Littérature scientifique sur le sujet « Leukaemia, Stem cells, Hypoxia, HIF »

Créez une référence correcte selon les styles APA, MLA, Chicago, Harvard et plusieurs autres

Choisissez une source :

Consultez les listes thématiques d’articles de revues, de livres, de thèses, de rapports de conférences et d’autres sources académiques sur le sujet « Leukaemia, Stem cells, Hypoxia, HIF ».

À côté de chaque source dans la liste de références il y a un bouton « Ajouter à la bibliographie ». Cliquez sur ce bouton, et nous générerons automatiquement la référence bibliographique pour la source choisie selon votre style de citation préféré : APA, MLA, Harvard, Vancouver, Chicago, etc.

Vous pouvez aussi télécharger le texte intégral de la publication scolaire au format pdf et consulter son résumé en ligne lorsque ces informations sont inclues dans les métadonnées.

Articles de revues sur le sujet "Leukaemia, Stem cells, Hypoxia, HIF"

1

Gezer, Deniz, Amelie V. Guitart, Milica Vukovic, Chithra Subramani, Karen Dunn, Patrick Pollard, Peter J. Ratcliffe, Tessa L. Holyoake et Kamil Kranc. « HIF-1α Is Not Essential For The Establishment Of MLL-Leukaemic Stem Cells ». Blood 122, no 21 (15 novembre 2013) : 3767. http://dx.doi.org/10.1182/blood.v122.21.3767.3767.

Texte intégral
Résumé :
Abstract Haematopoietic stem cells (HSCs) reside in hypoxic niches in the bone marrow (BM) and sustain long-life haematopoiesis. HSCs are largely quiescent, self-renew, undergo apoptosis and generate progenitor cells, which differentiate to multiple blood lineages. The strict regulation of the balance between these fate decisions is essential for haematopoiesis and their dysregulation in HSCs and progenitor cells can result in leukaemic transformation. HSCs and leukemic stem cells (LSCs) are suggested to share the same niche and are in need to adapt to hypoxic conditions. Hypoxia-inducible-factor-1α (HIF-1α) is a key mediator of cellular responses to hypoxia and is important for the maintenance of HSC functions under stressful conditions. Furthermore, in chronic myeloid leukaemia (CML) and acute myeloid leukaemia (AML) HIF-1α is essential for LSC maintenance and ablation or knockdown of HIF-1α leads to exhaustion of established LSCs. The aim of this study was to investigate the requirement for HIF-1α in the generation of pre-LSCs and the establishment of LSCs. To investigate the role of HIF-1α in the generation of pre-LSCs we retrovirally transduced haematopoietic stem and progenitor cells (HSPCs) from either WT or HIF1-αfl/fl Vav-iCre with MLL-ENL retroviruses. Next we performed serial re-plating assays under normoxic and hypoxic conditions to generate pre-LSCs. Surprisingly, WT and HIF-1α deficient HSPCs generated comparable numbers of colonies in normoxia and hypoxia (Fig. 1a). In addition no significant difference was found in the immunophenotypic profile of colonies (Figure 1b). Furthermore, microscopic examination indicated that colonies of all genotypes were dense consistent with their transformed shape (Fig. 1c). WT and HIF-1α-deficient pre-LSCs cultured under normoxia and hypoxia had similar cloning efficiency, which is known to directly correlate with the numbers of LSCs in vivo (Fig. 2). These results indicate that HIF-1α is dispensable for the generation of pre-LSCs. To test the role of HIF-1α in establishment of LSCs from pre-LSCs we transplanted pre-LSCs into lethally irradiated mice together with support BM and monitored the mice for disease development. No significant difference was found in disease latency (Fig. 3a) or frequency of LSCs in peripheral blood, bone marrow or spleens (Fig. 3b) indicating that pre-LSCs lacking HIF-1α can efficiently generate LSCs that cause aggressive AML. In conclusion, we provide genetic evidence that HIF-1α is dispensable for the generation of pre-LSCs and the establishment of LSCs from pre-LSCs. These surprising findings, together with published results indicating that HIF-1α is essential for maintenance of LSCs, imply that HIF-1α has different roles at different stages of leukaemic transformation. Further studies are required to explain the distinct roles of HIF-1α in different stages of leukaemogenesis. Disclosures: Ratcliffe: RedOx: Founder Other. Holyoake:Novartis: Membership on an entity’s Board of Directors or advisory committees; Bristol Myers Squibb: Membership on an entity’s Board of Directors or advisory committees; Ariad: Membership on an entity’s Board of Directors or advisory committees.
Styles APA, Harvard, Vancouver, ISO, etc.
2

Tomita, Mariko, Gregg L. Semenza, Canine Michiels, Takehiro Matsuda, Jun-Nosuke Uchihara, Taeko Okudaira, Yuetsu Tanaka, Naoya Taira, Kazuiku Ohshiro et Naoki Mori. « Activation of hypoxia-inducible factor 1 in human T-cell leukaemia virus type 1-infected cell lines and primary adult T-cell leukaemia cells ». Biochemical Journal 406, no 2 (13 août 2007) : 317–23. http://dx.doi.org/10.1042/bj20070286.

