended due to the higher obtainable cell numbers. Furthermore, a negative influence of the enzymatic digestion on the cell features was not detectable. However, to what extent the observed differences in vitro are relevant for in-vivo-applications needs to be further investigated
Wu, Yuenv. "Altered interactions between mesenchymal stromal cells and hematopoietic stem cells from MDS and AML through expression of FAK." Thesis, Lyon, 2019. http://www.theses.fr/2019LYSE1123.
Повний текст джерелаFAK is a cytoplasmic tyrosine kinase that regulates diverse cellular processes, including survival, proliferation, differentiation, and motility. Though various studies have demonstrated the importance of FAK in MDS and AML pathogenesis, the role of this molecule in MDS and AML tumor microenvironment remained to be further determined. By examining BM MSCs derived from MDS and AML patients, we have observed a continues increase of FAK expression and activation during MDS progression to AML, similar to those detected in hemopoietic counterparts. In LR-MDS, MSCs were found to be characterized by low FAK expression and activation. They exhibited altered morphology, immunophenotype, differentiation, and expression of hematopoiesis-supporting factors. Of note, these features could be largely reproduced in normal MSCs by FAK inhibition. Furthermore, FAK depletion in BM stromal cell line could induce massive expansion and apoptosis of normal HSPCs. Our results highlight a critical role of FAK in maintaining the functions of BM MSCs and provide evidence that dysregulation of FAK in MSCs contribute to the disturbed hematopoiesis and possibly the progression of myeloid malignancies. A greater understanding of the role that BM microenvironment plays in MDS and AML will enable an increased recognition of poor-risk patients and the development of therapies that target the defected MSCs, thereby improving the clinical outcome
Li, Yueying. "Vieillissement des cellules stromales mésenchymateuses de la moelle osseuse : implications en médecine régénérative." Thesis, Université de Lorraine, 2015. http://www.theses.fr/2015LORR0099/document.
Повний текст джерелаToday with their properties of differentiation into specific cells types, mesenchymal stromal cells (MSC) can be used in regenerative medicine. Bone marrow (BM) is the better characterized one. The researchers have proven that with increasing passage number in culture the proliferation and differentiation potential of MSC decrease. In parallel many researchers have showed the impact of donor age on MSC properties remains controversial. The aim of our study was to better understand the effect of donor age but also culture passages on the proliferation and differentiation ability of bone marrow mesenchymal stromal cells. The samples were separated into 4 groups depending on the donor age (<20 years; 20-40 years; 40-60 years; > 60 years) and The samples were cultured for 5 passages. The results obtained show that the MSC proliferative capacity obtained from young donors is greater than that of cells from older donors. In addition, the proliferative capacity decreases with increasing passage number in culture. In parallel, the ability of colony-forming unit-fibroblast, measured by the CFU-F assay, decreases slightly depending on the age of the donors but significantly depending on the passage. Finally, the MSC differentiation ability decreases according to the passage of the cells but also depending on the donor age. Our study shows that the properties of bone marrow derived MSC are modified not only during amplification in vitro but also in terms of donor age
Dubory, Arnaud. "Interface os-implant titane et ingénierie tissulaire A cadaveric validation of a method based on impact analysis to monitor the femoral stem insertion Bone marrow mesenchymal stem cells predict radiographic osseointegration of cementeless titanium hip cups Survival, adhesion, and expression of osteoblastic genes of human derived-bone marrow mesenchymal stromal cells on PEEK and titanium-coated PEEK lumbar interbody cages." Thesis, Paris Est, 2018. http://www.theses.fr/2018PESC0067.
