Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 31
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Oncoimmunology ; 12(1): 2244330, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37577144

RESUMEN

Malignant tumors often escape anticancer immune surveillance by suppressing the cytotoxic functions of T lymphocytes. While many of these immune evasion networks include checkpoint proteins, small molecular weight compounds, such as the amino acid L-kynurenine (LKU), could also substantially contribute to the suppression of anti-cancer immunity. However, the biochemical mechanisms underlying the suppressive effects of LKU on T-cells remain unclear. Here, we report for the first time that LKU suppresses T cell function as an aryl hydrocarbon receptor (AhR) ligand. The presence of LKU in T cells is associated with AhR activation, which results in competition between AhR and hypoxia-inducible factor 1 alpha (HIF-1α) for the AhR nuclear translocator, ARNT, leading to T cell exhaustion. The expression of indoleamine 2,3-dioxygenase 1 (IDO1, the enzyme that leads to LKU generation) is induced by the TGF-ß-Smad-3 pathway. We also show that IDO-negative cancers utilize an alternative route for LKU production via the endogenous inflammatory mediator, the high mobility group box 1 (HMGB-1)-interferon-gamma (IFN-γ) axis. In addition, other IDO-negative tumors (like T-cell lymphomas) trigger IDO1 activation in eosinophils present in the tumor microenvironment (TME). These mechanisms suppress cytotoxic T cell function, and thus support the tumor immune evasion machinery.


Asunto(s)
Quinurenina , Neoplasias , Humanos , Quinurenina/metabolismo , Quinurenina/farmacología , Evasión Inmune , Transducción de Señal , Linfocitos T , Microambiente Tumoral
2.
J Immunother Cancer ; 11(1)2023 01.
Artículo en Inglés | MEDLINE | ID: mdl-36599470

RESUMEN

BACKGROUND: Galectin-9 is a member of the family of lectin proteins and crucially regulates human immune responses, particularly because of its ability to suppress the anticancer activities of T lymphocytes and natural killer cells. Recent evidence demonstrated that galectin-9 is highly expressed in a wide range of human malignancies including the most aggressive tumors, such as high-grade glioblastomas and pancreatic ductal adenocarcinomas, as well as common malignancies such as breast, lung and colorectal cancers. However, solid tumor cells at rest are known to secrete either very low amounts of galectin-9 or, in most of the cases, do not secrete it at all. Our aims were to elucidate whether T cells can induce galectin-9 secretion in human cancer cells derived from solid malignant tumors and whether this soluble form displays higher systemic immunosuppressive activity compared with the cell surface-based protein. METHODS: A wide range of human cancer cell lines derived from solid tumours, keratinocytes and primary embryonic cells were employed, together with helper and cytotoxic T cell lines and human as well as mouse primary T cells. Western blot analysis, ELISA, quantitative reverse transcriptase-PCR, on-cell Western and other measurement techniques were used to conduct the study. Results were validated using in vivo mouse model. RESULTS: We discovered that T lymphocytes induce galectin-9 secretion in various types of human cancer cells derived from solid malignant tumors. This was demonstrated to occur via two differential mechanisms: first by translocation of galectin-9 onto the cell surface followed by its proteolytic shedding and second due to autophagy followed by lysosomal secretion. For both mechanisms a protein carrier/trafficker was required, since galectin-9 lacks a secretion sequence. Secreted galectin-9 pre-opsonised T cells and, following interaction with other immune checkpoint proteins, their activity was completely attenuated. As an example, we studied the cooperation of galectin-9 and V-domain Ig-containing suppressor of T cell activation (VISTA) proteins in human cancer cells. CONCLUSION: Our results underline a crucial role of galectin-9 in anticancer immune evasion. As such, galectin-9 and regulatory pathways controlling its production should be considered as key targets for immunotherapy in a large number of cancers.


