Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
1.
Blood ; 133(5): 446-456, 2019 01 31.
Artículo en Inglés | MEDLINE | ID: mdl-30401703

RESUMEN

Acute myeloid leukemia (AML) is an age-related disease that is highly dependent on the bone marrow (BM) microenvironment. With increasing age, tissues accumulate senescent cells, characterized by an irreversible arrest of cell proliferation and the secretion of a set of proinflammatory cytokines, chemokines, and growth factors, collectively known as the senescence-associated secretory phenotype (SASP). Here, we report that AML blasts induce a senescent phenotype in the stromal cells within the BM microenvironment and that the BM stromal cell senescence is driven by p16INK4a expression. The p16INK4a-expressing senescent stromal cells then feed back to promote AML blast survival and proliferation via the SASP. Importantly, selective elimination of p16INK4a+ senescent BM stromal cells in vivo improved the survival of mice with leukemia. Next, we find that the leukemia-driven senescent tumor microenvironment is caused by AML-induced NOX2-derived superoxide. Finally, using the p16-3MR mouse model, we show that by targeting NOX2 we reduced BM stromal cell senescence and consequently reduced AML proliferation. Together, these data identify leukemia-generated NOX2-derived superoxide as a driver of protumoral p16INK4a-dependent senescence in BM stromal cells. Our findings reveal the importance of a senescent microenvironment for the pathophysiology of leukemia. These data now open the door to investigate drugs that specifically target the "benign" senescent cells that surround and support AML.


Asunto(s)
Médula Ósea/patología , Senescencia Celular , Inhibidor p16 de la Quinasa Dependiente de Ciclina/metabolismo , Leucemia Mieloide Aguda/patología , Microambiente Tumoral , Animales , Médula Ósea/metabolismo , Proliferación Celular , Técnicas de Cocultivo , Femenino , Humanos , Leucemia Mieloide Aguda/metabolismo , Células Madre Mesenquimatosas/metabolismo , Células Madre Mesenquimatosas/patología , Ratones Endogámicos C57BL , NADPH Oxidasa 2/metabolismo , Superóxidos/metabolismo , Células Tumorales Cultivadas
2.
Blood ; 129(10): 1320-1332, 2017 03 09.
Artículo en Inglés | MEDLINE | ID: mdl-28049638

RESUMEN

Despite currently available therapies, most patients diagnosed with acute myeloid leukemia (AML) die of their disease. Tumor-host interactions are critical for the survival and proliferation of cancer cells; accordingly, we hypothesize that specific targeting of the tumor microenvironment may constitute an alternative or additional strategy to conventional tumor-directed chemotherapy. Because adipocytes have been shown to promote breast and prostate cancer proliferation, and because the bone marrow adipose tissue accounts for up to 70% of bone marrow volume in adult humans, we examined the adipocyte-leukemia cell interactions to determine if they are essential for the growth and survival of AML. Using in vivo and in vitro models of AML, we show that bone marrow adipocytes from the tumor microenvironment support the survival and proliferation of malignant cells from patients with AML. We show that AML blasts alter metabolic processes in adipocytes to induce phosphorylation of hormone-sensitive lipase and consequently activate lipolysis, which then enables the transfer of fatty acids from adipocytes to AML blasts. In addition, we report that fatty acid binding protein-4 (FABP4) messenger RNA is upregulated in adipocytes and AML when in coculture. FABP4 inhibition using FABP4 short hairpin RNA knockdown or a small molecule inhibitor prevents AML proliferation on adipocytes. Moreover, knockdown of FABP4 increases survival in Hoxa9/Meis1-driven AML model. Finally, knockdown of carnitine palmitoyltransferase IA in an AML patient-derived xenograft model improves survival. Here, we report the first description of AML programming bone marrow adipocytes to generate a protumoral microenvironment.


Asunto(s)
Adipocitos/patología , Células de la Médula Ósea/patología , Leucemia Mieloide Aguda/patología , Microambiente Tumoral/fisiología , Adipocitos/metabolismo , Adulto , Anciano , Anciano de 80 o más Años , Animales , Western Blotting , Células de la Médula Ósea/metabolismo , Técnicas de Cocultivo , Proteínas de Unión a Ácidos Grasos/metabolismo , Femenino , Citometría de Flujo , Xenoinjertos , Humanos , Inmunohistoquímica , Leucemia Mieloide Aguda/metabolismo , Masculino , Ratones , Persona de Mediana Edad , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
3.
Womens Health (Lond) ; 20: 17455057241231420, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38385267