Texte intégral
Résumé :
HTLV-1 (human T-cell leukaemia virus type 1) is the causative agent for ATL (adult T-cell leukaemia). HTLV-1 Tax can activate the PI3K (phosphoinositide 3-kinase)/Akt signalling pathway, which is responsible for survival of HTLV-1-infected T-cells. HIFs (hypoxia-inducible factors) are transcriptional regulators that play a central role in the response to hypoxia. Overexpression of HIF-1α in many cancers is associated with a poor response to treatment and increased patient mortality. Our objectives in the present study were to investigate whether HIF-1 was activated in HTLV-1-infected T-cells and to elucidate the molecular mechanisms of HIF-1 activation by focusing on the PI3K/Akt signalling pathway. We detected a potent pathway that activated HIF-1 in the HTLV-1-infected T-cells under a normal oxygen concentration. Enhanced HIF-1α protein expression and HIF-1 DNA-binding activity were exhibited in HTLV-1-infected T-cell lines. Knockdown of HIF-1α by siRNA (small interfering RNA) suppressed the growth and VEGF (vascular endothelial growth factor) expression of the HTLV-1-infected T-cell line. HIF-1 protein accumulation and transcriptional activity were enhanced by Tax, which was inhibited by dominant-negative Akt. Importantly, mutant forms of Tax that are defective in activation of the PI3K/Akt pathway failed to induce HIF-1 transcriptional activity. The PI3K inhibitor LY294002 suppressed HIF-1α protein expression, HIF-1 DNA-binding and HIF-1 transcriptional activity in HTLV-1-infected T-cell lines. In primary ATL cells, HIF-1α protein levels were strongly correlated with levels of phosphorylated Akt. The results of the present study suggest that PI3K/Akt activation induced by Tax leads to activation of HIF-1. As HIF-1 plays a major role in tumour progression, it may represent a molecular target for the development of novel ATL therapeutics.
Styles APA, Harvard, Vancouver, ISO, etc.
3

Choi, Jong Ho, Yun Bin Lee, Jieun Jung, Seong Gyu Hwang, IL-Hoan Oh et Gi Jin Kim. « Hypoxia Inducible Factor-1αRegulates the Migration of Bone Marrow Mesenchymal Stem Cells via Integrinα4 ». Stem Cells International 2016 (2016) : 1–11. http://dx.doi.org/10.1155/2016/7932185.

Texte intégral
Résumé :
Although hypoxic environments have been known to regulate the migratory ability of bone marrow-derived mesenchymal stem cells (BM-MSCs), which is a critical factor for maximizing the therapeutic effect, the underlying mechanisms remain unclear. Therefore, we aimed to confirm the effect of hypoxia-inducible factor-1α(HIF-1α) on the migration of BM-MSCs and to analyze the interaction between HIF-1αand integrin-mediated signals. Hypoxia-activated HIF-1αsignificantly increased BM-MSC migration. The expression of integrinα4was decreased in BM-MSCs by increased HIF-1αunder hypoxia, whereas the expression of Rho-associated kinase 1 (ROCK1) and Rac1/2/3 was increased. After downregulation of HIF-1αby YC-1, which is an inhibitor of HIF-1α, BM-MSC migration was decreased via upregulation of integrinα4and downregulation of ROCK1 and Rac1/2/3. Knockdown of integrinα4by integrinα4siRNA (siITGA4) treatment increased BM-MSC migration by upregulation of ROCK1, Rac1/2/3, and matrix metalloproteinase-2 regardless of oxygen tension. Moreover, siITGA4 treatment increased HIF-1αexpression and augmented the translocation of HIF-1αinto the nucleus under hypoxia. Taken together, the alternative expression of HIF-1αinduced by microenvironment factors, such as hypoxia and integrinα4, may regulate the migration of BM-MSCs. These findings may provide insights to the underlying mechanisms of BM-MSC migration for successful stem cell-based therapy.
Styles APA, Harvard, Vancouver, ISO, etc.
4

Bernard, Olivier, Florence Jeny, Yurdagül Uzunhan, Elisabetta Dondi, Rahma Terfous, Rabab Label, Angela Sutton et al. « Mesenchymal stem cells reduce hypoxia-induced apoptosis in alveolar epithelial cells by modulating HIF and ROS hypoxic signaling ». American Journal of Physiology-Lung Cellular and Molecular Physiology 314, no 3 (1 mars 2018) : L360—L371. http://dx.doi.org/10.1152/ajplung.00153.2017.

