• Refine Query
  • Source
  • Publication year
  • to
  • Language
  • 11
  • 6
  • 2
  • 1
  • 1
  • 1
  • Tagged with
  • 23
  • 23
  • 23
  • 7
  • 6
  • 5
  • 5
  • 5
  • 5
  • 4
  • 4
  • 4
  • 4
  • 4
  • 4
  • About
  • The Global ETD Search service is a free service for researchers to find electronic theses and dissertations. This service is provided by the Networked Digital Library of Theses and Dissertations.
    Our metadata is collected from universities around the world. If you manage a university/consortium/country archive and want to be added, details can be found on the NDLTD website.
11

CD271+ mesenchymal stromal cells for an intraoperative therapy of chondral defects

Petters, Oliver 01 April 2019 (has links)
Regenerative treatment of focal hyaline cartilage defects could prevent or delay the development of secondary osteoarthritis. Current surgical techniques result partly in i) the formation of mechanically inferior fibrous cartilage or ii) present the disadvantage of the donor site morbidity from harvesting cartilage biopsy as well as iii) the dedifferentiation of chondrocytes due to in vitro expansion and iv) the reduced re-differentiation potential of in vitro expanded chondrocytes. The self-healing capacities of injured and degenerated articular cartilage revealed a promising target cell population for a regenerative, autologous treatment of these defects using mesenchymal stromal cells (MSCs). Several case studies, randomized and controlled clinical studies showed the general ability of autologous, bone marrow-derived, expanded MSC transplantation to regenerate articular cartilage lesions [1]. However, these two-stage approaches are based on time- and cost-consuming expansion of MSCs under good manufacturing practice (GMP) conditions and hold a risk of contamination during this process. In 2010, CD271, the low-affinity nerve growth factor receptor, was described as a suitable surface marker to enrich MSCs from human bone marrow aspirate intraoperatively [2]. The aim of the present dissertation was to investigate the feasibility of generating cartilage grafts from either ovine (study no. 1) and human (study no. 2) non-expanded CD271+ bone marrow cells in a collagen type I hydrogel. Study no. 1 (“Point-of-care treatment of focal cartilage defects with selected chondrogenic mesenchymal stromal cells - An in vitro proof-of-concept study”) investigated several surface marker candidates for the prospective MSC separation and examined their potential of resulting colony-forming units, respective their yield of potent MSCs [3]. This study was conducted with ovine bone marrow samples. CD271 was the most effective surface marker to isolate the target cell population. Subsequently, CD271+, CD271- and unseparated mononuclear cells (MNCs), containing the MSCs, were used to generate cartilage grafts without an expansion of these cells in monolayer culture. It could be proven, that ovine CD271+ cells were able to generate a potent hyaline cartilage graft. Study no. 2 (“Single-stage preparation of human cartilage grafts generated from bone marrow-derived CD271+ mononuclear cells”) was performed as the final translational step from animal-derived bone marrow to human donor material and is therefore strengthening the therapeutically focus of the entire work [4]. Briefly, eight bone marrow aspirates were used for MNC isolation and subsequent magnetic cell separation (MACS). The resulting CD271+ and CD271- MNCs were compared to unseparated MNCs. Subsequently, they were seeded in a clinically approved collagen type I hydrogel and cultivated for up to 5 weeks to investigate the progression of the chondrogenic differentiation processes. Graft analysis included cell viability visualization by live/dead staining, determination of the DNA and the secreted sulphated glycosaminoglycan (sGAG) content as well as the immunohistochemical staining for typical chondrogenic differentiation markers and the extracellular matrix molecules aggrecan and collagen type II. A proliferation of cells in the generated grafts was shown of CD271+ and unsep, but not CD271- MNCs. Hence, the cell number was 2.8-fold higher after 35 days compared to the first day for CD271+ MNCs grafts, while CD271- MNCs did not proliferate in the grafts and unsep MNCs showed only a slight increase in cell number. The chondrogenic potential was measured by quantification of freshly produced sGAGs and the expression of chondrogenic markers. In grafts with CD271+ MNCs, sGAG production increased over time and reached its maximum at day 35, whereas grafts with CD271- MNCs showed no measurable sGAG deposition. The amount of sGAG in unsep MNC grafts increased only slightly over the whole cultivation period. Aggrecan and collagen type II staining varied considerably between the MNCs donors. Collagen type II positive staining was observed in CD271+ MNC grafts (5/8 donors) and unsep MNC (2/8) grafts. In comparison to macroscopically healthy cartilage, three-dimensional grafts of the CD271+ group yielded a proceeding extracellular matrix production. In summary, CD271+ MNCs showed the highest proliferation rate, cell viability, sGAG deposition and cartilage marker expression compared to the CD271- or unseparated MNC fractions in in vitro generated three-dimensional cartilage grafts. Therefore, the presented work demonstrated the feasibility of generating a cartilage graft from CD271+ bone marrow-derived MNCs in a clinically approved collagen type I hydrogel without a previous monolayer expansion of these cells. This will enable the intraoperative purification of CD271+ MNCs, which contain the majority of colony-forming MSCs, by MACS technology. The clinical application will be possible with currently available and clinical approved cell separation devices. Providing a cartilage graft with non-expanded CD271+ MNCs by a fast and simple intraoperative therapeutic approach fulfils the need for a “single-step, tissue-engineered solution to focal cartilage defects, and elimination of the morbidity of the donor defect” as requested by the editors of the journal Arthroscopy [5]. References of the summary 1. Filardo G, et al. (2016). Stem cells in articular cartilage regeneration. J Orthop Surg Res 11:42. 2. Jones E, et al. (2010). Large-scale extraction and characterization of CD271+ multipotential stromal cells from trabecular bone in health and osteoarthritis: implications for bone regeneration strategies based on uncultured or minimally cultured multipotential stromal cells. Arthritis Rheum. 62:1944–1954. 3. Petters O, et al. (2018). Point-of-care treatment of focal cartilage defects with selected chondrogenic mesenchymal stromal cells-An in vitro proof-of-concept study. J Tissue Eng Regen Med. 4. Petters O, et al. (2018). Single-Stage Preparation of Human Cartilage Grafts Generated from Bone Marrow-Derived CD271+ Mononuclear Cells. Stem Cells Dev 27:545–555. 5. Lubowitz JH and GG Poehling. (2009). Saving our cells: Advances in tissue engineering for focal cartilage defects. Arthroscopy: the journal of arthroscopic & related surgery: official publication of the Arthroscopy Association of North America and the International Arthroscopy Association 25:115–116.:1 Introduction 1 1.1 Articular cartilage 1 1.2 Cartilage lesions 2 1.3 Self-healing capability of articular cartilage 3 1.4 Treatment option for cartilage lesions 4 1.5 Mesenchymal stromal cells in cartilage regeneration 6 2 Rationale 8 3 Publication manuscripts 9 Point-of-care treatment of focal cartilage defects with selected chondrogenic mesenchymal stromal cells - An in vitro proof-of-concept study 9 Single-stage preparation of human cartilage grafts generated from bone marrow-derived CD271+ mononuclear cells 21 4 Summary 33 5 References 35 6 Appendix 42 7 Declaration of Authorship 47 8 Curriculum vitae 48 9 Publications 49 10 Acknowledgements 49
12