RESUMEN

BACKGROUND: Menstrual hygiene management plays a significant role in the achievement of sustainable development goals, including good health and well-being, quality education, gender equality, women's empowerment, and sanitation. Even though the United Nations emphasizes menstrual hygiene management as a global public health and human rights issue, studies from different parts of the world have shown there is a lack of policies in this area that affects adolescent girls. OBJECTIVE: The study aimed to assess the menstrual-related knowledge and practices of adolescent girls (10-18 years) attending urban and rural public schools of district Khairpur of province Sindh of Pakistan. DESIGN: This research is an analytical cross-sectional study design. METHODS: Adolescent girls (n = 310), 159 girls from urban and 159 girls from rural schools, were enrolled. Data were collected through self-administered questionnaires. Knowledge was considered satisfactory if the participant scored ⩾12/20, and <12/20 was considered unsatisfactory knowledge. Practice scores were considered good for >10 and poor for <10. Statistical Package for the Social Science (SPSS) version 20.0 was used for data analysis. Descriptive statistics such as mean and standard deviation were calculated. The chi-square test was used to assess and compare the differences between practices and knowledge of menstrual hygiene management between urban and rural adolescent school girls. RESULTS: We found significant differences in menstrual hygiene management among adolescent girls between urban and rural areas. Approximately two-thirds of the total school girls thought that school management had inadequate resources and toilet facilities (rural: 76% versus urban: 62%). The major source of knowledge in adolescent rural and urban girls for menstrual-related matters was mothers. More girls from the urban school (81%) were aware of using sanitary pads during menstruation. The majority of the girls from urban areas showed satisfactory knowledge whereas only 38% of rural girls were shown to have satisfactory results. Nearly 71% of urban girls were found to have good practices when compared with rural girls (12%). CONCLUSION: With further importance by the United Nations on female education in less developed regions, where the health of women is usually given less importance, policies are required on the inclusion of reproductive health of female adolescents in the curriculum.


Asunto(s)
Higiene , Menstruación , Femenino , Humanos , Adolescente , Estudios Transversales , Pakistán , Conocimientos, Actitudes y Práctica en Salud
4.
Cancers (Basel) ; 16(3)2024 Jan 29.
Artículo en Inglés | MEDLINE | ID: mdl-38339323

RESUMEN

BACKGROUND: Acute myeloid leukemia (AML) is the malignant proliferation of immature myeloid cells characterized by a block in differentiation. As such, novel therapeutic strategies to promote the differentiation of immature myeloid cells have been successful in AML, although these agents are targeted to a specific mutation that is only present in a subset of AML patients. In the current study, we show that targeting the epigenetic modifier enhancer of zeste homolog 2 (EZH2) can induce the differentiation of immature blast cells into a more mature myeloid phenotype and promote survival in AML murine models. METHODS: The EZH2 inhibitor EPZ011989 (EPZ) was studied in AML cell lines, primary in AML cells and normal CD34+ stem cells. A pharmacodynamic assessment of H3K27me3; studies of differentiation, cell growth, and colony formation; and in vivo therapeutic studies including the influence on primary AML cell engraftment were also conducted. RESULTS: EPZ inhibited H3K27me3 in AML cell lines and primary AML samples in vitro. EZH2 inhibition reduced colony formation in multiple AML cell lines and primary AML samples, while exhibiting no effect on colony formation in normal CD34+ stem cells. In AML cells, EPZ promoted phenotypic evidence of differentiation. Finally, the pretreatment of primary AML cells with EPZ significantly delayed engraftment and prolonged the overall survival when engrafted into immunodeficient mice. CONCLUSIONS: Despite evidence that EZH2 silencing in MDS/MPN can promote AML pathogenesis, our data demonstrate that the therapeutic inhibition of EZH2 in established AML has the potential to improve survival.