Повний текст джерелаIntroduction: Titanium, both used as a mean of fixing an implant to its biomechanical properties close to the bone and as a mean of osseointegration has taken a prominent place. The implant stability is essential for the durability of a titanium implant (TI); it depends on 2 phases: a primary phase, MECHANICAL, corresponding to the impaction or primary holding of the TI and a secondary phase, BIOLOGICAL, corresponding to the colonization of TI by bone tissue.The objective of this work was to evaluate and improve during these two phases the osseointegration of the titanium implant:(1) To evaluate the primary stability of uncemented titanium femoral stems by impact analysis corresponding to the measurement of impact over time.(2) To evaluate whether the amount of mesenchymal stromal cells (MSCs) contained in the iliac crest is correlated with the non-recovery survival of acetabular implants impacted in a context of aseptic osteonecrosis of the femoral head.(3) To improve the osseointegration of TI by cell therapy methods in vitro by studying the survival and division of human MSCs in contact with interbody lumbar cages coated with rough titanium alloy.Methods: The evaluation of the primary stability of cementless titanium femoral stems according to the impact analysis was carried out using a hammer equipped with a piezoelectric force sensor on 20 anatomical subjects, i.e. 40 hips. The number of hammer strokes was compared to obtain the ideal impaction of the prosthesis according to 3 different evaluation methods: number of impacts required by the surgeon (Nsurg), number of impacts required by the video analysis of the depression of the stem in the femur (Nvid), numbers of impacts needed by the impact analysis (Ni).To determine whether the amount of MSCs in the iliac crest could reflect the osseointegration of impacted acetabular implants and the risk of surgical revision. The rate of MSCs measured when performing a surgical cell therapy for aseptic osteonecrosis of the femoral head and the clinical and radiographic outcome of acetabular implants subsequently established for these same patients (n = 90), who had total hip arthroplasty in fine were compared. The mean follow-up was 15 years.The cell survival of bone marrow-derived MSCs was evaluated on lumbar interbody cages coated with titanium. Three groups (n = 5) were formed: a control group, a cage group with titanium surface, a cage group without titanium. On each implant, 1 microliter containing 106 human bone MSCs was cultured. The analysis of cell survival, cell proliferation and expression of osteoblastic genes were performed and compared.Results: Regarding the impact analysis of the cementless femoral stem impaction, the difference between NI, Nchir and Nvid was lower than 3 for more than 85% of the configurations performed.For the second study, a small number of MSCs in the iliac crest was a risk factor for surgical revision in patients treated with a cementless acetabular implant.The third study showed that MSCs could grow until 96 hours and could express osteoblastics genes 21 days after cell seed. No difference between PEEK cage and Titanium-coated PEEK has been found.Conclusion: The impact analysis provides objective data on the primary holding of the titanium impacted femoral stem. Titanium is also a favored biomaterial for the survival and proliferation of bone marrow-derived MSC predestined to become osteforming cells, especially since a small number of MSCs seems to be a risk of failure of osseointegration of cementless acetabular implants
Fievet, Loïc Marc André. "Caractérisation phénotypique et fonctionnelle des cellules stromales mésenchymateuses natives de la moelle osseuse humaine adulte." Thesis, Toulouse 3, 2020. http://www.theses.fr/2020TOU30137.
Повний текст джерелаWithin the bone marrow (BM), hematopoietic stem cells (HSC) are hosted in a specialized 3D microenvironment, called "niche", regulating their behavior (e.g. self-renewal, commitment to lineages, proliferation and survival). This niche is composed of several cells such as vascular endothelial, perivascular, and osteoblastic cells. Perivascular mesenchymal stromal cells (MSCs) play a key role in the formation of the microenvironment, both through expression of pro-hematopoietic factors, and their ability to differentiate towards osteoblastic lineage. Therefore, MSCs sub-populations are of crucial physiological importance in the regulation of hematopoiesis, but also for bone formation and regeneration. Recently described in mice at the single-cell level, BM MSCs subsets remain unexplored in humans, as well as their respective roles in the niche. By characterizing these sub-populations, and deciphering their native properties, it will be possible to shape ex vivo the physiological niches in 3D, addressing the major scientific challenges for understanding human hematopoiesis and osteogenesis. Here, we used single-cell RNA sequencing approaches to characterize the human BM stroma and described key hematopoietic niche factors highly conserved between species. We identified subsets of cells expressing different hematopoietic regulatory genes, spanning endothelial cells, mural cells, and especially MSCs with distinct osteoblastic and adipogenic trajectories. Of interest, our data suggest a simple branching differentiation hierarchy with the presence of a multipotent subset: the CXCL12-abundant reticular (CAR) cells at the origin of the other MSCs subpopulations. We confirmed the enrichment of the CXCL12-abundant reticular (CAR) cell subset expressing the Leptin receptor (LEPR+) in the CD45-/CD271+/CD200+ BM fraction as well as their in-situ localization using histological approaches on human biopsies. Secondly, we developed an ex vivo isolation method to preserve and amplify the BM CAR cells, and then studied their self-organization in 3D culture. We found that CAR derived organoids sustained high angiogenesis, secreted CSH niche cytokines, and spontaneously recapitulates early intramembranous bone formation in vitro. Using bioinformatics models and genome editing techniques, we highlighted the role of the WDR35 protein and the primary cilia in osteoblastic differentiation mediated by the Hedgehog and Wnt signaling pathways. Finally, we have shown that ectopic xenotransplantation of CSM-derived organoids could give rise to mature human osteoblasts while forming a niche for CSHs after hematopoietic humanization of immunocompromised NSG mice. Our study is the first map of the human BM stroma at a single-cell resolution, and CAR cells cultured in 3D represent a new tool useful in basic research as well as in regenerative medicine
Clutter, Suzanne Davis. "Chemotherapy disrupts bone marrow stromal cell function." Morgantown, W. Va. : [West Virginia University Libraries], 2006. https://eidr.wvu.edu/etd/documentdata.eTD?documentid=4528.
