Thèses sur le sujet « CXCR4 axi »
Créez une référence correcte selon les styles APA, MLA, Chicago, Harvard et plusieurs autres
Consultez les 23 meilleures thèses pour votre recherche sur le sujet « CXCR4 axi ».
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.
Parcourez les thèses sur diverses disciplines et organisez correctement votre bibliographie.
BIONDI, MARTA. « Enhancing AML CAR CIK therapeutic potency increasing the localization of engineered cells in the malignant niche and its selectivity by LSCs specific targeting ». Doctoral thesis, Università degli Studi di Milano-Bicocca, 2022. http://hdl.handle.net/10281/365153.
Texte intégralChimeric Antigen Receptor (CAR) T-cell therapy has produced remarkable clinical responses in patients affected by acute lymphoblastic leukemia. Unfortunately, CAR T-cells have not been equally successful in acute myeloid leukemia (AML) due to tumor heterogeneity, lack of truly AML-restricted target antigens and the role of leukemia microenvironment in blasts protection and leukemia stem cells (LSCs) maintenance. Specifically, the bone marrow (BM) niche, where LSCs reside, is involved in leukemia promoting activities whilst suppressing normal hematopoiesis. Therefore, we hypothesized that targeting LSCs at their location may enhance the potency and selectivity of CAR-T cells. To address this issue, we have designed two aims: 1) promote rapid and efficient localization of CAR T-cells within the BM niche, 2) select a leukemia-restricted antigen to specifically target AML blasts and LSCs. First, we proposed to harness CD33.CAR-redirected Cytokine-Induced Killer (CIK) cells, an alternative effector T-cell population with acquired NK-like cytotoxic activity as well as minimal alloreactivity, to selectively route their activity to leukemia transformed niche. The chemokine ligand 12 (CXCL12), released by mesenchymal stromal cells (MSCs) within the medullary niche, and its chemokine receptor 4 (CXCR4) are two pivotal players regulating leukocytes trafficking to the BM. In AML, CXCL12 interacts with CXCR4 overexpressed on blasts, promoting their migration and homing in the niche. Hence, taking advantage of this axis might facilitate CD33.CAR-CIK cells homing to the BM and therefore leukemia eradication. However, ex vivo manipulation protocols of CD33.CAR-CIK cells consistently downregulate CXCR4 expression and may affect the capacity of adoptively infused cells to migrate to BM and exert their anti-leukemic action. Therefore, to improve CD33.CAR-CIKs homing in the BM microenvironment we have developed CD33.CAR-CIK cells overexpressing CXCR4, in its wild-type or hyperactive mutant form. Notably, CIK cells engineering with CD33.CAR-CXCR4 constructs led to a consistent increase in CXCR4 expression, without altering CIK cells phenotype and CAR-related effector functions. Interestingly, compared to conventional CD33.CAR-CIK cells, CD33.CAR-CXCR4WT and especially CD33.CAR-CXCR4MUT-CIK cells demonstrated significantly superior in vitro chemotactic response toward CXCL12 and MSC-derived supernatants, and greater in vivo BM homing ability and persistence. Furthermore, to develop an effective anti-AML CAR T-cell therapy, it is fundamental to identify a LSC-specific marker, sparing the normal counterpart of hematopoietic stem cells (HSCs). T-cell immunoglobulin and mucin protein 3 (TIM-3) is an immune checkpoint molecule, it plays a central role in immune responses in AML and it is an LSC-specific marker, lacking expression on HSCs. Therefore, we designed a third-generation anti-TIM-3.CAR using the single-chain fragment variable (scFv) derived from an antagonistic ligand-blocking anti-TIM-3 antibody. In vitro, TIM-3.CAR-CIK cells efficiently killed both AML cell lines and primary AML blasts, but not normal TIM-3+ activated CIK cells, monocytes and NK-cells. Notably, we observed selective elimination of primary LSC-enriched population (CD34+ CD38-). Furthermore, TIM-3.CAR-CIK cells maintained their effector functions despite multiple in vitro restimulations, setting the basis for further exploration in in vivo models. Overall, both approaches, one improving CAR-CIK cells homing to the transformed niche and the other conferring superior safety and selectivity, might improve the efficacy of anti-AML CAR-CIK therapy.
