• Refine Query
  • Source
  • Publication year
  • to
  • Language
  • 33
  • 4
  • 3
  • 3
  • 1
  • Tagged with
  • 49
  • 19
  • 14
  • 9
  • 8
  • 7
  • 7
  • 6
  • 6
  • 6
  • 5
  • 5
  • 5
  • 5
  • 5
  • 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.
31

The role of Src homology 2 domain containing 5' inositol phosphatase 1 (SHIP) in hematopoietic cells

Desponts, Caroline 01 June 2006 (has links)
The principal isoform of Src homology (SH) 2-domain containing 5' inositol phosphatase protein 1 (SHIP) is a 145kDa protein primarily expressed by cells of the hematopoietic compartment. The enzymatic activity of SHIP is responsible for hydrolyzing the 5' phosphate of phosphatidylinositol-3,4,5-trisphosphate (PI(3,4,5)P3), and thereby preventing the recruitment of pleckstrin homology domain containing effector proteins. Furthermore, SHIP contains protein-protein interaction domains, such as an SH2 domain, two NPXY and several proline-rich motifs. All of these different domains endow SHIP with the capacity to impact signaling pathways important for proliferation, survival, differentiation and activation. Therefore, we hypothesized that SHIP-deficiency could result in the loss of hematopoietic cell homeostasis and functionTo this verify this hypothesis, we first studied the effect of SHIP ablation on hematopoietic stem cell (HSC) proliferation, survival, function and hom ing. Most interestingly we observed that SHIP impacts HSC homeostasis and their ability to home appropriately to the bone marrow. Then, since SHIP was shown to be activated after engagement of the c-mpl receptor by its ligand, thrombopoietin, we studied the impact of SHIP deletion on the function of megakaryocytes, the major target cell of that cytokine. We found that SHIP is also important for homeostasis of the megakaryocyte compartment. Thirdly, we studied the role of SHIP in natural killer (NK) cells biology. We observed that F4 generation SHIP-/- mice have increased NK cells in their spleen and that these cells exhibit a disrupted receptor repertoire. We verified the hypothesis that SHIP helps shape the receptor repertoire of NK cells, mainly through regulation of cell survival and proliferation. Also included, is a study on the role of a SHIP isoform lacking the SH2-domain, called stem cell-SHIP (s-SHIP) in the biology of embryonic stem (ES) cells. To date, this isoform i s expressed by stem/progenitor cells and not by normal differentiated cells. Due to its specific expression pattern, s-SHIP has the potential to have an important role in stem cell biology.
32

Molecular regulation of Megakaryopoiesis: the role of Fli-1 and IFI16

Johnson, Lacey Nicole, St George Clinical School, UNSW January 2006 (has links)
Megakaryocytes (Mks) are unique bone marrow cells, which produce platelets. Dysregulated Mk development can lead to abnormal platelet number and the production of functionally defective platelets, causing bleeding, thrombotic events, and leukaemia. Understanding the molecular mechanisms driving megakaryopoiesis may yield insights into the molecular genetics and cellular pathophysiology of a diversity of disorders. The primary aim of this thesis was to gain insight into the molecular events required for normal Mk development. As transcription factors and cytokines play a central role in driving Mk development, both of these processes were investigated. Fli-1 and GATA-1 are key transcription factors regulating Mk-gene expression, alone and co-operatively. To understand the mechanism of transcriptional synergy exerted by Fli-1 and GATA-1, in vitro assays were carried out investigating the interactions between Fli-1, GATA-1 and DNA that mediate synergy. A novel mechanism of synergy was identified, where Fli-1 DNA binding is not required, although an interaction between Fli-1 and GATA-1, and GATA-1 DNA binding is required. Importantly, the results demonstrate that Fli-1 DNA binding is not essential for promoting Mk-gene expression in primary murine bone marrow cells. Thrombopoietin (TPO) is the primary cytokine responsible for Mk and platelet development. Identifying novel TPO gene-targets may provide invaluable information to aid the understanding of the complex and unique processes required for Mk development. Using microarray technology, IFI16 was identified as a TPO-responsive gene that has not previously been studied in the Mk lineage. This work demonstrated that IFI16 is expressed in CD34+ HSC-derived Mks, and that the Jak/STAT pathway is essential for the activation of IFI16 by both TPO and IFN-??. Of biological significance, IFI16 was found to regulate both the proliferation and differentiation of primary Mks, suggesting that IFI16 may control the balance between these two essential processes. In conclusion, the data in this thesis presents a novel mechanism through which Fli-1 and GATA-1 regulate the synergistic activation of Mk genes. The identification and functional characterisation of a novel TPO-inducible gene, IFI16, involved in regulating the proliferation and differentiation of Mks is also described. These findings have implications for several congenital and malignant conditions affecting Mk and platelet development, and possibly a mechanism for IFN-induced thrombocytopaenia.
33