CCAAT/Enhancer-Binding Proteinβ Expressed by Bone Marrow Mesenchymal Stromal Cells Regulates Early B-Cell Lymphopoiesis / 骨髄間葉系ストローマ細胞に発現する転写因子C/EBPβは初期B細胞造血を制御する

Yoshioka, Satoshi 23 January 2014 (has links)
京都大学 / 0048 / 新制・課程博士 / 博士(医学) / 甲第17978号 / 医博第3842号 / 新制||医||1001(附属図書館) / 80822 / 京都大学大学院医学研究科医学専攻 / (主査)教授 長澤 丘司, 教授 河本 宏, 教授 江藤 浩之 / 学位規則第4条第1項該当 / Doctor of Medical Science / Kyoto University / DFAM
13

Detection of Collagen in Rat Abdominal Wound Healing: Contributions of Mesenchymal Stromal Cells and Platelet-Rich Plasma

Minteer, Tanya E. 28 September 2012 (has links)
No description available.
14

Functional characterisation of cardiac progenitors from patients with ischaemic heart disease

Zhang, Huajun January 2013 (has links)
Ischaemic heart disease (IHD) is the leading cause of death worldwide. Currently, even optimal medical therapies do not attenuate deterioration of the left ventricular (LV) function completely. Stem cell therapies, and recently cardiac stem cell therapies, have emerged as potential novel treatments for IHD. However, clinical evidence from randomised controlled studies has shown mixed results. Thus understanding what patient-related factors may affect the therapeutic performance of the cells may help improving treatment outcomes. The studies described in this thesis aim to understand how cardiac progenitor cells (CPCs) can re-vascularise ischaemic myocardium and promote functional repair of the heart. Resident CPCs were isolated and expanded from the right atrial appendage of 68 patients following the ‘cardiosphere’ method (cardiosphere-derived cells or CDCs). They resemble mesenchymal progenitors as they lack the expression of endothelial and haematopoietic cell surface markers but express mesenchymal progenitor cell markers (e.g. CD105, CD90). Cell function was evaluated by support of angiogenesis, mesenchymal lineage differentiation potential in vitro, and improvement in heart function in vivo. Notably in vitro, CDC from different patients differed in their angiogenic supportive and differentiation potentials. In a rodent model of myocardial infarction (MI), transplantation of CDC reduced infarct size significantly (p<0.05). However, only those CDCs with a robust pro-angiogenic ability in vitro improved vessel density and heart systolic function (p<0.05) in vivo. A multiple regression model, which accounted for 51% of the variability observed, identified New York Heart Association (NYHA) class, smoking, hypertension, type of ischaemic disease and diseased vessel as independent predictors of angiogenesis. In addition, gene expression analyses revealed that differential gene expression of several extracellular matrix components (e.g. CUX1, COL1A2, BMP1 genes and microRNA-29b) could explain the differences observed in CDC’s vascular supportive function. In summary, this is the first description of variability in the pro-angiogenic and differentiation potential of CDCs and its correlation with their therapeutic potential. This study indicates that patient stratification may need to be included in the design of future trials to improve the efficacy of cell-based therapies.
15