SOLARI, AGNESE. « CXCR4/CXCR7-CXCL11/CXCL12 AXIS PROMOTES MALIGNANT PHENOTYPE IN PUTATIVE STEM CELLS FROM HUMAN MENINGIOMAS ». Doctoral thesis, Università degli studi di Genova, 2019. http://hdl.handle.net/11567/946169.
Texte intégralMikami, Sakae. « Blockade of CXCL12/CXCR4 axis ameliorates murine experimental colitis ». Kyoto University, 2009. http://hdl.handle.net/2433/124258.
Texte intégralHoward, Cory M. « Characterization of the CXCR4-LASP1-eIF4F Axis in Triple-Negative Breast Cancer ». University of Toledo Health Science Campus / OhioLINK, 2020. http://rave.ohiolink.edu/etdc/view?acc_num=mco1596298549051863.
Texte intégralDas, Avik. « Ischemic stroke in type II diabetic mice : Deregulation of SDF-1a/CXCR4 axis ». Wright State University / OhioLINK, 2009. http://rave.ohiolink.edu/etdc/view?acc_num=wright1247594137.
Texte intégralRondeau, Vincent. « Rôle de la désensibilisation de CXCR4 dans la spécification lympho-myéloïde des progéniteurs hématopoïétiques multipotents. Lymphoid differentiation of hematopoietic stem cells requires efficient Cxcr4 desensitization New method to obtain lymphoid progenitors CXCR4-driven mitochondrial metabolic pathways shape the lympho-myeloid fate of hematopoietic multipotent progenitors ». Thesis, université Paris-Saclay, 2020. http://www.theses.fr/2020UPASQ022.
Texte intégralHematopoietic stem and progenitor cells (HSPCs), including the multipotent progenitors (MPPs), are responsible for replenishing immune cells. They reside in bone marrow (BM) endosteal and (peri)-vascular niches, which provide all cellular and molecular components required for their lifelong maintenance and fate. Among them, the CXCL12 chemokine and one of its receptor, CXCR4, exert a dominant role in promoting HSPC retention and quiescence. These processes are deregulated in the WHIM Syndrome (WS), a rare immunodeficiency caused by inherited heterozygous autosomal gain-of-function CXCR4 mutations that affect homologous desensitization of the receptor. Clinically, WS is notably characterized by severe, chronic circulating lymphopenia whose mechanisms remain to be elucidated. Using a mouse model carrying a naturally occurring WS-linked Cxcr4 mutation as well as human BM and blood samples, we explored the possibility that the lymphopenia in WS originates from defects at the HSPC level in BM. We reported that Cxcr4 desensitization is required for lymphoid differentiation of HSPCs and further identified the MPP stage as defective in mutant mice. The divergence between lymphoid and myeloid lineages occurs at the MPP stage, which is composed of distinct subpopulations, i.e., MPP2 and MPP3 are reported as distinct myeloid-biased MPP subsets that operate together with lymphoid-primed MPP4 to control blood leukocyte production. Our understanding of how cell-extrinsic niche-related and cell-intrinsic cues drive the lymphoid versus myeloid fate decision of MPPs is still fragmentary. Therefore, my PhD project aimed at determining whether and how CXCR4 signaling regulates bioenergetics demands of MPPs and at understanding how these metabolic pathways shape the lympho-myeloid fate of MPPs. We unraveled a myeloid skewing of the HSPC compartment in BM of WS mice and patients. In mutant mice, this partly relied on the contraction of the MPP4 pool and on cell-autonomous molecular and metabolic changes that reprogramed MPP4 away from lymphoid differentiation. Interestingly, chronic treatment with the CXCR4 antagonist AMD3100 normalized mitochondrial metabolism and fate of MPP4, while correcting circulating lymphopenia in WS mice. This study provides evidence that CXCR4 signaling acts as an essential gatekeeper for integrity of the mitochondrial machinery, which in turn controls lymphoid potential of MPP4
Goh, Poh. « Roles of protein kinase C and arrestin in migration of cells via CXCR4/CXCL12 signalling axis ». Thesis, University of East Anglia, 2018. https://ueaeprints.uea.ac.uk/67806/.
Texte intégralKato, Itaru. « Identification of hepatic niche harboring human acute lymphoblastic leukemic cells via the SDF-1/CXCR4 axis ». Kyoto University, 2012. http://hdl.handle.net/2433/157438.
Texte intégralSwidenbank, Isabella. « The role of the CXCR4-CXCL12 chemokine axis in melanoma metastasis to the normal and fibrotic liver ». Thesis, University of Newcastle upon Tyne, 2014. http://hdl.handle.net/10443/2612.