The diverse role of laminin isoforms in neuronal cells, human mast cells and blood platelets /

Sime, Wondossen, January 2007 (has links)
Diss. (sammanfattning) Stockholm : Karolinska institutet, 2007. / Härtill 4 uppsatser.
34

The role of Src homology 2 domain containing 5' inositol phosphatase 1 (SHIP) in hematopoietic cells /

Desponts, Caroline. January 2006 (has links)
Dissertation (Ph.D.)--University of South Florida, 2006. / Includes vita. Includes bibliographical references (leaves 154-187).. Also available online.
35

Cytokine receptor-like factor 3 (CRLF3) : a novel regulator of platelet biogenesis and potential drug target for thrombocythaemia

Bennett, Cavan January 2018 (has links)
Thrombocythaemia is defined as a circulating platelet count above 450x10$^9$/L in humans. The major cause of thrombocythaemia is reactive $(secondary)$ thrombocythaemia which occurs secondary to many conditions such as infection, cancer and inflammation. However, acquired clonal mutations in mainly Janus Kinas 2 $(JAK2)$, CALR and MPL cause essential thrombocythaemia $(ET)$. ET is a rare disease that leads to an increased risk of cardiovascular thrombotic events. Current treatment of ET uses combination of low dose aspirin to decrease platelet function and cytoreductive agents to decrease thrombopoiesis. The most commonly used cytoreductive agents are hydroxyurea, anagrelide and interferon-$alpha$ and all have unwanted side effects. Cytokine receptor-like factor 3 $(CRLF3)$ is a 2.4kb gene that is ubiquitously expressed throughout the haematopoietic system. Very little is known about the function of CRLF3, with only one peer reviewed journal article in the literature which shows that CRLF3 may negatively regulate the cell cycle at the G0/G1 phase. However, nothing is known about the role of CRLF3 in platelet biology. Using a Crlf3 knockout mouse $(Crlf3-/-)$ developed by the Wellcome Trust Sanger Institute we show CRLF3’s role in platelet biogenesis and how it could be used as a novel therapeutic target to treat ET. Crlf3-/- mice have an isolated and sustained 25-40$\%$ decrease in platelet count compared to wildtype $(WT)$ controls. Platelet function is unaffected as demonstrated in a range in a range of in vitro assays. The thrombocytopenia is a consequence of abnormalities in hematopoietic cells, as shown by bone marrow transplantations. Megakaryopoiesis is upregulated in Crlf3-/- mice and proplatelet morphology is unaffected, suggesting the thrombocytopenia is due to increased platelet clearance. Indeed, splenectomised Crlf3-/- mice show normalised platelet counts within 7 days, showing rapid splenic removal of platelets is responsible for the thrombocytopenia. Abnormal large platelet structures that resemble proplatelets shafts $(preplatelets)$ are abnormally present in the circulation of elderly Crlf3/- mice. Immunohistochemistry showed increased and aberrant tubulin expression in Crlf3-/- platelets compared to WT controls, especially in the preplatelet forms. Cold induced depolymerisation of microtubules was decreased in Crlf3-/- platelets, suggestive of increased tubulin stability, however, the ratio of detyrosinated to tyrosinated tubulin was not altered. We then crossbred Crlf3-/- mice with JAK2 V617F ET mice, to determine the effect of Crlf3 ablation of thrombocythaemia. Crossbred mice showed restoration of platelet counts to WT values without grossly affecting platelet function or other blood lineages, providing the rational for CRLF3 as a novel therapeutic target for treatment of ET. Finally, we aimed to resolve the crustal structure of CRLF3 and discover its interactome. To this end, we were able to resolve the crystal structure of a C-terminal portion of the full length protein containing the predicted fibronectin type III domain. To shed light on the interactome of CRLF3, endogenous CRLF3 was tagged with a tandem affinity purification $(TAP)$ tag using CRISPR/Cas9 technology in induced pluripotent stem cells $(iPSCs)$. We have been able to produce megakaryocytes from these TAP-tagged iPSCs by forward programming. However, as yet we have not been able to generate enough MKs to have adequate material to perform immunoprecipitation assays. Therefore, the interactome of CRLF3 in MKs remains unknown. In conclusion, we identified a mechanism by which Crlf3 controls platelet biogenesis. Slowed maturation of Crlf3-/- preplatelets in the peripheral circulation potentially due to increased structural stability leads to rapid removal of these immature forms by the spleen and therefore a decrease in platelet count. The isolated effect on platelet numbers and normalisation of platelet count in ET mice deficient of Crlf3 provides the rational for further study on CRLF3 drug targeting as a novel therapeutic strategy for ET.
36