Einfluss von Ursprungsquelle und Isolationsmethode auf zellbiologische Charakteristika equiner mesenchymaler Stromazellen / Influence of origin and isolation method on cell biological features of equine mesenchymal stromal cells

Gittel, Claudia 02 October 2014 (has links) (PDF)
Multipotente mesenchymale Stromazellen (MSCs) stellen nicht nur beim humanen Patienten, sondern auch in der Veterinärmedizin einen vielversprechenden Therapieansatz in der Behandlung erkrankter muskuloskelettaler Gewebe dar. Ziel der Behandlung ist dabei die Regeneration der betroffenen Strukturen im Vergleich zur Reparation nach konservativer Therapie. Vor allem im Bereich von Sehnenerkrankungen können nach MSC-Applikation vielversprechende Ergebnisse im Hinblick auf niedrigere Rezidivraten beobachtet werden. Dennoch sind noch nicht alle Umstände einer optimalen MSC-Anwendung geklärt. Hierbei sind unter anderem Fragen bezüglich der Herkunft und Gewinnung von MSCs offen, da Unterschiede von MSCs aufgrund ihrer Gewebezugehörigkeit bereits nachgewiesen wurden. Grundlegende umfassende Arbeiten zum Vergleich von equinen MSCs aus verschiedenen Quellen sowie deren mögliche Beeinflussung durch die Isolierung aus dem Gewebe lagen bislang noch nicht vor. Ziel dieser Studie war es daher, equine MSCs aus verschiedenen Quellen zu gewinnen und mögliche Unterschiede in vitro aufzuzeigen. Weiterhin sollten Unterschiede zwischen den Zelleigenschaften nach Anwendung verschiedener Isolationsprotokolle untersucht werden. In der hier vorliegenden Studie wurden MSCs aus Fett- und Sehnengewebe, Knochenmark, Nabelschnurblut und Nabelschnurgewebe von Pferden isoliert und vergleichend charakterisiert. Dabei wurden für die soliden Körpergewebe zwei unterschiedliche Isolationsmethoden, die Digestion und die Explantation, angewendet, um mögliche Einflüsse auf die gewonnen Zellen zu ermitteln. Die untersuchten Kriterien beinhalteten Zellertrag, Proliferation, Differenzierungspotenz und das Migrationsverhalten von MSCs. Hinblickend auf eine Anwendung von MSCs bei Sehnenerkrankungen wurde auch die Expression von Sehnenmarkern verglichen. In der vorliegenden Studie konnte gezeigt werden, dass sich die MSCs aus verschiedenen Quellen hinsichtlich der Zellausbeute und ihres Wachstumspotentials unterschieden. Aus soliden Geweben konnten mittels Digestion im Vergleich zu Körperflüssigkeiten signifikant mehr MSCs isoliert werden (p < 0,001). Dabei erbrachte die Isolation von MSCs mittels Digestionsmethode einen deutlich höheren Zellertrag nach der Passage 0 im Vergleich zur Explantationsmethode (p < 0,05). Im weiteren Verlauf der Kultivierung zeigten MSCs aus Sehnengewebe und Fettgewebe ein signifikant besseres Proliferationsverhalten im Vergleich zu Knochenmark-MSCs und Nabelschnurblut-MSCs. Im Hinblick auf das Differenzierungspotential konnten signifikante Unterschiede zwischen den MSCs aus den verschiedenen Quellen beobachtet werden. MSCs aus Knochenmark zeigten eine sehr gute osteogene Differenzierungsfähigkeit im Vergleich zu MSCs aus den geburtsassoziierten Geweben (p < 0,05). Im Gegensatz dazu zeichneten sich diese MSCs durch eine deutlich bessere chondrogene Differenzierung im Vergleich zu Knochenmark-MSCs aus (p < 0,05). Im Hinblick auf die Isolationsmethode konnten keine Unterschiede im Differenzierungspotential beobachtet werden. Weitere Unterschiede aufgrund der Zellquelle lassen sich in der Genexpression der Sehnenmarker erkennen. MSCs aus Fettgewebe und Sehnengewebe exprimierten Kollagen 1A2 auf höchstem Niveau. Sklexaris hingegen wurde von MSCs aus Nabelschnurblut und Sehnengewebe am höchstem exprimiert. Dabei zeigten MSCs, die mittels Digestionsmethode isoliert worden waren, ein signifikant höheres Expressionslevel von Skleraxis im Vergleich zur Explantationsmethode (p < 0,05). Die Ergebnisse der vorliegenden Studie lassen einen Einfluss der Zellquelle auf die Zellcharakteristika erkennen. MSCs aus Fettgewebe stellen dabei eine vielversprechende Alternative zu Knochenmark-MSCs dar. Allerdings scheint für eine klinische Anwendung von MSCs eine selektive Auswahl der Zellquelle entsprechend der vorliegenden Erkrankung von Vorteil zu sein. Dabei ist eine Isolierung von MSCs aus soliden Geweben mittels Digestionsverfahren zu empfehlen, da hier deutlich höhere Zellzahlen gewonnen werden können. Eine negative Beeinflussung der Zelleigenschaften durch die enzymatische Digestion lässt sich nach den vorliegenden Ergebnissen nicht vermuten. Inwiefern die beobachteten Unterschiede bei in-vivo-Anwendungen von Bedeutung sind, muss jedoch noch umfassend untersucht werden. / Not 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.
16