Texte intégralChow, Yan Ching Ken. « Role and Molecular Basis of the CXCL12-signalling Axis in the Pathogenesis of WHIM syndrome and the carcinogenesis associated with human papillomavirus (HPV) infection ». Paris 7, 2008. http://www.theses.fr/2008PA077129.
Texte intégralThe WHIM syndrome (WS) is a rare immunodeficiency characterised by severe leukoneutropenia (e. G. Myelokathexis) and profuse human papillomavirus (HPV)-associated skin lesions and malignant ano-genital cohdyloma. The disease links to dysfunctions of the CXCR4 chemokine receptor in response to its ligand SDF-1/CXCL12, and associates in many cases to heterozygous mutations causing truncation in the cytoplasmic tail of the receptor that is important for the β-arrestin (βarr)-mediated receptor desensitisation process. Such truncated receptor (e. G. CXCR4¹º¹³) displays no desensitisation and thus manifests a gain of function in response to CXCL12 in leukocytes derived from WS patients, which likely contribute to the pathogenesis of the disorder. In this study, we demonstrated that such dysfunctions are in fact dependent on an unexpected interaction between βarr2 and CXCR4¹º¹³. Upon CXCL12 stimulation, the CXCR4¹º¹³receptor displays an augmented and prolonged |3arr2-dépendent signalling that relies on the integrity of the third intracellular loop of the receptor. We have also observed the existence of CXCR4wt/CXCR4¹º¹³ heterodimer from which the possible enhanced parr2/CXCR4¹º¹³ interaction may contribute to the augmented response of the receptor to CXCL12. With the abnormal expression of CXCL12 we observed in HPV-induced lesions derived from both WS and non-WS patients, and the critical role of the chemokine in tumor growth and metastasis, we speculate on the existence of an HPV/CXCL12 interplay that could be crucial for the viral-mediated pathogenesis. Using keratinocytes immortalised by the subgenomic fragment of high-risk HPV, we showed an HPV-E6/7-dependent expression of CXCL12 and its receptors and the critical role of this signalling axis in the prolifération and motility of these cells. In WS, such HPV/CXCL12-interplay may synergise with the hyperfunctioning of CXCR4, and contribute to the malignant development of ano-genital condyloma that is unusually associated with low-risk HPV - the only viral subtype we identified in these lesions
Matsusue, Ryo. « Hepatic Stellate cells promote Liver Metastasis of Colon Cancer Cells by the Action of SDF-1/CXCR4 Axis ». Kyoto University, 2011. http://hdl.handle.net/2433/142051.
Texte intégralHassan, Saima. « The diagnostic and therapeutic role of the stromal cell-derived factor (SDF)-1/CXCR4 axis in breast cancer metastasis ». Thesis, McGill University, 2009. http://digitool.Library.McGill.CA:80/R/?func=dbin-jump-full&object_id=66712.
Texte intégralLe cancer du sein tue par le processus des métastases. Dans le but d'améliorer le pronostic des patients atteints du cancer du sein, une meilleure compréhension des facteurs sous-jacents qui conduisent à la transformation métastatique est nécessaire. Une théorie qui explique la transformation métastatique propose que les chimiokines, telles que le stromal cell-derived factor (SDF)-1, sont surexprimées dans des organes métastatiques distants spécifiques, tels que le poumon, le foie, et les os et servent à attirer les cellules cancéreuses qui expriment leurs récepteurs, tels CXCR4. L'hypothèse de cette t hèse est donc que l'axe SDF-1/CXCR4 joue un rôle important dans la transformation métastatique dans le cancer du sein, et que cet axe ligand/récepteur peut être exploité dans le diagnostic et la thérapie du cancer du sein. Le premier objectif de cette thèse était de déterminer si les niveaux circulants de SDF-1 peuvent prédire la présence de métastases du cancer du sein. Nous avons découvert que des niveaux peu élevés de SDF-1 dans le plasma représentent un bon déterminant pronostique indépendant, suggérant que le gradient de concentration de faible niveaux de SDF-1 dans le plasma et de niveaux élevés de SDF-1 dans l'organe métastasé peut être un événement critique dans le transfert des cellules cancéreuses de la circulation sanguine jusqu'à l'organe-cible. Nous avons de plus déterminé que les niveaux plasmatiques de SDF-1 sont indépendants des tumeurs, identifiant le premier marqueur sanguin, dérivé de l'hôte, de prédiction de métastases éloignées. Le second objectif était de déterminer si l'expression tumorale de CXCR4 pourrait moduler l'effet pronostique des niveaux plasmatiques de SDF-1. Nous avons découvert que les patients dont les tumeurs expriment de façon élevée la forme activée du récepteur, CXCR4 phosphorylé, et des niveaux plasmatiq
Mihic, Bojana [Verfasser], Irmgard [Akademischer Betreuer] Merfort et Jochen [Akademischer Betreuer] Seufert. « Macrophage migration inhibitory factor (MIF)/CXCR4 axis in migration of mesenchymal stem cells (MSC) towards pancreatic [beta]-cells under lipotoxic conditions ». Freiburg : Universität, 2017. http://d-nb.info/1179694651/34.