Forward programming of human pluripotent stem cells to a megakaryocyte-erythrocyte bi-potent progenitor population : an in vitro system for the production of platelets and red blood cells for transfusion medicine

Dalby, Amanda Louise January 2018 (has links)
There exists a need to produce platelets in vitro for use in transfusion medicine, due to increased platelet demands and short shelf life. Our lab uses human induced pluripotent stem cells (iPSCs), as an attractive alternative supply, as iPSCs can be cultured indefinitely and differentiate into almost any cell type. Using a technique called forward programming, we over express three key haematological transcription factors (TFs), pushing iPSCs towards the megakaryocyte lineage, to produce mature megakaryocytes, the platelet precursor cell type. A major limitation of the forward programming technique is a reliance of lentiviral transduction to overexpress the three TFs, which leads to a number of issues including heterogeneity and high experimental costs. To overcome this, I have developed an inducible iPSC line by inserting the forward programming TFs into a genomic safe harbour, using genome editing techniques. TF expression is strictly controlled, with the TFs expressed only after chemical induction. Inducing forward programming is an efficient method for producing mature megakaryocytes and these cells maintain higher purity in long-term cultures, when compared to cells produced by the lentiviral method. Removing the requirement of lentiviral transduction is a major advancement, making forward programming more amenable to scaling-up, thus moving this technology closer towards our goal of producing in vitro platelets for use in transfusion medicine. I have also shown that forward programming generates a bi-potent progenitor population, from which erythroblasts can be generated, by altering only media conditions. As for megakaryocyte cultures, inducing forward programming improves the purity of erythroblasts produced, compared to the lentiviral method. I have developed single cell progenitor assays combined with index sorting of different cell surface markers, to allow retrospective analysis of cells which successfully generate colonies. The aim of this work is to better characterise the progenitor cells produced by forward programming, to allow further study of this cell type. Single cell RNA-seq of megakaryocytes revealed heterogeneity in long-term cultures and also identified novel candidate surface markers that may help to further characterise the progenitor cell population.
37

Avaliação da infecção de megacariócitos e plaquetas pelo VHC e sua influência na fisiopatologia da hepatite C