Function of Fra1 in mesenchymal stromal cell differentiation & the potential immune modulatory role of Fra1

Drießler, Frank 06 August 2008 (has links)
Aktivator Protein-1 (AP-1) ist ein kollektiver Terminus für dimerische Transkriptionsfaktoren, die sich aus Fos- und Jun- Proteinen zusammensetzen. Diese Untereinheiten binden an eine gemeinsame, spezifische DNA-Sequenz, die AP-1 Bindungsstelle. Zusätzlich zu der gut dokumentierten Rolle des c-Fos Proteins in der Tumorgenese, wo dieses Gen als ein Aktivator beschrieben ist, übt AP-1 einen Einfluss auf mesenchymale Stromazellen und Immunzellen aus. Mesenchymale Knochenmarkszellen sind die Vorläuferzellen für Adipozyten, Osteoblasten, Chondrozyten, Myozyten und Fibroblasten. Die molekularen Mechanismen, welche die Differenzierungen regeln, sind noch weitgehend unerforscht. Der heterodimere Transkriptionsfaktor AP-1 übt eine wichtige Rolle in der Kontrolle der Zelldifferenzierung aus. Verschieden genetisch veränderte Mausmodelle untermauerten dies. Mäuse, welche das Fos-related antigen-1 (Fra1) oder eine kürzere Protein-Isoform von FosB (deltaFosB) überexpremieren, entwickelten, durch eine beschleunigte Differenzierung der Osteoblasten, eine Osteosklerose. Interessanterweise konnte gezeigt werden, dass die transgenen deltaFosB Mäuse weniger Fett haben. Die Stabilität und Aktivität von Fos Proteinen kann durch post-transkriptionale Modifizierungen geregelt werden. Basierend auf knockout Mausmodellen, wurde eine tragende Rolle für das wachstumsregulierende Enzym Rsk2 postuliert. Rsk2 spielt eine mögliche Rolle bei der Ausdifferenzierung von mesenchymalen Vorläuferzellen zu Osteoblasten und Adipozyten. Das Ziel dieser Arbeit war es molekulare Mechanismen zu finden, welche die unterschiedlichen Phänotypen (wild typ, fra1-tg, rsk2-defizient und fra1-tg/rsk2-defizient) charakterisieren. Die Knochenuntersuchungen der verschiedenen Genotypen zeigten, dass Fra1 und Rsk2, unabhängig voneinander, tragende Rollen im Knochenmetabolismus spielen. Quantitative Analysen von Adipozytenmarker, wie PPARgamma und C/EBPalpha zeigten, dass das Protein Fra1 die Adipozytenreifung in vivo und in vitro reguliert. Zusätzlich entwickelten die „doppel-mutierten“ fra1-tg/rsk2-/y Mäuse einen Lipodystrophy. Ein milderer Phänotyp wurde in den fra1-tg Tieren beobachtet, jedoch nicht in den Rsk2-knockout Mäusen. Zusätzlich wurde beobachtet, dass mesenchymale Zellen, welche Fra1 überexprimieren, gegen Glucocorticoid-induzierte Wachstumshemmung resistent waren. Diese Wirkung kann am wahrscheinlichsten durch die Fra1-vermittelte Suppression des Glucocorticoidrezeptors erklärt werden. Außerdem beeinflusste die Überexpression von Fra1 die Milzentwicklung. Leber und Herzanalysen zeigten, dass Fra1 kollagenhaltiges Gewebe induziert. Krankheiten wie Cholangitis und Fibrosen waren die Folge. / AP-1 transcription factor is a general name for multiple dimers formed by the association of Fos (or ATF) and Jun proteins. AP-1 acts as a sensor of changes in the cellular environment and thus, it is implicated in the modulation of cell proliferation, differentiation, transformation and cell death. Besides the well-documented role of c-Fos protein in oncogenesis, where this gene can function as a tumor promoter, AP-1 proteins are being recognized as regulators for mesenchymal stromal cell development and as regulators of immune cells. The mesenchymal stromal cells are the common progenitors for various mesenchymal lineages such as adipocytes, osteoblasts, chondrocytes, myocytes and fibroblasts. AP-1 seems to play a key role in the control of mesenchymal cell fate decision and differentiation. This is suggested by phenotypes of mice with a genetic modifications in either the Jun or the Fos component of AP-1. In particular, mice overexpressing the Fos-related antigen-1 (Fra1) or the short isoform of FosB (deltaFosB) have been found to develop osteosclerosis due to an accelerated differentiation of osteoblasts. Interestingly, mice overexpressing deltaFosB also developed less fat tissue. The activity of Fos proteins can be regulated by post-transcriptional modification. Based on knockout mouse model, a role for the growth factor regulated kinase Rsk2 was proposed in the differentiation of mesenchymal stromal cells to osteoblasts as well as in fat tissue development. Goal in this study was to identify the molecular mechanisms explaining the differences between the wild type, fra1-tg, rsk2-deficient and fra1-tg/rsk2-deficient phenotypes. The comparison of the bones of the different mice genotypes revealed, that Fra1 and Rsk2 were independently regulating bone metabolism. Quantitative analysis of adipocyte markers expressions, like PPARgamma and C/EBPalpha revealed, that Fra1 overexpression was blocking adipocyte maturation in vivo and in vitro. Moreover, the in vivo results show that the fra1-tg/rsk2-/y mice develop a severe lipodystrophy. A milder phenotype was observed in the parental fra1-tg strain but not in the Rsk2 knockout strain. Additionally, it was been observed, that mesenchymal cells overexpressing Fra1 were resistant to glucocorticoid-induced growth inhibition. This effect can most likely be explained by Fra1-mediated downregulation of the glucocorticoid receptor. Furthermore, Fra1 overexpression influenced spleen development. Liver and heart analyses showed that Fra1 overexpression induced collagen tissue. Diseases like cholangitis and fibrosis were the outcome.
17