5.
Cells ; 12(16)2023 08 11.
Artículo en Inglés | MEDLINE | ID: mdl-37626855

RESUMEN

Cellular senescence is a durable cell cycle arrest as a result of the finite proliferative capacity of cells. Senescence responds to both intrinsic and extrinsic cellular stresses, such as aging, mitochondrial dysfunction, irradiation, and chemotherapy. Here, we report on the use of mass cytometry (MC) to analyze multiple model systems and demonstrate MC as a platform for senescence analysis at the single-cell level. We demonstrate changes to p16 expression, cell cycling fraction, and histone tail modifications in several established senescent model systems and using isolated human T cells. In bone marrow mesenchymal stromal cells (BMSCs), we show increased p16 expression with subsequent passage as well as a reduction in cycling cells and open chromatin marks. In WI-38 cells, we demonstrate increased p16 expression with both culture-induced senescence and oxidative stress-induced senescence (OSIS). We also use Wanderlust, a trajectory analysis tool, to demonstrate how p16 expression changes with histone tail modifications and cell cycle proteins. Finally, we demonstrate that repetitive stimulation of human T cells with CD3/CD28 beads induces an exhausted phenotype with increased p16 expression. This p16-expressing population exhibited higher expression of exhaustion markers such as EOMES and TOX. This work demonstrates that MC is a useful platform for studying senescence at a single-cell protein level, and is capable of measuring multiple markers of senescence at once with high confidence, thereby improving our understanding of senescent pathways.


Asunto(s)
Histonas , Investigación , Humanos , Envejecimiento , Antígenos CD28 , Ciclo Celular
6.
Br J Haematol ; 159(2): 191-203, 2012 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-22934750

RESUMEN

The novel multi-kinase inhibitor TG02 has selectivity against cell cycle and transcriptional cyclin dependent kinases (CDKs) as well as fms-like tyrosine kinase receptor-3 (FLT3). Inhibition of transcriptional CDKs preferentially depletes short-lived proteins such as MCL1. We evaluated the in vitro toxicity of TG02 to primary acute myeloid leukaemia (AML) cells in the presence of survival signalling pathway activation by cytokines and fibronectin. One hundred nanomolar TG02 induced a median decrease of 40% in bulk cell survival and 43% in the CD34(+) CD38(-) CD123(+) subset. A 90% inhibitory concentration of 500 nmol/l indicated that TG02 toxicity is not halted by protective cell cycle arrest. Samples with FLT3 internal tandem duplication were not preferentially targeted. By flow cytometry, TG02 treatment caused loss of RNA Polymerase II serine 2 phosphorylation in patient samples, which correlated strongly with BAX activation (R(2) =0·89), suggesting these as potential biomarkers for clinical studies. MCL1 and XIAP expression also decreased. Repeated brief exposure to TG02 in MOLM-13 cells did not result in compensatory up-regulation of survival protein expression. In conclusion, TG02 is potently cytotoxic towards CD34(+) CD38(-) CD123(+) and bulk AML cells, despite protective signalling pathway activation. This antitumour activity is most likely mediated by dephosphorylation of RNA Polymerase II leading to depletion of survival molecules such as MCL1 and XIAP, with subsequent BAX activation and apoptosis.


Asunto(s)
Apoptosis/efectos de los fármacos , Compuestos Heterocíclicos de 4 o más Anillos/farmacología , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Transducción de Señal/efectos de los fármacos , Proteína Inhibidora de la Apoptosis Ligada a X/metabolismo , Antígenos CD , Ensayos de Selección de Medicamentos Antitumorales , Femenino , Células HL-60 , Humanos , Leucemia Mieloide Aguda , Masculino , Proteína 1 de la Secuencia de Leucemia de Células Mieloides , Fosforilación/efectos de los fármacos , ARN Polimerasa II/metabolismo , Células U937 , Proteína X Asociada a bcl-2/metabolismo
7.
Front Microbiol ; 11: 474, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32351457

RESUMEN

Induction of the AR2 acid response system of Escherichia coli occurs at a moderately low pH (pH 5.5) and leads to high levels of resistance to pH levels below 2.5 in the presence of glutamate. Induction is mediated in part by the EvgAS two component system. Here, we show that the bacterial signaling molecule indole inhibits the induction of key promoters in the AR2 system and blocks the development of glutamate-dependent acid resistance. The addition of tryptophan, the precursor for indole biosynthesis, had the same effects, and this block was relieved in a tnaA mutant, which is unable to synthesize indole. Expression of a constitutively active EvgS protein was able to relieve the inhibition caused by indole, consistent with EvgS being inhibited directly or indirectly by indole. Indole had no effect on autophosphorylation of the isolated cytoplasmic domain of EvgS. This is consistent with a model where indole directly or indirectly affects the ability of EvgS to detect its inducing signal or to transduce this information across the cytoplasmic membrane. The inhibitory activity of indole on the AR2 system is not related to its ability to act as an ionophore, and, conversely, the ionophore CCCP had no effect on acid-induced AR2 promoter activity, showing that the proton motive force is unlikely to be a signal for induction of the AR2 system.