Asunto(s)
Proteínas de Punto de Control Inmunitario , Neoplasias Pancreáticas , Humanos , Animales , Ratones , Galectinas/metabolismo , Linfocitos T Citotóxicos/metabolismo , Terapia de Inmunosupresión
3.
Front Med (Lausanne) ; 9: 790995, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35223897

RESUMEN

Immune checkpoint proteins play crucial roles in human embryonic development but are also used by cancer cells to escape immune surveillance. These proteins and biochemical pathways associated with them form a complex machinery capable of blocking the ability of cytotoxic immune lymphoid cells to attack cancer cells and, ultimately, to fully suppress anti-tumor immunity. One of the more recently discovered immune checkpoint proteins is V-domain Ig-containing suppressor of T cell activation (VISTA), which plays a crucial role in anti-cancer immune evasion pathways. The biochemical mechanisms underlying regulation of VISTA expression remain unknown. Here, we report for the first time that VISTA expression is controlled by the transforming growth factor beta type 1 (TGF-ß)-Smad3 signaling pathway. However, in T lymphocytes, we found that VISTA expression was differentially regulated by TGF-ß depending on their immune profile. Taken together, our results demonstrate the differential biochemical control of VISTA expression in human T cells and various types of rapidly proliferating cells, including cancer cells, fetal cells and keratinocytes.

4.
Int Immunopharmacol ; 100: 108155, 2021 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-34543981

RESUMEN

Galectin-9 is a member of the galectin family of proteins, which were first identified to specifically bind to carbohydrates containing ß-galactosides. Galectin-9 is conserved through evolution and recent evidence demonstrated its involvement in innate immune reactions to bacterial infections as well as the suppression of cytotoxic immune responses of T and natural killer cells. However, the molecular mechanisms underlying such differential immunological functions of galectin-9 remain largely unknown. In this work we confirmed that soluble galectin-9 derived from macrophages binds to Gram-negative bacteria by interacting with lipopolysaccharide (LPS), which forms their cell wall. This opsonisation effect most likely interferes with the mobility of bacteria leading to their phagocytosis by innate immune cells. Galectin-9-dependent opsonisation also promotes the innate immune reactions of macrophages to these bacteria and significantly enhances the production of pro-inflammatory cytokines - interleukin (IL) 6, IL-1ß and tumour necrosis factor alpha (TNF-α). In contrast, galectin-9 did not bind peptidoglycan (PGN), which forms the cell wall of Gram-positive bacteria. Moreover, galectin-9 associated with cellular surfaces (studied in primary human embryonic cells) was not involved in the interaction with bacteria or bacterial colonisation. However, galectin-9 expressed on the surface of primary human embryonic cells, as well as soluble forms of galectin-9, were able to target T lymphocytes and caused apoptosis in T cells expressing granzyme B. Furthermore, "opsonisation" of T cells by galectin-9 led to the translocation of phosphatidylserine onto the cell surface and subsequent phagocytosis by macrophages through Tim-3, the receptor, which recognises both galectin-9 and phosphatidylserine as ligands.


Asunto(s)
Apoptosis , Escherichia coli/metabolismo , Galectinas/metabolismo , Inmunidad Innata , Macrófagos/metabolismo , Opsonización , Linfocitos T/metabolismo , Citocinas/metabolismo , Escherichia coli/inmunología , Escherichia coli/patogenicidad , Granzimas/metabolismo , Interacciones Huésped-Patógeno , Humanos , Mediadores de Inflamación/metabolismo , Células Jurkat , Macrófagos/inmunología , Macrófagos/microbiología , Macrófagos/patología , Linfocitos T/inmunología , Linfocitos T/patología , Células THP-1
5.
Front Immunol ; 12: 675731, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34234778