Texte intégralOgawa, Ryotaro. « Loss of SMAD4 Promotes Colorectal Cancer Progression by Recruiting Tumor-Associated Neutrophils via the CXCL1/8-CXCR2 Axis ». Kyoto University, 2019. http://hdl.handle.net/2433/245315.
Texte intégralZimmerman, Grant Robert. « T-ALL LEUKEMIA DYSREGULATES STROMAL BONE MARROW ENVIRONMENT AND DISRUPTS NICHE-STEM CELL SIGNALING AXIS ». Case Western Reserve University School of Graduate Studies / OhioLINK, 2015. http://rave.ohiolink.edu/etdc/view?acc_num=case1436293859.
Texte intégralLi, Yin [Verfasser], Andreas [Akademischer Betreuer] Knopf, Murat [Gutachter] Bas et Andreas [Gutachter] Knopf. « CXCR4-CXCL12 axis in head and neck squamous cell carcinoma regarding HPV status / Yin Li ; Gutachter : Murat Bas, Andreas Knopf ; Betreuer : Andreas Knopf ». München : Universitätsbibliothek der TU München, 2021. http://d-nb.info/1236343069/34.
Texte intégralMachado, Isabel Daufenback. « Mecanismos moleculares da ação dos glicocorticóides endógenos e da anexina-A1 sobre o tráfego de neutrófilos : caracterização da ação sobre os eixos SDF-1α/CXCR4 e IL-17/IL-23/G-CSF ». Universidade de São Paulo, 2013. http://www.teses.usp.br/teses/disponiveis/9/9136/tde-27022014-105135/.
Texte intégralThe traffic leukocytes is a complex process dependent on the action of severals chemical mediators, in addition to perfect cell interaction. Therefore, this study aimed to evaluate the effect of GCe and ANXA1 on SDF-1α/CXCR4 and IL-17/IL-23/G-CSF and on the expression of adhesion molecules CD18, CD49d and CD62L. Balb/C wild type and ANXA1-/- male mice were employed. The analysis were performed at physiological conditions, in the presence of high concentrations of GCe and during of inflammatory process induced by ACTH administration (5 µg/animal, i.p.) or LPS injection (100 µg/kg, i.p.), respectively or in the absence of GCe action, by the action of RU 38486 (RU, 10 mg/kg , i.p.). The involvement of the receptor FPR2 and ANXA1 was assessed by pre-treatment with Ac2-26 (1 mg/kg, i.p.) or BOC2 (10 µg/animal, i.p.) for 4 days, once a day. The quantification of total and differential cell was performed in a Neubauer chamber and stained smears by May-Grunwald and flow cytometry. Quantification of expression of CXCR2, CXCR4, FPR2, CD18, CD49d, CD62L and granulocytic maturation (CD11b/Ly6G) in the bone marrow and circulation were performed by flow cytometry. The expression of ANXA1 on tissues was performed by western blotting and on cells from bone marrow and blood by immunocytochemistry. Quantification of IL-17, IL-23, G-CSF, SDF-1α and corticosterone were performed by ELISA. The chemotaxis of neutrophils from the bone marrow and blood was tested in the chemotaxis chamber with filter pore of 8 microns. The phagocytosis of apoptotic neutrophils by bone marrow macrophages was assessed by in vitro assay. To investigate the effects of ACTH in the migration of neutrophils in the inflammatory process, the model employed was air pouch (100 µg/ ml, LPS), and the behavior of circulating leukocytes from animals treated with ACTH were evaluated by intravital microscopy. The results obtained, which are presented in three sections, showed that: 1) neutrophils from the bone marrow and blood expressed ANXA1 in the cytoplasm and membrane, as well as FPR2, constitutively and the expression of both is regulated by GCe. The ANXA1 via FPR2 receptor expressed in bone marrow cells, controls the neutrophilic maturation and traffic of these cells from the bone marrow into the blood. The ANXA1 via interaction to FPR2 controls the clearance of neutrophils from the blood to the bone marrow by modulating the SDF-1α/CXCR4 axis; 2) the administration of ACTH induces neutrophilia and the circulating neutrophils are ANXA1+, CD18+, CD49d+ and CD62L+, showing that the injection of ACTH in vivo alters the phenotype of these cells in the blood. These modifications alter the behavior of neutrophils in the blood, as well as the migration to the air pouch in the presence of inflammation and to the tissue clearance, and these effects may be dependent, at least in part, on inhibition of migration oriented events, as chemotaxis in response to fMLP or SDF-1α were reduced. Further, the clearance of neutrophils is reduced in animals treated with ACTH due to the lower phagocytic and secretory activity of medullary macrophages; 3) Animals treated with RU 38486 and ANXA1-/- mobilize granulocytes from bone marrow