Watanabe, Caroline Mitiká January 2016 (has links)
Orientador: Paulo Eduardo de Abreu Machado / Resumo: A hepatite C acomete cerca de 130-150 milhões de pessoas em todo o mundo, sendo que grande parcela dos portadores do vírus da hepatite C (VHC) permanecem assintomáticos por longos períodos. O diagnóstico da doença acontece muitas vezes devido a sintomas extra-hepáticos como fadiga crônica, alterações endócrinas, dermatológicas e hematológicas, porém, a patogênese das manifestações extra-hepática é pouco conhecida. Assim, modelos que reproduzam a infecção in vitro pelo VHC se tornam necessários para que se possa compreender e esclarecer a relação entre estas desordens e o VHC. O VHC é um Hepacivírus da família Flaviviridae, sua entrada em células suscetíveis, como os hepatócitos, pode ocorrer por infecção direta, mediada principalmente pelos receptores CD81 e Claudina-1 (CLDN1), desencadeando uma série de processo para internalização e replicação viral, ou por infecção por contato célula-a-célula, mediada por CLDN1 e Ocludina (OCLN), não sendo necessário, por esta via, CD81. Estudos evidenciam a interação entre plaquetas e VHC, no entanto, não demonstram claramente se estas células somente aderem às partículas virais ou se são infectadas pelo vírus. Assim, estudos que contribuam à elucidação deste processo são salutares. Desta forma, o objetivo deste trabalho foi avaliar a infecção de megacariócitos e plaquetas pelo VHC e verificar a influência na fisiopatologia da hepatite C. Amostras de megacariócitos, provenientes de doadores de medula óssea, e amostras de plaquetas perif... (Resumo completo, clicar acesso eletrônico abaixo) / Abstract: Hepatitis C affects about 130-150 million people worldwide. It´s caused by Hepacivirus and the diagnosis are by extrahepatic symptoms such as chronic fatigue, endocrine, dermatologic and hematologic changes. However, the pathogenesis of extrahepatic manifestations is little known and requires further studies on the relationship of these disorders and the hepatitis C vírus (HCV), therefore infection in vitro model can improve this kind of knowledge. HCV is a family of Flaviviridae, its entry into susceptible cells, such as hepatocytes, can occur by direct infection mediated primarily by CD81 receptors and Claudin-1 (CLDN1), triggering a process serie for internalization and viral replication ocurr. Other road is by cell-to-cell mediated CLDN1 and occludin (OCLN), not being necessary the presence of CD81. Studies describe the interaction between platelets and HCV, however, does not clearly denotes how this process happens, due to this, studies can contribute to the elucidation of this process relevance. The aim of this study is the evaluation of the infection of megakaryocytes and platelets HCV and the influence in pathophysiology of hepatitis C. Megakaryocytes samples, from bone marrow donors, and samples of peripheral platelets, both obtained from healthy donos are infected in vitro with HCV positive plasma. Infected samples were evaluated by flow cytometry and confocal microscopy. The parameters analyzed were the presence or absence of viral and expression of CLDN1 and CD81... (Complete abstract click electronic access below) / Mestre
38

Apport de pathologies plaquettaires rares à la compréhension des rôles de CalDAG-GEFI et des kindlines dans l'activation de l'intégrine αIIbß3