RESUMEN

High mobility group box 1 (HMGB1) is a non-histone protein which is predominantly localised in the cell nucleus. However, stressed, dying, injured or dead cells can release this protein into the extracellular matrix passively. In addition, HMGB1 release was observed in cancer and immune cells where this process can be triggered by various endogenous as well as exogenous stimuli. Importantly, released HMGB1 acts as a so-called "danger signal" and could impact on the ability of cancer cells to escape host immune surveillance. However, the molecular mechanisms underlying the functional role of HMGB1 in determining the capability of human cancer cells to evade immune attack remain unclear. Here we report that the involvement of HMGB1 in anti-cancer immune evasion is determined by Toll-like receptor (TLR) 4, which recognises HMGB1 as a ligand. We found that HGMB1 induces TLR4-mediated production of transforming growth factor beta type 1 (TGF-ß), displaying autocrine/paracrine activities. TGF-ß induces production of the immunosuppressive protein galectin-9 in cancer cells. In TLR4-positive cancer cells, HMGB1 triggers the formation of an autocrine loop which induces galectin-9 expression. In malignant cells lacking TLR4, the same effect could be triggered by HMGB1 indirectly through TLR4-expressing myeloid cells present in the tumour microenvironment (e. g. tumour-associated macrophages).


Asunto(s)
Galectinas/biosíntesis , Proteína HMGB1/fisiología , Neoplasias/inmunología , Receptor Toll-Like 4/fisiología , Humanos , Tolerancia Inmunológica , Células THP-1 , Factor de Crecimiento Transformador beta1/fisiología
6.
Front Immunol ; 11: 580557, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33329552

RESUMEN

Acute myeloid leukemia (AML), a blood/bone marrow cancer, is a severe and often fatal malignancy. AML cells are capable of impairing the anti-cancer activities of cytotoxic lymphoid cells. This includes the inactivation of natural killer (NK) cells and killing of T lymphocytes. Here we report for the first time that V-domain Ig-containing suppressor of T cell activation (VISTA), a protein expressed by T cells, recognizes galectin-9 secreted by AML cells as a ligand. Importantly, we found that soluble VISTA released by AML cells enhances the effect of galectin-9, most likely by forming multiprotein complexes on the surface of T cells and possibly creating a molecular barrier. These events cause changes in the plasma membrane potential of T cells leading to activation of granzyme B inside cytotoxic T cells, resulting in apoptosis.


Asunto(s)
Antígenos B7/metabolismo , Galectinas/metabolismo , Linfocitos T Citotóxicos/inmunología , Antígenos de Neoplasias , Apoptosis , Citotoxicidad Inmunológica , Granzimas/metabolismo , Humanos , Terapia de Inmunosupresión , Ligandos , Potenciales de la Membrana , Unión Proteica , Multimerización de Proteína , Células THP-1 , Escape del Tumor
7.
Aging (Albany NY) ; 12(23): 23478-23496, 2020 12 08.
Artículo en Inglés | MEDLINE | ID: mdl-33295886

RESUMEN

Galectin-9 is one of the key proteins employed by a variety of human malignancies to suppress anti-cancer activities of cytotoxic lymphoid cells and thus escape immune surveillance. Human cancer cells in most cases express higher levels of galectin-9 compared to non-transformed cells. However, the biochemical mechanisms underlying this phenomenon remain unclear. Here we report for the first time that in human cancer as well as embryonic cells, the transcription factors hypoxia-inducible factor 1 (HIF-1) and activator protein 1 (AP-1) are involved in upregulation of transforming growth factor beta 1 (TGF-ß1) expression, leading to activation of the transcription factor Smad3 through autocrine action. This process triggers upregulation of galectin-9 expression in both malignant (mainly in breast and colorectal cancer as well as acute myeloid leukaemia (AML)) and embryonic cells. The effect, however, was not observed in mature non-transformed human cells. TGF-ß1-activated Smad3 therefore displays differential behaviour in human cancer and embryonic vs non-malignant cells. This study uncovered a self-supporting biochemical mechanism underlying high levels of galectin-9 expression operated by the human cancer and embryonic cells employed in our investigations. Our results suggest the possibility of using the TGF-ß1 signalling pathway as a potential highly efficient target for cancer immunotherapy.