into the blood, and from this compartment to the focus of inflammation with higher intensity than that observed in the control group. The axis IL-17/IL-23/G-CSF seems to be involved in granulopoiesis and mobilization of neutrophils into the blood during inflammation, but it is not the target of action of ANXA1 and GCe at this step of inflammatory process. Additionally, it was observed that in the presence of peritonitis, the adhesion molecules, CD49d and CD62L are involved in the migration of neutrophils from the bone marrow into the blood. The results obtained allow concluding that the GCe and ANXA1 are relevant to the granulopoiesis and the traffic of neutrophils from bone marrow under physiological conditions and in the presence of inflammation. Furthermore, together with literature data, the data presented here may suggest the involvement of ANXA1 the GCe in phenotypic plasticity of neutrophils according to the stimuli that are submitted, and may support to understand the new concepts of production, half-life, location and function of neutrophils.
Patil, Ashwini [Verfasser], et Joachim [Akademischer Betreuer] Göthert. « The CXCL10/CXCR3 axis cross-talk between emerging T cell acute lymphoblastic leukemia and thymic epithelial cells / Ashwini Patil ; Betreuer : Joachim Göthert ». Duisburg, 2021. http://d-nb.info/1225294649/34.
Texte intégralDiez, Tejerina Santiago. « La expresión del eje CXCL 12/CXCR4 como predictor de la respuesta a nivel regional en pacientes con carcinomas escamosos de cabeza y cuello ». Doctoral thesis, Universitat Autònoma de Barcelona, 2016. http://hdl.handle.net/10803/378021.
Texte intégralPurpose: The CXCL12 / CXCR4 axis is a chemokine system (ligand) and its receptor, which function normally and are necessary for the homeostasis of various body systems. It is suspected that some head and neck carcinomas using this axis to spread to regional lymph from the primary tumor. Few studies have substantiated this pathway as a possible marker to predict the occurrence of lymph node recurrences. We studied the prognostic axis CXCL12 / CXCR4 on possible nodal recurrence in patients treated for HNSCC. Experimental design and results: Healthy and tumor samples from 111 patients with HNSCC (oral cavity, oropharynx, larynx and hypopharynx) and treated with radical intent with minimum follow-up of 3 years were analyzed. The appearance of lymph node recurrence, regional recurrence free survival, local or remote and survival adjusted depending on the expression of CXCL12/CXCR4 axis expression and other inflammatory pathways associated with response to treatment and was rated capacity known spread (COX, NFkB, IL-1, CD-45, MPO, MPC-1, MMP-2 and 9, Hsp-90 and SOD). We found that the expression of CXCL12 was lower and CXCR4 higher in the tumor than in healthy mucosa. Patients were distributed through an analysis method of recursive partitioning, in groups according to the expression of CXCL12 and CXCR4. Patients with low expression of CXCR4 and high CXCR4 and CXCL12 high, had a low risk of tumor recurrence at nodal level, 2.6% and 4.5% respectively. The patient group consisting of high expression of CXCR4 CXCL12 and counted down with a high risk of local relapse, 34.0%. Multivariate analysis calculated a 10.7 times greater risk of regional recurrence in these patients compared to the previous group. All this regardless of the category of initial tumor extension at regional level, the type of treatment performed on nodal areas or local control of the disease. Additionally, a significant correlation between the transcriptional expression of CXCL12 and CXCR4 with PGIS (PGI2), SOD-2 and COX-2, PGIS, VEGF and MMP-2 respectively is shown. Conclusions: CXCL12 expression was significantly lower and upper CXCR4 in tumor samples corresponding to samples of healthy mucosa. Expression levels of CXCL12/CXCR4 not differ depending on the location of the tumor nor HPV status. The low expression of CXCL12 and CXCR4 elevated in patients with HNSCC shows a clear increase in the risk of suffering a relapse during evolution nodal treatment of these patients, decreasing significantly with survival compared to patients with high expression of CXCL12 and high CXCR4 or CXCR4 expression low. There is a correlation between CXCL12/CXCR4 and the expression of various genes related to response to treatment and the ability to spread the HNSCC as PGIS, COX-2, VEGF and MMP-2.