Uso de meio condicionado por células estromais mesenquimais uterinas durante o cultivo in vitro de embriões bovinos

Cintra, Lais do Nascimento January 2018 (has links)
Orientador: Fernanda da Cruz Landim-Alvarenga / Resumo: As tecnologias de reprodução assistida, tais como a fertilização in vitro (FIV), transferência de embriões, transgenia e clonagem, ainda não tem o impacto comercial desejado devido a baixa produção embrionária. Apenas 30 a 40% dos blastocistos desenvolvidos são obtidos de oócitos após a MIV, fertilização e cultivo dos embriões, embora 80% dos oócitos maturados in vitro sejam fertilizados com sucesso. O soro fetal bovino (SFB) é o suplemento mais utilizado no cultivo de embriões in vitro, uma vez que melhora o desenvolvimento dos blastocistos. Apesar disso, sua presença está relacionada a alterações do metabolismo embrionário, perda de qualidade e indução de modificações na expressão de vários genes embrionários. Na tentativa de minimizar os efeitos deletérios do SFB, várias citocinas e fatores de crescimento têm sido acrescentados aos meios de cultivo embrionários in vitro, com a intenção de mimetizar as condições de cultivo in vivo. O presente experimento tem como objetivo avaliar e comparar os efeitos da adição do SFB e de meio condicionado por células mesenquimais estromais (MSCs) durante o cultivo embrionário. Os parâmetros analisados foram a viabilidade embrionária, apoptose e o perfil transcricional de genes relacionados à qualidade dos embriões. Não foi observado uma diferença (P≥0,05) na clivagem dos blastocistos, porém observou-se que a taxa de produção de embriões utilizando SFB no CIV foi maior (P≤0,05) quando comparada com MC, mas não diferiu (P≥0,05) do grupo pro... (Resumo completo, clicar acesso eletrônico abaixo) / Abstract: Endometrial mesenchymal stromal cell (eMSCs) secretes bioactive molecules such as cytokines and growth factors which are released as soluble factors or through extracellular vesicles (EVs). Conditioned medium (CM) of the MSCs maintains the immunomodulation and regenerative potential properties of the cells that produced it. This study investigated the use of CM by eMSC plus BSA as alternative to FBS in embryo culture medium. The developmental ability and quality of bovine embryos were determined by assessing their cell number and gene expression. The percentage of embryos that underwent cleavage was similar (P>0.05) among the groups but blastocyst formation was higher (P<0.05) in FBS group. The total cell number was higher in CM group, but not statistically different from the others (P>0.05). The relative mRNA expression of ELOVL6 was higher in the CM group, CASP3 in the BSA group, ACSL3 and VEGF in the FBS group. Taken together, these data suggest that CM can be used as an alternative supplement to FBS. We observed a different gene expression profile, suggesting the CM inhibited an increase in the relative mRNA levels for CASP3. Moreover, the CM favored the total number cells, inhibited the percentage of cells in apoptosis and produces better quality embryo. / Mestre
18