Texte intégral
Résumé :
Distal lung diseases, such as pulmonary fibrosis or acute lung injury, are commonly associated with local alveolar hypoxia that may be deleterious through the stimulation of alveolar epithelial cell (AEC) apoptosis. In various murine models of alveolar injury, administration of allogenic human mesenchymal stem cells (hMSCs) exerts an overall protective paracrine effect, limiting lung inflammation and fibrosis. However, the precise mechanisms on lung cells themselves remain poorly understood. Here, we investigated whether hMSC-conditioned medium (hMSC-CM) would protect AECs from hypoxia-induced apoptosis and explored the mechanisms involved in this cytoprotective effect. Exposure of rat primary AECs to hypoxia (1.5% O2 for 24 h) resulted in hypoxia-inducible factor (HIF)-1α protein stabilization, partly dependent on reactive oxygen species (ROS) accumulation, and in a twofold increase in AEC apoptosis that was prevented by the HIF inhibitor 3-(5′-hydroxymethyl-2′-furyl)-1-benzyl-indazole and the antioxidant drug N-acetyl cysteine. Incubation of AECs with hMSC-CM significantly reduced hypoxia-induced apoptosis. hMSC-CM decreased HIF-1α protein expression, as well as ROS accumulation through an increase in antioxidant enzyme activities. Expression of Bnip3 and CHOP, two proapoptotic targets of HIF-1α and ROS pathways, respectively, was suppressed by hMSC-CM, while Bcl-2 expression was restored. The paracrine protective effect of hMSC was partly dependent on keratinocyte growth factor and hepatocyte growth factor secretion, preventing ROS and HIF-1α accumulation.
Styles APA, Harvard, Vancouver, ISO, etc.
5

Eliasson, Pernilla M., et Jan-Ingvar Jönsson. « A Hypoxic Niche in the Mouse Bone Marrow Diminishes Proliferation and Differentiation of Hematopoietic Stem Cells ». Blood 112, no 11 (16 novembre 2008) : 4777. http://dx.doi.org/10.1182/blood.v112.11.4777.4777.

Texte intégral
Résumé :
Abstract In the bone marrow hematopoietic stem cells (HSCs) reside in specialized niches in close contact with stromal cells and endosteal osteoblasts. It is thought that this environment is hypoxic in nature, where HSCs are maintained in a quiescent state to prevent their depletion. Hypoxia stabilizes the transcription factor HIF-1α which triggers angiogenesis as well as genes slowering the cell cycle, promoting cell survival, and leading to a decrease in cellular metabolism. In this study, hypoxic effects of the maintenance of Lin−Sca1+c-kit+* (LSK) cells derived from mouse bone marrow and the involvement of the transcription factor hypoxia inducible factor 1 α (HIF-1α) were investigated. Hypoxic culture conditions led to an increase in numbers of primitive colony-forming progenitor cells and a preferential expansion of immature blast-like appearing cells. Concurrently, the immature c-kit Sca-1 phenotype was better maintained in hypoxia compared to ambient oxygen levels. Moreover, hypoxia decreased the proliferation of HSCs as measured by CFSE or PKH26 staining. This was confirmed by cell cycle analysis, and hypoxic cultivation decreased the percentage of cells in S-phase whereas cells in G0/G1 phase increased. Cells infected with a constitutively active form of HIF-1α showed the same pattern as cells cultured in hypoxia. To verify that the effect is HIF-1α mediated, we silenced HIF-1α in LSK cells with shRNA. The decrease in proliferation in hypoxic cultivation of cells infected with shRNA against HIF-1α was markedly diminished, indicating that HIF-1α play an important role in controlling proliferation of hematopoietic stem cells. These results suggest that a major function of hypoxia is to counteract proliferation and possibly differentiation, thereby sustaining maintenance. Furthermore, hypoxic culture conditions may have beneficial clinical implications for ex vivo purposes and may improve the yields of stem cells. In our ongoing-studies, we are investigating whether HIF-1α and hypoxia is an absolute prerequisite for the proper maintenance of HSCs in the bone marrow.
Styles APA, Harvard, Vancouver, ISO, etc.
6

Hu, Cheng-Jun, Sangeeta Iyer, Aneesa Sataur, Kelly L. Covello, Lewis A. Chodosh et M. Celeste Simon. « Differential Regulation of the Transcriptional Activities of Hypoxia-Inducible Factor 1 Alpha (HIF-1α) and HIF-2α in Stem Cells ». Molecular and Cellular Biology 26, no 9 (1 mai 2006) : 3514–26. http://dx.doi.org/10.1128/mcb.26.9.3514-3526.2006.

Texte intégral
Résumé :
ABSTRACT Transcriptional responses to hypoxia are primarily mediated by hypoxia-inducible factors (HIFs), HIF-1α and HIF-2α. The HIF-1α and HIF-2α subunits are structurally similar in their DNA binding and dimerization domains but differ in their transactivation domains, implying they may have unique target genes and require distinct transcriptional cofactors. Our previous results demonstrated that HIF-1α and HIF-2α regulate distinct target genes. Here, we report that HIF-2α is not transcriptionally active in embryonic stem (ES) cells, as well as possible inhibition by a HIF-2α-specific transcriptional repressor. Using DNA microarray analysis of hypoxia-inducible genes in wild-type (WT), Hif-1α − / − , and Hif-2α − / − ES cells, we show that HIF-1α induces a large number of both confirmed and novel hypoxia-inducible genes, while HIF-2α does not activate any of its previously described targets. We further demonstrate that inhibition of HIF-2α function occurs at the level of transcription cofactor recruitment to endogenous target gene promoters. Overexpression of WT and, notably, a DNA-binding-defective HIF-2α mutant restores endogenous HIF-2α protein activity, suggesting that ES cells express a HIF-2α-specific corepressor that can be titrated by overexpressed HIF-2α protein. HIF-2α repression may explain why patients with mutations in the VHL tumor suppressor gene display cancerous lesions in specific tissue types.
Styles APA, Harvard, Vancouver, ISO, etc.
7

Tang, Di, Junhui Zhang, Tiantian Yan, Jingyu Wei, Xupin Jiang, Dongxia Zhang, Qiong Zhang, Jiezhi Jia et Yuesheng Huang. « FG-4592 Accelerates Cutaneous Wound Healing by Epidermal Stem Cell Activation via HIF-1α Stabilization ». Cellular Physiology and Biochemistry 46, no 6 (2018) : 2460–70. http://dx.doi.org/10.1159/000489652.