Ahmed, Tahir. « Selective neuronal loss, microglial activation and adult neurogenesis after stroke : an in vitro and in vivo analysis of the SDF-1alpha/CXCR4 axis in focal ischaemia and endogenous mechanisms of neural repair ». Thesis, University of Cambridge, 2012. http://ethos.bl.uk/OrderDetails.do?uin=uk.bl.ethos.610788.
Texte intégralChiang, Tsaiyu, et 江彩語. « Lipid-Based Nanoparticles Inhibiting SDF1-α/ CXCR4 axis and Delivering Anti-angiogenic siRNA for Ttreating Liver Fibrosis ». Thesis, 2015. http://ndltd.ncl.edu.tw/handle/rs3vu4.
Texte intégralMartins, Ana Catarina Correia. « Does hyperglycemia impact the crosstalk between AML cells and BM stroma ? : the role of CXCL12-CXCR4 chemoattraction axis ». Master's thesis, 2015. http://hdl.handle.net/1822/47403.
Texte intégralThe crosstalk between acute myeloid leukemia (AML) cells and their neighboring stromal cells is often key for cancer cells survival, resistance to treatment and disease relapse. Importantly, altered metabolic conditions of the bone marrow (BM) niche can influence stem cell behavior. Type 2 diabetes mellitus (T2DM) induces persistent changes in the BM microenvironment, disturbing stem cell niches and consequently hematopoiesis and BM cells function. T2DM and AML share the average age of onset, both disorders more prevalent in the elderly. Moreover, T2DM is often associated with poorer AML outcomes. Yet the association between these two disorders has not been clarified or dissected. The CXCL12/CXCR4 chemoattraction axis is a possible mechanism by which diabetes affects AML progression. The CXCL12/CXCR4 axis altered function is known to impair hematopoietic stem cells mobilization in a diabetic context, and to enhance survival and chemoresistance of AML cells. Thus, the aim of this project was to dissect the role of the CXCL12/CXCR4 axis on the AML poorer outcomes associated with diabetes‐induced damage. Specifically, we addressed the activity of the CXCL12/CXCR4 chemoattraction axis on the crosstalk between BM stromal cells and AML cells in a diabetes context. Using primary human samples, a deregulation of the CXCL12/CXCR4 patterns within the BM diabetic microenvironment was observed. Therefore, we next evaluated the effect of such alteration on the crosstalk between the AML and stromal cells. Submitting two AML cell lines (HL‐60 and KG‐1) to either BMSCs‐conditioned media or a BMSCs‐AML direct contact co‐culture system, upon normal and diabetic conditions, a strong association between CXCR4 activation and AML cells tumorigenicity was demonstrated in a diabetic context. In fact, BM stromal cells from diabetic patients were shown to activate CXCR4 receptor in HL‐60 cells, which led to activation of the Akt/MEK survival pathways, and increase of the cancer cells’ tumorigenicity. Briefly, our results support the hypothesis that the CXCL12/CXCR4 axis may play a critical role on the prognostic of AML patients that also suffer from metabolic disorders, as diabetes. This project unveils the very “tip of the iceberg” on the diabetes facilitating‐role on AML progression and tumorigenicity, within BM microenvironment.