Asunto(s)
Galectinas/metabolismo , Células Madre Embrionarias Humanas/metabolismo , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Neoplasias/metabolismo , Factor de Crecimiento Transformador beta1/metabolismo , Comunicación Autocrina , Galectinas/genética , Regulación Neoplásica de la Expresión Génica , Células HEK293 , Células HaCaT , Humanos , Subunidad alfa del Factor 1 Inducible por Hipoxia/genética , Células MCF-7 , Neoplasias/genética , Neoplasias/inmunología , Neoplasias/patología , Transducción de Señal , Proteína smad3/genética , Proteína smad3/metabolismo , Células THP-1 , Factor de Transcripción AP-1/genética , Factor de Transcripción AP-1/metabolismo , Factor de Crecimiento Transformador beta1/genética , Escape del Tumor , Hipoxia Tumoral , Microambiente Tumoral
8.
Front Immunol ; 10: 1594, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31354733

RESUMEN

Human cancer cells operate a variety of effective molecular and signaling mechanisms which allow them to escape host immune surveillance and thus progress the disease. We have recently reported that the immune receptor Tim-3 and its natural ligand galectin-9 are involved in the immune escape of human acute myeloid leukemia (AML) cells. These cells use the neuronal receptor latrophilin 1 (LPHN1) and its ligand fibronectin leucine rich transmembrane protein 3 (FLRT3, and possibly other ligands) to trigger the pathway. We hypothesized that the Tim-3-galectin-9 pathway may be involved in the immune escape of cancer cells of different origins. We found that studied breast tumors expressed significantly higher levels of both galectin-9 and Tim-3 compared to healthy breast tissues of the same patients and that these proteins were co-localized. Increased levels of LPHN2 and expressions of LPHN3 as well as FLRT3 were also detected in breast tumor cells. Activation of this pathway facilitated the translocation of galectin-9 onto the tumor cell surface, however no secretion of galectin-9 by tumor cells was observed. Surface-based galectin-9 was able to protect breast carcinoma cells against cytotoxic T cell-induced death. Furthermore, we found that cell lines from brain, colorectal, kidney, blood/mast cell, liver, prostate, lung, and skin cancers expressed detectable amounts of both Tim-3 and galectin-9 proteins. The majority of cell lines expressed one of the LPHN isoforms and FLRT3. We conclude that the Tim-3-galectin-9 pathway is operated by a wide range of human cancer cells and is possibly involved in prevention of anti-tumor immunity.


Asunto(s)
Neoplasias de la Mama/metabolismo , Galectinas/metabolismo , Receptor 2 Celular del Virus de la Hepatitis A/metabolismo , Línea Celular Tumoral , Femenino , Humanos , Leucemia Mieloide Aguda/metabolismo , Células MCF-7 , Glicoproteínas de Membrana/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Transducción de Señal/fisiología , Linfocitos T Citotóxicos/metabolismo
10.
Front Pharmacol ; 10: 333, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30984005

RESUMEN

Calcineurin inhibitors potentially prevent pro-allergic mediator release from basophils and mast cells but are rarely used systemically due to ubiquitous expressions of target signaling proteins. However, specific targeting of allergic effector cells with these inhibitors could circumvent unwanted side effects. We recently demonstrated the biocompatibility of gold nanoparticles (AuNPs) as a platform for non-toxic delivery of signaling inhibitors due to unique physicochemical properties of these nanomaterials. Since AuNPs can be conjugated with both anti-allergic drugs and antibodies or other proteins that specifically recognize basophils and mast cells, our aims were to assess specific targeting of allergic effector cell function using AuNPs conjugated with the calcineurin inhibitor ascomycin. Purified human basophils and LAD2 human mast cells were used for investigations with AuNPs conjugated either to CD203c antibodies or containing stem cell factor (SCF), respectively, which were amine-coupled to acidic groups of reduced glutathione (GSH). GSH was also used as a spacer for immobilization of ascomycin on the gold surface. AuNPs conjugated with anti-CD203c and ascomycin strikingly blocked IgE-dependent degranulation of both purified basophils and those present in mixed leukocyte preparations, suggesting specific targeting of these cells. In contrast, LAD2 mast cell responses were not inhibited using anti-CD203c-containing nanoconjugates but were when the conjugates contained SCF. Successful targeting of allergic effector cells using gold nanoconjugates indicates that this technology may have therapeutic potential for the treatment of allergies by specifically delivering highly effective signaling inhibitors with reduced side effects.