Texte intégral
Résumé :
Background/Aims: Regional hypoxia promptly develops after trauma because of microvascular injury and increased oxygen consumption. This acute hypoxia plays a positive role in early skin wound healing. One of the mechanisms underlying the beneficial effects of acute hypoxia on wound healing may be increased hypoxia-inducible factor-1 (HIF-1α) expression. HIF-1α may affect the wound-healing process through many aspects, including angiogenesis, metabolism, and extra-cellular matrix synthesis and remodelling. Epidermal stem cells (EpSCs) are important participants in wound repair; however, whether these cells are regulated by hypoxia is unclear. This study aimed to elucidate the regulatory mechanism by which hypoxia acts on EpSCs. Methods: CCK8 assays, western blots and live cell station observation were employed to compare the viability, proliferation and motility of EpSCs cultured under normoxic conditions (21% O2) with those cultured under hypoxic conditions (2% O2). Moreover, we used FG-4592 (a prolyl hydroxylase inhibitor that stabilizes HIF-1α in normoxia), KC7F2 (a selective inhibitor of HIF-1α transcription) and siRNA against HIF-1α to regulate HIF-1α expression. Results: Acute hypoxia caused EpSCs to switch from a quiescent state to an activated state with higher viability and motility, as well as an earlier proliferation peak. We demonstrated that the HIF-1 signalling pathway mediated hypoxia-induced activation of EpSCs. Finally, the in vivo experiments showed that exogenous FG-4592 effectively accelerates wound healing, shortens healing times and even induces epidermal hyperplasia. Conclusion: This study demonstrated that both hypoxia and exogenous FG-4592 improve EpSC proliferation and motility by stabilizing HIF-1α, and its results suggest that HIF-1α is an important target through which wound healing can be accelerated and that FG-4592 is a promising new drug for wound repair.
Styles APA, Harvard, Vancouver, ISO, etc.
8

Chang, Yao-Lung, Shuenn-Dyh Chang, An-Shine Chao, Martin Sieber, Chia-Lung Tsai et Po-Jen Cheng. « Effect of Hypoxia on Glucose Transporter 1 and 3 Gene Expression in Placental Mesenchymal Stem Cells Derived from Growth-Restricted Fetuses ». Genes 13, no 5 (25 avril 2022) : 752. http://dx.doi.org/10.3390/genes13050752.

Texte intégral
Résumé :
(1) Background: Glucose is transferred from maternal blood to the fetus by glucose transporters. What is the effect of hypoxia on the gene expression of placenta glucose transporter 1 (GLUT1) and glucose transporter 3 (GLUT3) in growth-restricted fetus is interesting. (2) Methods: The gene expression of GLUT1 and GLUT3 and the protein expression of HIF-1α were evaluated under nonhypoxic conditions and after 4 and 8 h under hypoxic conditions in placental mesenchymal stem cells derived from monochorionic twin pregnancies with selective intrauterine growth restriction. (3) Results: The gene expressions of GLUT1 and GLUT3 under hypoxia conditions were higher in placental mesenchymal stem cells derived from appropriate-for-gestational-age fetuses than in those from selective intrauterine growth-restricted fetuses. However, the protein expression of hypoxia induced factor-1 α (HIF-1α) at hypoxia condition was not lower in placenta mesenchymal stem cells from selective intrauterine growth-restricted fetuses than in placental mesenchymal stem cells from appropriate-for-gestational-age fetuses. (4) Conclusions: Hypoxia-induced upregulation of GLUT1 and GLUT3 expression was decreased in placental mesenchymal stem cells from selective intrauterine growth-restricted fetuses but not due to decreased HIF-1α expression. Selective growth-restricted fetuses have less capacity for hypoxia-induced upregulation of placental glucose transport.
Styles APA, Harvard, Vancouver, ISO, etc.
9

Chang, Yao-Lung, Shuenn-Dyh Chang, An-Shine Chao, Martin Sieber, Chia-Lung Tsai et Po-Jen Cheng. « Effect of Hypoxia on Glucose Transporter 1 and 3 Gene Expression in Placental Mesenchymal Stem Cells Derived from Growth-Restricted Fetuses ». Genes 13, no 5 (25 avril 2022) : 752. http://dx.doi.org/10.3390/genes13050752.