Einfluss von Ursprungsquelle und Isolationsmethode auf zellbiologische Charakteristika equiner mesenchymaler Stromazellen

Gittel, Claudia 17 June 2014 (has links)
Multipotente mesenchymale Stromazellen (MSCs) stellen nicht nur beim humanen Patienten, sondern auch in der Veterinärmedizin einen vielversprechenden Therapieansatz in der Behandlung erkrankter muskuloskelettaler Gewebe dar. Ziel der Behandlung ist dabei die Regeneration der betroffenen Strukturen im Vergleich zur Reparation nach konservativer Therapie. Vor allem im Bereich von Sehnenerkrankungen können nach MSC-Applikation vielversprechende Ergebnisse im Hinblick auf niedrigere Rezidivraten beobachtet werden. Dennoch sind noch nicht alle Umstände einer optimalen MSC-Anwendung geklärt. Hierbei sind unter anderem Fragen bezüglich der Herkunft und Gewinnung von MSCs offen, da Unterschiede von MSCs aufgrund ihrer Gewebezugehörigkeit bereits nachgewiesen wurden. Grundlegende umfassende Arbeiten zum Vergleich von equinen MSCs aus verschiedenen Quellen sowie deren mögliche Beeinflussung durch die Isolierung aus dem Gewebe lagen bislang noch nicht vor. Ziel dieser Studie war es daher, equine MSCs aus verschiedenen Quellen zu gewinnen und mögliche Unterschiede in vitro aufzuzeigen. Weiterhin sollten Unterschiede zwischen den Zelleigenschaften nach Anwendung verschiedener Isolationsprotokolle untersucht werden. In der hier vorliegenden Studie wurden MSCs aus Fett- und Sehnengewebe, Knochenmark, Nabelschnurblut und Nabelschnurgewebe von Pferden isoliert und vergleichend charakterisiert. Dabei wurden für die soliden Körpergewebe zwei unterschiedliche Isolationsmethoden, die Digestion und die Explantation, angewendet, um mögliche Einflüsse auf die gewonnen Zellen zu ermitteln. Die untersuchten Kriterien beinhalteten Zellertrag, Proliferation, Differenzierungspotenz und das Migrationsverhalten von MSCs. Hinblickend auf eine Anwendung von MSCs bei Sehnenerkrankungen wurde auch die Expression von Sehnenmarkern verglichen. In der vorliegenden Studie konnte gezeigt werden, dass sich die MSCs aus verschiedenen Quellen hinsichtlich der Zellausbeute und ihres Wachstumspotentials unterschieden. Aus soliden Geweben konnten mittels Digestion im Vergleich zu Körperflüssigkeiten signifikant mehr MSCs isoliert werden (p < 0,001). Dabei erbrachte die Isolation von MSCs mittels Digestionsmethode einen deutlich höheren Zellertrag nach der Passage 0 im Vergleich zur Explantationsmethode (p < 0,05). Im weiteren Verlauf der Kultivierung zeigten MSCs aus Sehnengewebe und Fettgewebe ein signifikant besseres Proliferationsverhalten im Vergleich zu Knochenmark-MSCs und Nabelschnurblut-MSCs. Im Hinblick auf das Differenzierungspotential konnten signifikante Unterschiede zwischen den MSCs aus den verschiedenen Quellen beobachtet werden. MSCs aus Knochenmark zeigten eine sehr gute osteogene Differenzierungsfähigkeit im Vergleich zu MSCs aus den geburtsassoziierten Geweben (p < 0,05). Im Gegensatz dazu zeichneten sich diese MSCs durch eine deutlich bessere chondrogene Differenzierung im Vergleich zu Knochenmark-MSCs aus (p < 0,05). Im Hinblick auf die Isolationsmethode konnten keine Unterschiede im Differenzierungspotential beobachtet werden. Weitere Unterschiede aufgrund der Zellquelle lassen sich in der Genexpression der Sehnenmarker erkennen. MSCs aus Fettgewebe und Sehnengewebe exprimierten Kollagen 1A2 auf höchstem Niveau. Sklexaris hingegen wurde von MSCs aus Nabelschnurblut und Sehnengewebe am höchstem exprimiert. Dabei zeigten MSCs, die mittels Digestionsmethode isoliert worden waren, ein signifikant höheres Expressionslevel von Skleraxis im Vergleich zur Explantationsmethode (p < 0,05). Die Ergebnisse der vorliegenden Studie lassen einen Einfluss der Zellquelle auf die Zellcharakteristika erkennen. MSCs aus Fettgewebe stellen dabei eine vielversprechende Alternative zu Knochenmark-MSCs dar. Allerdings scheint für eine klinische Anwendung von MSCs eine selektive Auswahl der Zellquelle entsprechend der vorliegenden Erkrankung von Vorteil zu sein. Dabei ist eine Isolierung von MSCs aus soliden Geweben mittels Digestionsverfahren zu empfehlen, da hier deutlich höhere Zellzahlen gewonnen werden können. Eine negative Beeinflussung der Zelleigenschaften durch die enzymatische Digestion lässt sich nach den vorliegenden Ergebnissen nicht vermuten. Inwiefern die beobachteten Unterschiede bei in-vivo-Anwendungen von Bedeutung sind, muss jedoch noch umfassend untersucht werden. / Not 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.
19