Ghalloussi, Dorsaf 15 March 2016 (has links)
L’étude de l’identification des défauts moléculaires mis jeu dans les pathologies héréditaires plaquettaires est d’un apport considérable pour améliorer la compréhension des mécanismes physiologiques. Durant ma thèse, j’ai étudié les plaquettes d’individus appartenant à deux familles distinctes souffrant de dysfonctions plaquettaires à l’origine d’hémorragies sévères. Par séquençage entier des exons, nous avons identifié pour la première famille une mutation du gène RASGRP2 à l’origine de la substitution Gβ48W empêchant l’activation de CalDAG-GEFI. Les plaquettes des individus porteurs de la mutation à l’état homozygote ont une capacité réduite à activer Rap1 et l’intégrine αIIbß3 en réponse à de faibles doses d'agonistes. La présence d'un allèle non muté (hétérozygotie) est suffisante pour prévenir lessaignements mais ne permet pas de rétablir totalement une fonction plaquettaire normale. La deuxième famille est porteuse d’une mutation du gène FERMT3 (pN54RfsX142) conduisant à une absence complète de kindline-3. Les plaquettes homozygotes pour cette mutation sont incapables d’activer l’intégrine αIIbß3. Elles forment des filopodes et desnodules d’actine mais ne peuvent étendre des lamellipodes même en présence de Mn2+. La kindline-3 s’est révélée essentielle à la régulation de l’activité de Cdc4β et au réarrangement au cytosquelette d'actine lors de la signalisation «outside-in» de l’intégrineαIIbß3. Seule la kindline-3 a jusqu’ici été impliquée dans l'activation des intégrinesplaquettaires. Nous mettons en évidence la présence de kindline-2 dans les plaquettes et les mégacaryocytes humains. Des localisations différentes ont été mises en évidence pour ces deux kindlines. Dans le mégacaryocyte la kindline-2 se situe dans les zones d’adhérence focales et s’associe préférentiellement avec les intégrines ß3. Dans les plaquettes, seule la kindline-3 est présente dans nodules d’actine. Ces résultats sont en faveur de rôles non redondants des kindlines-2 et -γ et d’une implication potentielle de la kindline-2 dans la mégacaryopoïèse. / Inherited platelet disorders are rare diseases that give rise to severe bleeding when platelets fail to fulfill their hemostatic function upon vessel injury. Identifying the molecular mechanisms involved brings important insight into platelet pathophysiology. During my PhD, I studied platelets isolated from members of two families suffering severe bleedings among those one had no established diagnosis. In the first family, using whole exome sequencing, we identified a RASGRP2 mutation causing a G248W substitution leaving CalDAG-GEFI inactive. Platelets from individualscarrying the mutation exhibit a reduced ability to activate Rap1 and to perform proper Inherited platelet disorders are rare diseases that give rise to severe bleeding when platelets fail to fulfill their hemostatic function upon vessel injury. Identifying the molecular mechanisms involved brings important insight into platelet pathophysiology. During my PhD, I studied platelets isolated from members of two families suffering severe bleedings among those one had no established diagnosis. In the first family, using whole exome sequencing, we identified a RASGRP2 mutation causing a G248W substitution leaving CalDAG-GEFI inactive. Platelets from individuals carrying the mutation exhibit a reduced ability to activate Rap1 and to perform proper αIIbß3 integrin inside-out signaling in response to low doses agonists. The presence of a single normal allele is sufficient to prevent bleeding but does not allow normal platelet function. integrin inside-out signaling in response to low doses agonists. The presence of a single normal allele is sufficient to prevent bleeding but does not allow normal platelet function. Members of the second family carry a FERMT3 mutation leading to a completekindlin-3 deficiency (pN54RfsX142). Platelets from the homozygous patient are unable to perform proper integrin αIIbß3 activation. We now observe that kindlin-3 deficient platelets form filipodia and actin nodules but are unable to extend lamellipodia even in presence of Mn2+. We demonstrate that kindlin-3 is essential for Cdc42 activity regulation and actincytoskeleton remodeling during αIIbß3 integrin outside-in signaling To date, only the kindlin-3 has been involved in integrin activation. We show that kindlin-2 is present in human platelets and megakaryocytes. Both kindlins exhibit distinctlocalizations. In megakaryocytes, kindlin-2 specifically localizes within focal adhesion and associates preferentially with ß3 integrins. In platelets, unlike kindline-2, kindline-3 is located in actin nodule. All together these data argue in favor of specific roles played by each kindlins and a possible implication of kindlin-2 in megakaryocytopoiesis.
39

Role of NFAT (Nuclear Factor of Activated T Cells) Transcription Factors in Hematopoiesis: Role of NFAT (Nuclear Factor of Activated T Cells) Transcription Factors in Hematopoiesis