Texte intégral
Résumé :
(1) Background: Glucose is transferred from maternal blood to the fetus by glucose transporters. What is the effect of hypoxia on the gene expression of placenta glucose transporter 1 (GLUT1) and glucose transporter 3 (GLUT3) in growth-restricted fetus is interesting. (2) Methods: The gene expression of GLUT1 and GLUT3 and the protein expression of HIF-1α were evaluated under nonhypoxic conditions and after 4 and 8 h under hypoxic conditions in placental mesenchymal stem cells derived from monochorionic twin pregnancies with selective intrauterine growth restriction. (3) Results: The gene expressions of GLUT1 and GLUT3 under hypoxia conditions were higher in placental mesenchymal stem cells derived from appropriate-for-gestational-age fetuses than in those from selective intrauterine growth-restricted fetuses. However, the protein expression of hypoxia induced factor-1 α (HIF-1α) at hypoxia condition was not lower in placenta mesenchymal stem cells from selective intrauterine growth-restricted fetuses than in placental mesenchymal stem cells from appropriate-for-gestational-age fetuses. (4) Conclusions: Hypoxia-induced upregulation of GLUT1 and GLUT3 expression was decreased in placental mesenchymal stem cells from selective intrauterine growth-restricted fetuses but not due to decreased HIF-1α expression. Selective growth-restricted fetuses have less capacity for hypoxia-induced upregulation of placental glucose transport.
Styles APA, Harvard, Vancouver, ISO, etc.
10

Carcelén, María, Carlos Velásquez, Veronica Vidal, Olga Gutierrez et Jose L. Fernandez-Luna. « HIF2α Upregulates the Migration Factor ODZ1 under Hypoxia in Glioblastoma Stem Cells ». International Journal of Molecular Sciences 23, no 2 (11 janvier 2022) : 741. http://dx.doi.org/10.3390/ijms23020741.

Texte intégral
Résumé :
Background: Glioblastoma (GBM) remains a major clinical challenge due to its invasive capacity, resistance to treatment, and recurrence. We have previously shown that ODZ1 contributes to glioblastoma invasion and that ODZ1 mRNA levels can be upregulated by epigenetic mechanisms in response to hypoxia. Herein, we have further studied the transcriptional regulation of ODZ1 in GBM stem cells (GSCs) under hypoxic conditions and analyzed whether HIF2α has any role in this regulation. Methods: We performed the experiments in three primary GSC cell lines established from tumor specimens. GSCs were cultured under hypoxia, treated with HIF regulators (DMOG, chetomin), or transfected with specific siRNAs, and the expression levels of ODZ1 and HIF2α were analyzed. In addition, the response of the ODZ1 promoter cloned into a luciferase reporter plasmid to the activation of HIF was also studied. Results: The upregulation of both mRNA and protein levels of HIF2α under hypoxia conditions correlated with the expression of ODZ1 mRNA. Moreover, the knockdown of HIF2α by siRNAs downregulated the expression of ODZ1. We found, in the ODZ1 promoter, a HIF consensus binding site (GCGTG) 1358 bp from the transcription start site (TSS) and a HIF-like site (CCGTG) 826 bp from the TSS. Luciferase assays revealed that the stabilization of HIF by DMOG resulted in the increased activity of the ODZ1 promoter. Conclusions: Our data indicate that the HIF2α-mediated upregulation of ODZ1 helps strengthen the transcriptional control of this migration factor under hypoxia in glioblastoma stem cells. The discovery of this novel transcriptional pathway identifies new targets to develop strategies that may avoid GBM tumor invasion and recurrence.
Styles APA, Harvard, Vancouver, ISO, etc.

Thèses sur le sujet "Leukaemia, Stem cells, Hypoxia, HIF"

1

Cochran, David M. Ph D. Massachusetts Institute of Technology. « The role of HIF-1 alpha in the localization of embryonic stem cells with respect to hypoxia within teratomas ». Thesis, Massachusetts Institute of Technology, 2005. http://hdl.handle.net/1721.1/33841.

Texte intégral
Résumé :
Thesis (Ph. D.)--Harvard-MIT Division of Health Sciences and Technology, 2005.
Includes bibliographical references (leaves 172-183).
In embryonic stem (ES) cell tumors, the hypoxia-inducible transcription factor, HIF- 1[alpha], has been shown to be a tumor suppressor, and HIF-1[alpha]-expressing cells have been shown to localize preferentially in vivo to regions near tumor vasculature. These differences were proposed to be due to increased hypoxia-induced apoptosis and growth arrest of HIF-1[alpha]-expressing ES cells. This thesis presents a careful investigation into the localization of ES cells in vitro and in vivo with respect to hypoxia. A sandwich culture system was utilized in which controlled gradients of oxygen and nutrients are developed in the vicinity of the tumor cells. A diffusion-consumption model was utilized to predict the oxygen and glucose concentration profiles within the system. Oxygen and glucose consumption rates were measured and used as inputs into the model, and the concentration profiles were found to depend on a single experimental parameter, the cell density within the system. The optimum cell density was found in which stable, measurable oxygen gradients develop over 2-3 mm. The model demonstrated excellent agreement between the predicted oxygen concentration profiles and experimentally determined oxygen gradients. In vitro, there was no difference in localization with respect to hypoxia between tumor cells expressing or lacking HIF-1[alpha].
(cont.) In addition, there was no difference in apoptosis, proliferation, or migration of the tumor cells in vitro based on HIF-1[alpha] expression. Likewise, a quantitative study on localization of tumor cells within tumors in vivo demonstrated no difference between localization of HIF-1[alpha]-expressing vs. HIF-1[alpha]-lacking ES cells within tumors with respect to blood vessels or hypoxia. These results differ from previous studies, perhaps due to clonal variation of the cell phenotype or the interplay of other complex environmental factors that were not considered in this study. Interestingly, the HIF-1[alpha]-lacking cells were found to exhibit increased tumor growth relative to the HIF-1[alpha]-expressing cells, perhaps due to a normalization of the blood vessels within the HIF-1[alpha]-lacking tumors. These studies reveal the complex role of HIF-1[alpha] in tumor growth and tumor cell localization, as well as develop a useful quantitative experimental model for studying the role of the microenvironment in tumors or in embryonic stem cell biology.
by David M. Cochran.
Ph.D.
Styles APA, Harvard, Vancouver, ISO, etc.
2