Adult and Embryonic Stem Cell Sources for Use in a Canine Model of In Utero Transplantation

Vaags, Andrea Kathleen 05 March 2012 (has links)
Dogs are useful preclinical models for the translation of cell transplantation therapies from the bench to the bedside. In order for canine models to be utilized for stem cell transplantation research, it is necessary to advance discoveries in the fields of canine stem cell biology and transplantation. The use of side population hematopoietic stem cells (HSCs) has garnered much interest for the purification of mouse HSCs and has been translated to several other species, including human. In order to assess if this method of purification of HSCs could be useful for stem cell therapies in humans, safety and efficacy studies in a large animal model, such as the dog would be required. With this objective in mind, we isolated canine bone marrow-derived side population (SP) stem cells and assessed their multilineage differentiation in vitro and engraftment potential in vivo. Utilizing a pregating strategy to enrich for small, agranular SP cells we were able to enrich for blast cells, expressing the ABCG2 transmembrane pump known to be associated with murine and human SP cells. Canine SP cells were also enriched for C-KIT positive cells and lacked expression of CD34 as identified in other species. The small, agranular SP fraction had high CFU potential after long-term culture with canine bone marrow stromal cells and cytokine supplementation. Yet, canine SP cells demonstrated low-level engraftment within the NOD/SCID-β2m-/- xenotransplantation model as compared to unfractionated canine bone marrow, which was indicative of suboptimal activation of quiescent canine SP cells within the murine bone marrow niche. A second source of transplantable canine stem cells was examined through the derivation of canine embryonic stem cells (cESCs). The cESC lines described herein were determined to have similar pluripotent stem cell characteristics to human embryonic stem cells, in that they were maintained in an undifferentiated state upon extended passaging as determined by their expression of the human stem cell markers, OCT3/4, NANOG, SOX2, SSEA3, SSEA4, TRA1-60, TRA1-81 and alkaline phosphatase. In addition, cESCs could be induced to differentiate to cells of the three germ layers within in vitro embryoid body cultures and adherent differentiation cultures. Importantly, these cESC lines were the first reported to differentiate in vivo within teratomas. One method of transplanting stem cells to canine recipients involves the delivery of donor cells to the yolk sacs of developing fetuses in utero. Utilizing cells labeled with supraparamagnetic particles conjugated to a Dragon Green fluorophore and the intracellular fluorescent dye, CMTMR, donor cells were tracked from the yolk sac injection site to fetal tissues after transplantation in early (day-25) and mid (day-35) gestation canine fetuses. Labeled cells were localized primarily to the fetal liver and developing bone marrow cavities when examined at gestational day 32, and had been redistributed to not only the fetal liver and bone marrow by day 42, but also to nonhematopoietic tissues, including the lungs and hearts. No labeled cells were detected within the yolk sacs of transplanted fetuses at either time point. These studies demonstrated the efficacy of yolk sac in utero transplantation for the delivery of donor cells to fetal tissues. Collectively, these results indicate that canine stem cells with characteristics similar to human can be isolated and their engraftment, proliferation and differentiation may be assessed in future studies utilizing the canine in utero transplantation model employing yolk sac delivery.
20

Rôle de la niche mésenchymateuse dans la régulation du phénotype SP des progéniteurs hématopoïétiques humains / Role of the mesenchymal niche in SP phenotype regulation of human hematopoietic progenitors