Arabanian, Laleh Sadat 07 November 2012 (has links)
Understanding the transcriptional mechanisms that control hematopoiesis and the interaction between hematopoietic stem cells and the bone marrow (BM) microenvironment in vivo is of considerable interest. The calcineurin-dependent transcription factor NFAT (Nuclear Factor of Activated T cells) is known as master regulator of cytokine production in T lymphocytes and therefore central for T cell-dependent immune reactions, but has also been shown to regulate a process of differentiation and tissue adaptation in various cell types. The activation of NFAT is dependent on the calcium level within the cell. In resting cells, calcium levels are low and NFAT is cytoplasmic and inactive. A sustained increase in the internal calcium concentration within an external stimuli leads to activation of the calcium-dependent calcineurin, followed by dephosphorylation and nuclear translocation of NFAT. We have previously shown that NFATc2, a member of the NFAT family, is expressed in CD34+ hematopoietic stem cells (HSC). A mouse model harboring NFATc2 deficiency provides the opportunity for in vivo investigation of the role of NFATc2 in hematopoiesis. Our recent observations showed that aged mice lacking the transcription factor NFATc2 develop peripheral blood anemia and thrombocytopenia, BM hypoplasia and extramedullary hematopoiesis in spleen and liver. The proliferation and differentiation of NFATc2-deficient hematopoietic stem cells ex vivo, however, was found to be intact. It remained therefore unclear whether the disturbed hematopoiesis in NFATc2-deficient mice was caused by the hematopoietic or the stroma component of the BM hematopoietic niche. In the current study we dissected the relative contribution of hematopoietic and stroma cells to the phenotype of the NFATc2-deficent mice by transplanting immuno-magnetically purified NFATc2-deficient (KO) HSCs to lethally irradiated wild type (WT) mice, and vice versa. After a post-transplantation period of 6-8 months, peripheral blood, BM as well as spleen and liver of the transplanted animals were analyzed and compared to WT and KO mice transplanted with control cells. Transplantation of NFATc2-deficient HSCs into WT recipients (KO WT) induced similar hematological abnormalities as those occurring in non-transplanted KO mice or in KO mice transplanted with KO cells (KO KO). Compared to WT mice transplanted with WT cells (WT WT), KO WT mice showed evidence of anemia, thrombocytopenia and a significantly reduced number of hematopoietic cells in their BM. Likewise, KO WT mice developed clear signs of extramedullary hematopoiesis in spleen and liver, which was not the case in WT WT control animals. In addition to the hematopoietic abnormalities, transplantation of NFATc2-deficient HSC also induced osteogenic abnormalities such as BM sclerosis and fibrosis in WT mice. This phenomenon was rather subtle and of incomplete penetrance, but never seen in mice transplanted with WT cells. These data demonstrate for the first time, that the NFATc2 transcription factor directly regulates the intrinsic function of hematopoietic stem cells in vivo. However, the transcriptional targets for NFAT in these cells are yet unknown. In addition to hematopoietic stem cells, NFATc2 has been shown to be expressed in a lineage-specific manner during myeloid differentiation and, notably, is maintained during megakaryopoiesis while it is suppressed during the differentiation of neutrophils. Bone marrow megakaryocytes are the precursors of peripheral blood platelets and therefore constitute an integral part of primary hemostasis, thrombosis and wound healing. The biological role of NFAT in megakaryocytes is unknown. We have recently shown that NFATc2 is not necessary for megakaryocytic differentiation. On the other hand, recent evidence suggests that NFATc2 is required for the transcription of specific megakaryocytic genes. In this study, we showed that activation of the calcineurin/NFAT pathway in either primary megakaryocytes or CMK megakaryocytic cells forces the cells to go into apoptosis. Cell death in megakaryocytes is induced by treating the cells with the calcium ionophore ionomycin and suppressed by either the pan-caspase inhibitor zVAD or the calcineurin inhibitor cyclosporin A (CsA). Ionomycin stimulation of megakaryocytes leads to the expression of Fas Ligand (FASLG), a pro-apoptotic member of the tumor necrosis factor superfamily. Expression of FASLG was detectable as early as four hours after stimulation on the membrane of ionomycin-treated megakaryocytes, was augmented in cells stably overexpressing NFATc2, and was suppressed in cells either pretreated with CsA or expressing the specific peptide inhibitor of NFAT, VIVIT. To investigate the physiological relevance of FASLG expression on megakaryocytes, we performed co-cultures of megakaryocytes with Fas-expressing T-lymphocytes, in which CMK cells were left either unstimulated or pre-stimulated with ionomycin and then added to Jurkat cells. The presence of ionomycin-stimulated CMK cells, but not of unstimulated cells or cells stimulated in the presence of CsA, significantly induced apoptosis in Jurkat cells. Overexpression of NFATc2 in CMK cells enhanced their potency to induce apoptosis in Jurkat cells, while cells expressing VIVIT were less effective. Apoptosis induction of Jurkat cells by stimulated CMK cells was partially blocked by the presence of either a neutralizing antibody against FASLG or an antagonistic antibody to Fas during the co-culture period, indicating involvement of the FASLG/Fas apoptosis pathway. These results represent the first clear evidence for a biological function of the calcineurin/NFAT pathway in megakaryocytes, namely the regulation of Fas/FASLG-dependent apoptosis. Second, they underline that the biological role of megakaryocytes is not restricted to the production of proteins and other cellular structures for platelet assembly, but that this population of cells fulfills an independent regulatory function in the context of the surrounding tissue. Finally, we have identified by RNA sequencing analysis of NFATc2-expressing and -deficient cells, the entire set of genes which is induced by NFATc2 in stimulated megakaryocytes. Functional pathway analysis suggests an involvement of NFATc2 in pro-inflammatory pathways in these cells. The significance of these findings has to be addressed in further studies.
40