Krieg, Marie-Louise. « Impact of Oxygen-Release Material on Human Urine-Derived Stem Cells’ Differentiation and Proliferation in Hypoxic Condition In Vitro ». Thesis, Uppsala universitet, Tillämpad materialvetenskap, 2010. http://urn.kb.se/resolve?urn=urn:nbn:se:uu:diva-126039.

Texte intégral
Résumé :
One of today’s most widely spread health problems is urinary incontinence, affecting 60-80% of the US population from age 15 and up. Treatment based on the possibility to implant a scaffold seeded with the patients’ own urine-derived stem cells, hUSC, to regenerate the damaged muscle tissue, would prove effective. A main challenge in regenerating new tissue from cell-seeded scaffolds is the limited cell survival due to insufficient oxygen diffusion to the center of the scaffold. Ways of enhancing cell survival, and thereby, proliferation and differentiation, is by hypoxic preconditioning of the cells or implantation in an oxygen-release material. Hypoxic preconditioning has shown to enhance proliferation as well as the expression of vascular endothelial growth factor, VEGF, in for example human bone marrow derived stem cells, hBMSC. VEGF is involved in the establishment of vasculature structures and an upregulation of its expression may therefore help promote quicker angeogenisis, increasing the oxygen supply and the cell survival. Oxygen-release materials have shown to enhance cell survival and growth both in vitro and in vivo. This study aims to investigate the effect of hypoxia on hUSC, during 9 days of hypoxic culturing (2.0% ± 0.1% O2) with and without oxygen-release material (PLGA 75:25 with 5 w% CPO) in vitro. hBMSC, and human smooth muscle cells, hSMC, have been used as control groups. Cell proliferation, morphology, differentiation, production of VEGF, and expression of hypoxia inducible factor HIF-1α have been studied. According to the results, combining hypoxic preconditioning of hUSC with implantation in oxygen-release material could be an effective way to regenerate muscular tissue. Hypoxic preconditioning enhanced cell proliferation, production of VEGF, and HIF-1α expression. The increase of VEGF and HIF-1α would promote vascularization when implanted. The oxygen-release material showed possible promotion of cell differentiation, which would augment the hUSCs’ myogenic differentiation, while supplying oxygen until the tissue’s vascular structure has been established.
Styles APA, Harvard, Vancouver, ISO, etc.
3

Wozny, Anne-sophie. « Mécanismes moléculaires spécifiques de la réponse aux ions carbone dans les cellules tumorales (souches et non souches) des cancers des Voies Aéro-Digestives Supérieures ». Thesis, Lyon, 2018. https://n2t.net/ark:/47881/m6m044rb.

Texte intégral
Résumé :
L’hadronthérapie par ions carbone est une alternative à la radiothérapie photonique dans le traitement des cancers des VADS, en raison d’une balistique précise et d’une efficacité biologique élevée, y compris au sein des zones tumorales hypoxiques. Ces cancers sont de mauvais pronostic en raison d’un risque élevé de récidives liées à la présence de cellules souches cancéreuses (CSCs). L’objectif de ce travail était de déterminer les spécificités moléculaires de la réponse aux ions carbone par rapport aux photons dans deux lignées cellulaires de cancer des VADS et leur sous-population CSCs en hypoxie et normoxie. Il s’est focalisé sur le rôle de la protéine HIF-1α dans la survie cellulaire, dans la mesure où l’hypoxie favorise sa stabilisation mais également la radiorésistance; sur la transition épithélio-mésenchymateuse (EMT) et la détection-réparation des cassures double-brin (CDBs) de l’ADN. HIF-1α est stabilisée plus précocement dans les CSCs par rapport aux non-CSCs. Son activation, tout comme celle des voies de l’EMT (STAT3, MEK/p38/JNK et Akt/mTOR) est dépendante des radicaux libres oxygénés (RLO), dont la production est homogène dans la cellule en réponse aux photons. Par contre, les RLO produits dans la trace des ions carbone ne permettent pas d’activer HIF-1α et les voies de l’EMT. Sous hypoxie, une relation a été établie entre l’activation d’HIF-1α et celles des voies de détection (ATM) et de réparation (Rad51) des CDBs (Recombinaison Homologue). Ces travaux démontrent que l’avantage thérapeutique des ions carbone repose sur la répartition spatiale des RLO à l’échelle nanométrique et consécutivement sur la non-activation de voies clés de la défense cellulaire tumorale
Hadrontherapy using carbon ions is an alternative to photon irradiation in the treatment of Head and Neck cancers, because of accurate ballistics and high biological efficiency, including hypoxic tumor areas. These cancers are of poor prognosis because of a high risk of recurrences related to the presence of cancer stem cells (CSCs).The aim of this work was to determine the molecular specificities of the response to carbon ion irradiations compared to photons in two cancer cell lines and their CSCs’ subpopulation, in hypoxic and normoxic conditions. This work focused on the role of the HIF-1α protein in cell survival, since hypoxia promotes its stabilization, but also in the radioresistance; the epithelial-mesenchymal transition (EMT) and the detection and repair of DNA double-strand breaks (DSBs). HIF-1α is stabilized earlier in CSCs compared to non-CSCs. Its activation, as well as the EMT pathways (STAT3, MEK/p38/JNK and Akt/mTOR), are dependent on reactive oxygen species (ROS), whose production is homogeneous in response to photons. At the opposite, the ROS produced in the carbon ion tracks are insufficient to activate HIF-1α and the upstream EMT pathways. Under hypoxic conditions, a relationship has been established between HIF-1α activation and that of the DSBs detection (ATM) and repair (Rad51) pathways (Homologous Recombination). These studies demonstrate that the therapeutic advantage of carbon ions is based on the spatial ROS distribution at the nanoscale and consequently on the non-activation of key pathways involved in tumor cell defense
Styles APA, Harvard, Vancouver, ISO, etc.
4