Malfuson, Jean-Valère 05 June 2013 (has links)
L’hématopoïèse est un processus finement régulé pour permettre sa pérennité et son adaptation aux contraintes physiologiques et pathologiques. Ce potentiel repose en grande partie sur les capacités de quiescence, auto-renouvellement, division asymétrique et multipotence des cellules souches hématopoïétiques (CSH). Les CSH et progéniteurs hématopoïétiques (CSPH) sont principalement régulés de façon extrinsèque au sein des niches hématopoïétiques médullaires et cette régulation fait intervenir, des contacts intercellulaires et des facteurs diffusibles. Le phénotype « side-population » (SP), secondaire à l’efflux actif d’un colorant fluorescent (Hoechst 33342) par des pompes de type multidrugresistance, est une caractéristique des cellules souches de la plupart des tissus. Au sein de l’hématopoïèse, le phénotype SP est un excellent moyen pour identifier les CSH murines et est associé à leur quiescence et à leur adhésion à la niche endostéale, mais sa valeur comme marqueur des CSH est plus discutée chez l’homme. Les cellules SP, de par leur nature, sont également étudiées en oncologie, et sont associées aux cellules tumorales les plus résistantes et les plus tumorogènes. La compréhension des mécanismes régulant la fonctionnalité SP devrait permettre d’ouvrir des pistes en physiologie quand à la compréhension de la régulation des CSPH par les niches mésenchymateuses et en pathologie pour cibler les mécanismes de chimiorésistance.Dans ce travail nous montrons pour la première fois chez l’homme que l’acquisition du phénotype SP est un phénomène dynamique et versatile sous le contrôle du stroma médullaire. Le stroma médullaire est en effet capable de maintenir le phénotype SP de CSPH médullaires et d’induire le phénotype SP de CSPH circulants. L’acquisition du phénotype SP par les cellules circulantes nécessite à la fois un « nichage » au sein du stroma et des facteurs diffusibles. Les cellules circulantes capables d’acquérir le phénotype SP contiennent des CSPH au regard de (i) leur expression du CD34, (ii) leur richesse en cellules quiescentes, (iii) leur capacité clonogénique et proliférative en cultures secondaires, (iv) leur expression des gènes de « nichage » et de « souchitude », (v) leur capacité de migration en réponse à un gradient de CXCL12, (vi) leur activité LT-SRC in vivo. De plus nous avons mis en évidence, au sein de ces CSPH SP+CD34+ révélés par le stroma médullaire, une sous-population CD44-/faible qui pourrait contenir les cellules plus immatures en raison de sa quiescence et de l’intensité de son efflux du Hoechst 33342. Les études mécanistiques montrent que l’acquisition du phénotype SP par les cellules circulantes est sous la dépendance de l’intégrine VLA-4 et du CD44. La transduction du signal implique des protéines G et la famille des Src-kinases. Nous montrons également que le stroma médullaire peut induire/maintenir/amplifier la fonctionnalité SP de blastes circulants de leucémie aigüe myéloblastique de façon ß1-intégrine dépendante et que cette fonctionnalité est associée à une capacité d’efflux de Mitoxantrone. Ce mécanisme de modulation de l’activité d’ABC-transporteurs par l’adhésion au stroma correspond à un mécanisme encore jamais décrit de CAM-DR. / Hematopoiesis is a finely tuned process to allow its long-term efficiency and its adaptation to various physiological and pathological stresses. Hematopoietic stem cell (HSC) is the keystone of hematopoiesis through its multipotency, quiescence, asymmetrical division and self-renewing properties. HSC bone marrow (BM) niches mainly regulate hematopoietic stem and progenitor cells (HSPC) through intercellular contacts and diffusible factors. Side-population (SP) cells are characterized by their capability to actively efflux Hoechst 33342 dye through multidrug resistance-like pumps. SP phenotype is a characteristic of stem cells in many tissues and especially, it is a stringent criterion to purify murine HSCs. In mice, this phenotype has been demonstrated to be related to quiescence and resistance to drugs/environmental stresses and to be controlled by endosteal niche adhesion. SP cells are also studied in oncology and are associated to chemo-resistance and tumor initiating capacity. At steady state, SP cells are mainly present in the BM and are mostly absent from the circulation except in stress conditions, raising the hypothesis of the versatility of the SP functionality. Therefore, studying SP phenotype regulation is of importance to understand how BM niches regulate HSPC and how to interfere with cancer cells chemo-resistance.In this work, we demonstrate for the first time and in human that SP phenotype acquisition is a dynamic phenomenon under control of stromal BM cells. Stromal cells from healthy donors maintain SP phenotype of BM HSPC and promote SP phenotype acquisition in circulating ones. SP phenotype promotion depends of stroma nesting and of diffusible factors secretion. This stroma-induced circulating SP cell fraction contains HSPC, as ascertained by (i) CD34 expression, (ii) proportion of cells in G0, (iii) clonogenic and proliferative potential, (iv) nesting and “stemness” gene expression, (v) CXCL12-related migration capability and (vi) LT-SRC activity. Moreover, we describe an SP+CD34+CD44-/low sub-population that could contain most immature HSPCs with regards to their quiescence and Hoechst efflux intensity. Mechanistic studies show that the stoma-mediated SP promoting effect is VLA-4/4ß1-integrin and CD44 dependent, and implicate G-protein and Src-kinase pathways. We also demonstrate that BM stroma from healthy donors can induce/maintain/amplify in a ß1-integrin dependent manner an SP sub-population with mitoxantrone efflux capability in blast cells from acute myeloid leukemia. The existence of a similar mechanism in circulating leukemic blasts suggests the possibility to interfere with the chemo-resistant phenotype of blast cells through integrin/CD44 axis blockade.

Page generated in 0.0565 seconds