A comunicação entre as células de leucemia mielóide aguda (LMA) e as células estromais vizinhas é, muitas vezes, crucial para a sobrevivência das células cancerígenas, resistência ao tratamento e recaída. Mais importante, condições metabólicas alteradas do nicho da medula óssea (MO) podem influenciar o comportamento das células estaminais. Diabetes mellitus tipo 2 (T2DM) induz alterações persistentes no microambiente da MO, perturbando os nichos das células estaminais e, consequentemente, a hematopoiese e as funções das células da MO. T2DM e LMA partilham a idade média de diagnóstico, sendo ambas doenças prevalentes na população idosa. Além disso, T2DM é frequentemente associada com maus prognósticos da LMA. No entanto, a associação entre as duas doenças não foi ainda estudada. O eixo de quimioatração CXCL12/CXCR4 é um mecanismo possível pelo qual diabetes afeta a progressão da LMA. A função alterada do eixo CXCL12/CXCR4 prejudica a mobilização das células estaminais hematopoiéticas num contexto de diabetes, e aumenta a sobrevivência e quimioresistência das células de LMA. Assim sendo, o objetivo deste projeto era avaliar os possíveis efeitos que o dano induzido pela diabetes na MO teria na progressão da LMA, tendo em conta o eixo de quimioatração CXCL12/CXCR4. Usando amostras humanas primárias, foi observada uma desregulação dos padrões de CXCL12/CXCR4 no microambiente diabético da MO. Portanto, de seguida avaliamos o efeito de tal alteração na comunicação entre células de LMA e células estromais. Submetendo duas linhas de AML (HL‐60 e KG‐1) a meio condicionado de células estromais derivadas da MO, ou a um sistema de co‐cultura direta, em condições normal e diabética, uma forte associação entre a ativação do CXCR4 e a tumorigenicidade das células de LMA foi demonstrada no contexto diabético. Na verdade, as células estromais da MO de pacientes diabéticos são capazes de ativar o receptor CXCR4 nas células HL‐60, levando à ativação das cascatas de sobrevivência Akt/MEK, e consequente aumento da tumorigenicidade das células de AML. Em resumo, os resultados deste projeto sugerem a hipótese de que o eixo de quimioatração CXCL12/CXCR4 tem um papel importante no prognóstico de pacientes de LMA que também sofrem de doenças metabólicas, como a diabetes. Este projeto desvenda a “ponta do iceberg” do papel facilitador da diabetes na progressão e tumorigenicidade da LMA, no microambiente da MO.
Chou, Andy Shau-Bin, et 周紹賓. « Cancer progression associated with dysregulation of selected cytokine pathways and the CXCR4/CXCL12 axis in nasopharyngeal carcinoma and CEA-producing lymph node metastatic carcinoma cell lines : role of cancer stem cells ». Thesis, 2011. http://ndltd.ncl.edu.tw/handle/79429262520807143447.
Texte intégral長庚大學
臨床醫學研究所
99
We have divided this PhD thesis into three parts. While each is independent, these three parts of study are closely related in the subjects of cancer metastasis, tumor microenvironment and cancer stem cells. In the first part, since the understanding of immunobiology of bone marrow metastases (designated BM-NPC) versus primary tumors (P-NPC) of the nasopharynx is far from complete, the goal of this part of study was to determine if there would be differences between cultured P-NPCs and BM-NPCs with respect to (i) the expression levels of constitutive IL-6 and the gp80 subunit of IL-6 receptor (IL-6Rα) in the spent media of nontransduced P-NPC and BM-NPC cells, and (ii) the expression levels of IL-6 and IL-6Rα in the spent media of P-NPC and BM-NPC cells transduced with a retroviral vector containing the IFN–γ gene. Our results showed that in day 3 culture supernatants, low levels of soluble IL-6 were detected in 5/5 cultured tumor cell lines derived from P-NPCs, while much higher constitutive levels of IL-6 were detected in 3/3 metastasis-derived NPC cell lines including one originated from ascites; the difference was significant (p = 0.025). An inverse relationship was found between IL-6Rα and IL-6 in their release levels in cultured P-NPCs and metastasis-derived NPCs. In IFN-γ-transduced-P-NPCs, IL-6 production increased and yet IL-6Rα decreased substantially, as compared to nontransduced counterparts. At variance with P-NPC cells, the respective ongoing IL-6 and IL-6Rα release patterns of BM-NPC cells were not impeded as much following IFN-γ transduction. These observations were confirmed by extended kinetic studies with representative NPC cell lines and clonal sublines. The latter observation with the clonal sublines also indicates that selection for high IL-6 or