CHELONI, GIULIA. « Chronic myeloid leukaemia stem cells are sensitive to the pharmacological inhibition of hypoxia inducible factor-1a ». Doctoral thesis, 2015. http://hdl.handle.net/2158/1042876.

Texte intégral
Styles APA, Harvard, Vancouver, ISO, etc.
5

Li, Zhizhong. « Differential Angiogenic Capability and Hypoxia Responses in Glioma Stem Cells ». Diss., 2009. http://hdl.handle.net/10161/1149.

Texte intégral
Résumé :

Malignant gliomas are highly lethal cancers characterized by florid angiogenesis. Glioma stem cells (GSCs), enriched through CD133 (Prominin1) selection, are highly tumorigenic and therapy resistance. However, the mechanism through which GSCs promote tumor growth was largely unknown. As we noticed that tumors derived from GSCs contain widespread tumor angiogenesis, necrosis, and hemorrhage, we examined thepotential of GSCs to support tumor angiogenesis. We measured the expression of a panel of angiogenic factors secreted by GSCs. In comparison with matched non-GSC populations, GSCs consistently secreted markedly elevated levels of vascular endothelial growth factor (VEGF), which were further induced by hypoxia. In an in vitro model of angiogenesis, GSC-conditioned medium significantly increased endothelial cell migration and tube formation compared with non-GSC glioma cell-conditioned medium. The proangiogenic effects of GSCs on endothelial cells were specifically abolished by the anti-VEGF neutralizing antibody bevacizumab, which is in clinical use for cancer therapy. Furthermore, bevacizumab displayed potent antiangiogenic efficacy in vivo and suppressed growth of xenografts derived from GSCs but limited efficacy against xenografts derived from a matched non-GSC population. As hypoxia is a key regulator of angiogenesis, I further examined hypoxic responses in GSCs to determine the molecular mechanisms underlying their angiogenic drive. I demonstrated that multiple hypoxia response genes, including the hypoxia-inducible factors (HIFs)-1a and -2a(EPAS-1) were differentially expressed in GSCs in comparison to non-stem glioma cells and normal neural progenitors. GSCs preferentially induced HIF2a; and HIF2a-regulated genes under hypoxia in comparison to non-stem glioma cells. In contrast, neural progenitor/stem cells did not induce HIF2a in response to hypoxia suggesting that the HIF2a hypoxic response is not a general stem cell response. Targeting HIF1a or HIF2a in GSCs using short hairpin RNA (shRNA) inhibited neurosphere formation efficiency, indicating a requirement for HIFs in cancer stem cell self-renewal. HIF1a and HIF2a were also necessary for VEGF expression in GSCs, but HIF2a was not required in matched non-stem glioma cells. In vivo experiments determined that knockdown of HIFs significantly attenuated the tumorigenic capacity of GSCs and increased survival of immunocompromised mice. Together, our work provides the first evidence that that GSCs can be a crucial source of key angiogenic factors in cancers due to their differential hypoxia responses. It also suggests that anti-angiogenic therapies can be designed to target GSC-specific molecular mechanisms of neoangiogenesis, including the expression and/or activity of HIF2a.


Dissertation
Styles APA, Harvard, Vancouver, ISO, etc.
6

TANTURLI, MICHELE. « Ruolo dell'adattamento all'ipossia di cellule di leucemia mieloide cronica nella loro resistenza al trattamento ». Doctoral thesis, 2011. http://hdl.handle.net/2158/997010.

Texte intégral
Styles APA, Harvard, Vancouver, ISO, etc.
7

Wozny, Anne-Sophie. « Mécanismes moléculaires spécifiques de la réponse aux ions carbone dans les cellules tumorales (souches et non souches) des cancers des Voies Aéro-Digestives Supérieures ». Thesis, 2018. http://www.theses.fr/2018LYSE1120.