Identification of Hox Genes Controlling Thrombopoiesis in Zebrafish

Sundaramoorthi, Hemalatha 12 1900 (has links)
Thrombocytes are functional equivalents of mammalian platelets and also possess megakaryocyte features. It has been shown earlier that hox genes play a role in megakaryocyte development. Our earlier microarray analysis showed five hox genes, hoxa10b, hoxb2a, hoxc5a, hoxc11b and hoxd3a, were upregulated in zebrafish thrombocytes. However, there is no comprehensive study of genome wide scan of all the hox genes playing a role in megakaryopoiesis. I first measured the expression levels of each of these hox genes in young and mature thrombocytes and observed that all the above hox genes except hoxc11b were expressed equally in both populations of thrombocytes. hoxc11b was expressed only in young thrombocytes and not in mature thrombocytes. The goals of my study were to comprehensively knockdown hox genes and identify the specific hox genes involved in the development of thrombocytes in zebrafish. However, the existing vivo-morpholino knockdown technology was not capable of performing such genome-wide knockdowns. Therefore, I developed a novel cost- effective knockdown method by designing an antisense oligonucleotides against the target mRNA and piggybacking with standard control morpholino to silence the gene of interest. Also, to perform knockdowns of the hox genes and test for the number of thrombocytes, the available techniques were both cumbersome or required breeding and production of fish where thrombocytes are GFP labeled. Therefore, I established a flow cytometry based method of counting the number of thrombocytes. I used mepacrine to fluorescently label the blood cells and used the white cell fraction. Standard antisense oligonucleotide designed to the central portion of each of the target hox mRNAs, was piggybacked by a control morpholino and intravenously injected into the adult zebrafish. The thrombocyte count was measured 48 hours post injection. In this study, I found that the knockdown of hoxc11b resulted in increased number of thrombocytes and knockdown of hoxa10b, hoxb2a, hoxc5a, and hoxd3a showed reduction in the thrombocyte counts. I then screened the other 47 hox genes in the zebrafish genome using flow sorting method and found that knockdown of hoxa9a and hoxb1a also resulted in decreased thrombocyte number. Further, I used the dye DiI, which labels only young thrombocytes at specific concentrations and observed that the knockdown of hoxa10b, hoxb2a, hoxc5a, hoxd3a, hoxa9a and hoxb1a, lead to a decrease in young thrombocytes; whereas hoxc11b knockdown lead to increase in number of young thrombocytes. Using bromodeoxyuridine, I also showed that there is increase in release of young thrombocytes into peripheral circulation in hoxc11b knockdown fish which suggests that hoxc11b significantly promotes cell proliferation rather effecting apoptosis. In conclusion, I found six hox genes that are positive regulators and one hox gene is a negative regulator for thrombocyte development.

Page generated in 0.0536 seconds