Повний текст джерелаTitle from document title page. Document formatted into pages; contains x, 180 p. : ill. (some col.). Vita. Includes abstract. Includes bibliographical references.
Valle, Paulo Roberto Del. "Perfil transcricional de fibroblastos de tumor primário, linfonodo e medula óssea de pacientes com câncer de mama." Universidade de São Paulo, 2013. http://www.teses.usp.br/teses/disponiveis/5/5155/tde-26032013-143422/.
Повний текст джерелаmay influence tumor development in the primary site of breast cancer, as well as in regional and distant metastatic sites. In this context, fibroblasts are important stromal cells which influence proliferation and migration of cancer cells and may also provide an appropriate niche to tumor development. Objectives: The main objective of this work is the comparison of stromal cells from the primary tumor (PT), lymph node metastasis (N+) and bone marrow (BM) obtained from breast cancer patients, through gene expression profile. Patients and Methods: The gene expression profile was analyzed in fibroblasts primary culture from 11 breast cancer patients. The expression profiles of PT cells (n=4), N+ cells (n=3) and BM cells (n=4) were determined through a customized cDNA microarray platform (containing 4800 immobilized sequences which represents 4600 genes approximately). The analysis were performed by SAM multiclass (TMEV; FDR 0%), followed by SAM two classes test (TMEV; FDR 0%). Functional analysis was performed using DAVID v6.7. Technical validation was performed in same 6 samples that were previously analyzed in microarray experiments and biological validation was performed in fibroblasts obtained from other group of 16patients by RT-qPCR Results: The expression profile of fibroblasts obtained from three sites revealed 267 differentially expressed genes, which appropriately clustered fibroblasts in three different branches, in accordance with their origin (PT vs. N+ vs. BM). Although the differences between PT and N+ were represented by 20 genes, differences between PT vs. BM and N+ vs. BM were more significant (235 and 245 differentially expressed genes respectively). Functional analysis revealed enrichment of functions related to development and morphogenesis. Afterwards, the expression of some selected genes were analyzed in a different batch of samples (biological validation).Thereby, NOTCH2 confirmed high expression in N+ (vs. PT), and ADCY2, HECTD1, HNMT, LOX, MACF1 and USP16 confirmed high expression in BM (vs. PT). Conclusion: In breast cancer patients, stromal cells obtained from different origins present a differential gene expression profile, which may influence tumor behavior
Dennis, James Edmund. "Mesenchymal progenitor cells in adult marrow." Case Western Reserve University School of Graduate Studies / OhioLINK, 1995. http://rave.ohiolink.edu/etdc/view?acc_num=case1062516436.
Повний текст джерелаLiu, Limin. "Expression of follicular dendritic cell determinants by mouse bone marrow stromal cells." Thesis, National Library of Canada = Bibliothèque nationale du Canada, 1997. http://www.collectionscanada.ca/obj/s4/f2/dsk2/ftp03/MQ37142.pdf.
Повний текст джерела