11.
Front Pharmacol ; 10: 342, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31024310

RESUMEN

The Tim-3-galectin-9 secretory pathway is known to protect various types of cancer cells against host immune surveillance. We found that pharmacologically induced mitochondrial dysfunction leads to a reduced galectin-9 expression/exocytosis in human colorectal cancer cells and re-distribution of this protein (the effect described for various cellular proteins) into mitochondria.

13.
Cell Mol Immunol ; 15(11): 989-991, 2018 11.
Artículo en Inglés | MEDLINE | ID: mdl-29872115

RESUMEN

Acute myeloid leukaemia (AML) is a blood/bone marrow cancer originating from myeloid cell precusors capable of self-renewing. AML cells implement biochemical mechanisms which allow them not only to survive, but also to successfully escape immune surveillance. ln this work, we discuss crucial molecular mechanisms used by human AML cells in order to evade immune attack.


Asunto(s)
Leucemia Mieloide Aguda , Escape del Tumor , Humanos , Leucemia Mieloide Aguda/inmunología , Leucemia Mieloide Aguda/metabolismo , Leucemia Mieloide Aguda/patología
14.
Oncoimmunology ; 7(6): e1438109, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29872582

RESUMEN

High mobility group box 1 (HMGB1) is a non-histone protein localised in the cell nucleus, where it interacts with DNA and promotes nuclear transcription events. HMGB1 levels are elevated during acute myeloid leukaemia (AML) progression followed by participation of this protein in triggering signalling events in target cells as a pro-inflammatory stimulus. This mechanism was hypothesised to be employed as a survival pathway by malignant blood cells and our aims were therefore to test this hypothesis experimentally. Here we report that HMGB1 triggers the release of tumour necrosis factor alpha (TNF-α) by primary human AML cells. TNF-α induces interleukin 1 beta (IL-1ß) production by healthy leukocytes, leading to IL-1ß-induced secretion of stem cell factor (SCF) by competent cells (for example endothelial cells). These results were verified in mouse bone marrow and primary human AML blood plasma samples. In addition, HMGB1 was found to induce secretion of angiogenic vascular endothelial growth factor (VEGF) and this process was dependent on the immune receptor Tim-3. We therefore conclude that HMGB1 is critical for AML progression as a ligand of Tim-3 and other immune receptors thus supporting survival/proliferation of AML cells and possibly the process of angiogenesis.

15.
Nanoscale ; 10(13): 5827-5833, 2018 Mar 29.
Artículo en Inglés | MEDLINE | ID: mdl-29538473

RESUMEN

In this study we used 5 nm gold nanoparticles as delivery platforms to target cancer cells expressing the immune receptor Tim-3 using single chain antibodies. Gold surfaces were also covered with the cytotoxic drug rapamycin which was immobilised using a glutathione linker. These nanoconjugates allowed highly specific and efficient delivery of cytotoxic rapamycin into human malignant blood cells.


Asunto(s)
Antineoplásicos/administración & dosificación , Sistemas de Liberación de Medicamentos , Leucemia Mieloide Aguda/tratamiento farmacológico , Nanoconjugados , Anticuerpos de Cadena Única/administración & dosificación , Galectinas/metabolismo , Oro , Receptor 2 Celular del Virus de la Hepatitis A/metabolismo , Humanos , Nanopartículas del Metal , Células THP-1
16.
EBioMedicine ; 22: 44-57, 2017 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-28750861