low IL-6Rα producing subpopulations did not occur as a result of IFN-γ-transduction process. P-NPCs, which secreted constitutively only marginal levels of IFN-γ (8.4 ~ 10.5 pg/ml), could be enhanced to produce higher levels of IFN-γ (6.8- to 10.3-fold increase) after IFN-γ gene transduction. Unlike P-NPCs, BM-NPCs spontaneously released IFN-γ at moderate levels (83.8 ~ 100.7 pg/ml), which were enhanced by 1.3 to 2.2-fold in the spent media of their IFN-γ-transduced counterparts. In conclusion, our results clearly showed that cultured P-NPCs and BM-NPCs could be distinguished from one another on the basis of their differential baseline secretion pattern of IFN-γ, IL-6 and IL-6Rα, and their differential response profiles to IFN-γ gene transfer of the production of these three soluble molecules. These results suggest that the IL-6 and IFN-γ pathways in a background of genetic instability be involved in the acquisition of metastatic behavior in BM-NPCs. In the second part of the study, we looked into the mechanisms underlying tumor dormancy in human P-NPCs and BM-NPCs. The specific aim of this part of study was to determine differences in the fates of P-NPC cells, IFN-γ-transduced primary NPC (IFN-γ-P-NPC), bone marrow metastatic NPC (BM-NPC) and IFN-γ-transduced BM-NPC (IFN-γ-BM-NPC) cells following xenotransplantation into 4 different groups of SCID mice through subcutaneous injection of 5 x 106 cells/site/animal (4 animals/group). The injected mice were monitored for tumor development at the sites of injection. In only the group injected with IFN-γ-P-NPC cells, the resulting nodules remained small throughout the 60-days observation period after injection, but gradually became palpably prickly. Histopathological examination revealed that these lesions invariably consisted of mostly structures of horny pearls and keratin bridges with occasional apoptotic and degenerative cells. In contrast, animals injected with non-transduced-P-NPC cells developed tumors progressively with occasional central necroses. In the two groups injected with IFN-γ-NPC-BM and NPC-BM cells, progressive growths of tumors were noted with the latter being at slightly faster rates; while the xenografts of both groups showed a poorly differentiated phenotype with abundant vascularity. Our results highlight the high susceptibility of P-NPC, but not BM-NPC, cells, following IFN-γ gene transfer to the induction of tumor dormancy which is mediated via induced cell differentiation. Thus induced cell differentiation by agents such as IFN-γ could provide a new mechanism by which tumor dormancy is initiated and established. In the last part of the study, we looked into how the metastatic cancer stem cells (mCSCs) were converted from cancer stem cells (CSCs). As an advanced status of CSCs, mCSC shave been proposed to be the essential seeds that initiate tumor metastasis. In this study, we used a lymph node metastatic CEA-producing carcinoma cell line, UP-LN1, characterized by the persistent appearance of adherent (A) and floating (F) cells in culture, to determine the distribution of CSCs and mechanisms for the induction of mCSCs. F and A cells displayed distinct phenotypes, CD44high/CD24low and CD44low/CD24high, respectively. The CSC-rich nature of F cells was typified by stronger expression of multiple drug resistance genes and a 7.8-fold higher frequency of tumor-initiating cells in NOD/SCID mice when compared to A cells. F cells exhibited a greater depression in HLA class I expression and an extreme resistance to NK/LAK-mediated cytolysis. Moreover, the NK/LAK-resistant F cells were highly susceptible to IFN-γ-mediated induction of surface CXCR4 with concomitant down-regulation of cytoplasmic CXCL12 expression, whereas these two parameters remained essentially unchanged in NK/LAK-sensitive A cells. Following the induction of surface CXCR4, enhanced migratory/invasive potential of F cells was demonstrated by in vitro assays. Confocal immunofluorescence microscopy showed the two distinct phenotypes of F and A cells could be correspondingly identified in monodispersed and compact tumor cell areas within the patient’s LN tumor lesion. In response to IFN-γ or activated NK/LAK cells, the CXCR4+ mCSCs could be only induced from the CSCs which were harbored in the highly tumorigenic CD44high/CD24low F subset. Collectively, these results revealed the complexity and heterogeneity of the CSC of this cell line/tumor, localization of the major niche of CSCs/mCSCs in F subsets, and the differential immunomodulatory functions of F and A subsets.