Texte intégral
Résumé :
L’hadronthérapie par ions carbone est une alternative à la radiothérapie photonique dans le traitement des cancers des VADS, en raison d’une balistique précise et d’une efficacité biologique élevée, y compris au sein des zones tumorales hypoxiques. Ces cancers sont de mauvais pronostic en raison d’un risque élevé de récidives liées à la présence de cellules souches cancéreuses (CSCs). L’objectif de ce travail était de déterminer les spécificités moléculaires de la réponse aux ions carbone par rapport aux photons dans deux lignées cellulaires de cancer des VADS et leur sous-population CSCs en hypoxie et normoxie. Il s’est focalisé sur le rôle de la protéine HIF-1α dans la survie cellulaire, dans la mesure où l’hypoxie favorise sa stabilisation mais également la radiorésistance; sur la transition épithélio-mésenchymateuse (EMT) et la détection-réparation des cassures double-brin (CDBs) de l’ADN. HIF-1α est stabilisée plus précocement dans les CSCs par rapport aux non-CSCs. Son activation, tout comme celle des voies de l’EMT (STAT3, MEK/p38/JNK et Akt/mTOR) est dépendante des radicaux libres oxygénés (RLO), dont la production est homogène dans la cellule en réponse aux photons. Par contre, les RLO produits dans la trace des ions carbone ne permettent pas d’activer HIF-1α et les voies de l’EMT. Sous hypoxie, une relation a été établie entre l’activation d’HIF-1α et celles des voies de détection (ATM) et de réparation (Rad51) des CDBs (Recombinaison Homologue). Ces travaux démontrent que l’avantage thérapeutique des ions carbone repose sur la répartition spatiale des RLO à l’échelle nanométrique et consécutivement sur la non-activation de voies clés de la défense cellulaire tumorale
Hadrontherapy using carbon ions is an alternative to photon irradiation in the treatment of Head and Neck cancers, because of accurate ballistics and high biological efficiency, including hypoxic tumor areas. These cancers are of poor prognosis because of a high risk of recurrences related to the presence of cancer stem cells (CSCs).The aim of this work was to determine the molecular specificities of the response to carbon ion irradiations compared to photons in two cancer cell lines and their CSCs’ subpopulation, in hypoxic and normoxic conditions. This work focused on the role of the HIF-1α protein in cell survival, since hypoxia promotes its stabilization, but also in the radioresistance; the epithelial-mesenchymal transition (EMT) and the detection and repair of DNA double-strand breaks (DSBs). HIF-1α is stabilized earlier in CSCs compared to non-CSCs. Its activation, as well as the EMT pathways (STAT3, MEK/p38/JNK and Akt/mTOR), are dependent on reactive oxygen species (ROS), whose production is homogeneous in response to photons. At the opposite, the ROS produced in the carbon ion tracks are insufficient to activate HIF-1α and the upstream EMT pathways. Under hypoxic conditions, a relationship has been established between HIF-1α activation and that of the DSBs detection (ATM) and repair (Rad51) pathways (Homologous Recombination). These studies demonstrate that the therapeutic advantage of carbon ions is based on the spatial ROS distribution at the nanoscale and consequently on the non-activation of key pathways involved in tumor cell defense
Styles APA, Harvard, Vancouver, ISO, etc.

Chapitres de livres sur le sujet "Leukaemia, Stem cells, Hypoxia, HIF"

1

Ude, Arinzechukwu, et Kelechi Okeke. « Leukaemia : The Purinergic System and Small Extracellular Vesicles ». Dans Purinergic System [Working Title]. IntechOpen, 2022. http://dx.doi.org/10.5772/intechopen.104326.

Texte intégral
Résumé :
Haematopoiesis is a tightly regulated process, by intrinsic and extrinsic factors, to produce lifelong blood cell lineages within the bone marrow. In the bone marrow microenvironment, mesenchymal stem cells and haematopoietic stem cells play important roles to ensure that haematopoiesis is maintained. These cells contain purines and pyrimidines that control intercellular process such as energy transport. However, in some cases, this process may be misregulated thus leading to the production of various diseases, including leukaemia. As a result, bone marrow cells may be stimulated via stress or induced hypoxia, and this leads to the release of purine and pyrimidine nucleotides and nucleosides into the extracellular space, and activation of autocrine/paracrine feedback loops. These extracellular nucleotides and nucleosides, and their respective cell surface receptors are involved in purinergic signaling that control different physiologic functions in cells including proliferation, differentiation, and cell death. These extracellular nucleotides and nucleosides include ATP, UTP, adenosine diphosphate (ADP), UDP and adenosine however the most important players are ATP and its metabolite adenosine. ATP is degraded via a sequential activity of ectonucleotidases. ATP, adenosine and these ectonucleotidases play very important roles in the tumour microenvironment crucial to disease development, progression, and aggressiveness by modulating immune response to leukaemia treatment and increasing homing of leukaemic cells.
Styles APA, Harvard, Vancouver, ISO, etc.
Nous offrons des réductions sur tous les plans premium pour les auteurs dont les œuvres sont incluses dans des sélections littéraires thématiques. Contactez-nous pour obtenir un code promo unique!

Vers la bibliographie