RESUMEN

Acute myeloid leukemia (AML) is a severe and often fatal systemic malignancy. Malignant cells are capable of escaping host immune surveillance by inactivating cytotoxic lymphoid cells. In this work we discovered a fundamental molecular pathway, which includes ligand-dependent activation of ectopically expressed latrophilin 1 and possibly other G-protein coupled receptors leading to increased translation and exocytosis of the immune receptor Tim-3 and its ligand galectin-9. This occurs in a protein kinase C and mTOR (mammalian target of rapamycin)-dependent manner. Tim-3 participates in galectin-9 secretion and is also released in a free soluble form. Galectin-9 impairs the anti-cancer activity of cytotoxic lymphoid cells including natural killer (NK) cells. Soluble Tim-3 prevents secretion of interleukin-2 (IL-2) required for the activation of cytotoxic lymphoid cells. These results were validated in ex vivo experiments using primary samples from AML patients. This pathway provides reliable targets for both highly specific diagnosis and immune therapy of AML.


Asunto(s)
Galectinas/metabolismo , Receptor 2 Celular del Virus de la Hepatitis A/metabolismo , Leucemia Mieloide Aguda/inmunología , Leucemia Mieloide Aguda/metabolismo , Animales , Comunicación Autocrina , Línea Celular Tumoral , Humanos , Interleucina-2/metabolismo , Células Jurkat , Células K562 , Células Asesinas Naturales/citología , Células Asesinas Naturales/inmunología , Ratones , Receptores Acoplados a Proteínas G/metabolismo , Receptores de Péptidos/metabolismo , Transducción de Señal , Células THP-1
17.
Oncotarget ; 7(29): 45575-45583, 2016 Jul 19.
Artículo en Inglés | MEDLINE | ID: mdl-27322212

RESUMEN

Acute myeloid leukaemia (AML) is a blood cancer affecting cells of myeloid lineage. It is characterised by rapid growth of malignant leukocytes that accumulate in the bone marrow and suppress normal haematopoiesis. This systemic disease remains a serious medical burden worldwide. Characterisation of protein antigens specifically expressed by malignant cells, but not by healthy leukocytes, is vital for the diagnostics and targeted treatment of AML. Here we report, for the first time, that the neuronal receptor latrophilin-1 is expressed in human monocytic leukaemia cell lines and in primary human AML cells. However, it is absent in healthy leukocytes. Latrophilin-1 is functional in leukaemia cells tested, and its biosynthesis is controlled through the mammalian target of rapamycin (mTOR), a master regulator of myeloid cell translational pathways. Our findings demonstrate that latrophilin-1 could be considered as a novel biomarker of human AML, which offers potential new avenues for AML diagnosis and treatment.


Asunto(s)
Biomarcadores de Tumor/análisis , Leucemia Mieloide Aguda/metabolismo , Receptores Acoplados a Proteínas G/biosíntesis , Receptores de Péptidos/biosíntesis , Humanos , Células Tumorales Cultivadas
18.
Oncotarget ; 6(30): 28678-92, 2015 Oct 06.
Artículo en Inglés | MEDLINE | ID: mdl-26384306

RESUMEN

Correction of human myeloid cell function is crucial for the prevention of inflammatory and allergic reactions as well as leukaemia progression. Caffeine, a naturally occurring food component, is known to display anti-inflammatory effects which have previously been ascribed largely to its inhibitory actions on phosphodiesterase. However, more recent studies suggest an additional role in affecting the activity of the mammalian target of rapamycin (mTOR), a master regulator of myeloid cell translational pathways, although detailed molecular events underlying its mode of action have not been elucidated. Here, we report the cellular uptake of caffeine, without metabolisation, by healthy and malignant hematopoietic myeloid cells including monocytes, basophils and primary acute myeloid leukaemia mononuclear blasts. Unmodified caffeine downregulated mTOR signalling, which affected glycolysis and the release of pro-inflammatory/pro-angiogenic cytokines as well as other inflammatory mediators. In monocytes, the effects of caffeine were potentiated by its ability to inhibit xanthine oxidase, an enzyme which plays a central role in human purine catabolism by generating uric acid. In basophils, caffeine also increased intracellular cyclic adenosine monophosphate (cAMP) levels which further enhanced its inhibitory action on mTOR. These results demonstrate an important mode of pharmacological action of caffeine with potentially wide-ranging therapeutic impact for treating non-infectious disorders of the human immune system, where it could be applied directly to inflammatory cells.


Asunto(s)
Cafeína/farmacología , Linaje de la Célula , Células Mieloides/efectos de los fármacos , Serina-Treonina Quinasas TOR/metabolismo , Xantina Oxidasa/metabolismo , Proteínas Angiogénicas/metabolismo , Basófilos/efectos de los fármacos , Basófilos/enzimología , Cafeína/metabolismo , Línea Celular Tumoral , Citocinas/metabolismo , Relación Dosis-Respuesta a Droga , Regulación hacia Abajo , Glucólisis/efectos de los fármacos , Humanos , Mediadores de Inflamación/metabolismo , Leucemia Mieloide Aguda/enzimología , Leucemia Mieloide Aguda/patología , Monocitos/efectos de los fármacos , Monocitos/enzimología , Células Mieloides/enzimología , Células Mieloides/patología , Transducción de Señal/efectos de los fármacos
19.
J Biomed Nanotechnol ; 10(7): 1259-66, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24804546

RESUMEN

In this study we demonstrate for the first time a new approach for highly specific targeting of viable human leukocytes by functionalised gold nanoconjugates made on the basis of citrate-stabilised gold nanoparticles. This immobilisation involves highly specific recognition of the cells of interest by plasma membrane-associated receptor ligands or functional antibodies attached to gold nanoconjugates. Using this technology we managed to deliver pharmacological inhibitors into the target cells. Also we immobilized fully functional human leukocytes utilising both cell lines and rare primary human cells (basophils). This approach demonstrates possibilities of using nanoconjugates for diagnostics, drug delivery and immune therapy.


Asunto(s)
Oro/farmacología , Leucocitos/citología , Nanopartículas del Metal/química , Nanoconjugados/química , Anticuerpos/metabolismo , Basófilos/citología , Basófilos/efectos de los fármacos , Basófilos/metabolismo , Materiales Biocompatibles/farmacología , Línea Celular , Células Inmovilizadas/citología , Células Inmovilizadas/efectos de los fármacos , Células Inmovilizadas/metabolismo , Ensayo de Inmunoadsorción Enzimática , Humanos , Leucocitos/efectos de los fármacos , Leucocitos/metabolismo , Lipopolisacáridos/farmacología , Tacrolimus/análogos & derivados , Tacrolimus/farmacología
20.
Cell Mol Life Sci ; 71(4): 699-710, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-23872956

RESUMEN

Mammalian myeloid cells are crucial effectors of host innate immune defense. Normal and pathological responses of these cells require adaptation to signaling stress through the hypoxia-inducible factor 1 (HIF-1) transcription complex. Adapted cells activate the mammalian target of rapamycin (mTOR), via S2448 phosphorylation, which induces de novo translation of vital signaling proteins. However, the molecular mechanisms underlying this signaling dogma remain unclear. Here, we demonstrate for the first time that inactivation of HIF-1, by silencing its inducible alpha subunit, significantly decreases mTOR S2448 phosphorylation caused by ligand-dependent activation of human myeloid leukemia cells. This shows that HIF-1 is essential for the activation of mTOR and serves at a crucial juncture of myeloid cell function in both in vitro and in vivo systems.


Asunto(s)
Factor 1 Inducible por Hipoxia/metabolismo , Células Mieloides/metabolismo , Serina-Treonina Quinasas TOR/metabolismo , Proteínas Quinasas Activadas por AMP/metabolismo , Animales , Células Sanguíneas/metabolismo , Línea Celular , Activación Enzimática , Humanos , Factor 1 Inducible por Hipoxia/genética , Masculino , Ratones , Fosforilación , Interferencia de ARN , ARN Interferente Pequeño/genética , Receptor Toll-Like 2/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...