8.
Cancer Lett ; 412: 37-45, 2018 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-29031566

RESUMEN

Multiple myeloma (MM) is an incurable disease characterized by clonal plasma cell proliferation. The stress response transcription factor Nuclear factor erythroid 2 [NF-E2]-related factor 2 (NRF2) is known to be activated in MM in response to proteasome inhibitors (PI). Here, we hypothesize that the transcription factor NRF2 whose physiological role is to protect cells from reactive oxygen species via the regulation of drug metabolism and antioxidant gene plays an important role in MM cells survival and proliferation. We report for the first time that NRF2 is constitutively activated in circa 50% of MM primary samples and all MM cell lines. Moreover, genetic inhibition of constitutively expressed NRF2 reduced MM cell viability. We confirm that PI induced further expression of NRF2 in MM cell lines and primary MM. Furthermore, genetic inhibition of NRF2 of PI treated MM cells increased ER-stress through the regulation of CCAAT-enhancer-binding protein homologous protein (CHOP). Finally, inhibition of NRF2 in combination with PI treatment significantly increased apoptosis in MM cells. Here we identify NRF2 as a key regulator of MM survival in treatment naive and PI treated cells.


Asunto(s)
Apoptosis , Estrés del Retículo Endoplásmico/fisiología , Mieloma Múltiple/patología , Factor 2 Relacionado con NF-E2/fisiología , Línea Celular Tumoral , Glutatión/metabolismo , Humanos , Mieloma Múltiple/tratamiento farmacológico , Mieloma Múltiple/metabolismo , Factor 2 Relacionado con NF-E2/análisis , Factor 2 Relacionado con NF-E2/antagonistas & inhibidores , Inhibidores de Proteasoma/farmacología , Factor de Transcripción CHOP/análisis
9.
PLoS One ; 13(6): e0199361, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29928062

RESUMEN

The presence of cancer stem cells (CSCs) and the induction of epithelial-to-mesenchymal transition (EMT) in tumors are associated with tumor aggressiveness, metastasis, drug resistance, and poor prognosis, necessitating the development of reagents for unambiguous detection of CSC- and EMT-associated proteins in tumor specimens. To this end, we generated novel antibodies to EMT- and CSC-associated proteins, including Goosecoid, Sox9, Slug, Snail, and CD133. Importantly, unlike several widely used antibodies to CD133, the anti-CD133 antibodies we generated recognize epitopes distal to known glycosylation sites, enabling analyses that are not confounded by differences in CD133 glycosylation. For all target proteins, we selected antibodies that yielded the expected target protein molecular weights by Western analysis and the correct subcellular localization patterns by immunofluorescence microscopy assay (IFA); binding selectivity was verified by immunoprecipitation-mass spectrometry and by immunohistochemistry and IFA peptide blocking experiments. Finally, we applied these reagents to assess modulation of the respective markers of EMT and CSCs in xenograft tumor models by IFA. We observed that the constitutive presence of human hepatocyte growth factor (hHGF) in the tumor microenvironment of H596 non-small cell lung cancer tumors implanted in homozygous hHGF knock-in transgenic mice induced a more mesenchymal-like tumor state (relative to the epithelial-like state when implanted in control SCID mice), as evidenced by the elevated expression of EMT-associated transcription factors detected by our novel antibodies. Similarly, our new anti-CD133 antibody enabled detection and quantitation of drug-induced reductions in CD133-positive tumor cells following treatment of SUM149PT triple-negative breast cancer xenograft models with the CSC/focal adhesion kinase (FAK) inhibitor VS-6063. Thus, our novel antibodies to CSC- and EMT-associated factors exhibit sufficient sensitivity and selectivity for immunofluorescence microscopy studies of these processes in preclinical xenograft tumor specimens and the potential for application with clinical samples.


Asunto(s)
Anticuerpos Monoclonales/farmacología , Antineoplásicos/farmacología , Transición Epitelial-Mesenquimal/efectos de los fármacos , Células Madre Neoplásicas/patología , Microambiente Tumoral/efectos de los fármacos , Antígeno AC133/metabolismo , Animales , Anticuerpos Monoclonales/biosíntesis , Antineoplásicos/uso terapéutico , Benzamidas/farmacología , Benzamidas/uso terapéutico , Carcinoma de Pulmón de Células no Pequeñas/metabolismo , Carcinoma de Pulmón de Células no Pequeñas/patología , Línea Celular Tumoral , Femenino , Técnicas de Sustitución del Gen , Factor de Crecimiento de Hepatocito/genética , Humanos , Indicadores y Reactivos , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/patología , Ratones Transgénicos , Células Madre Neoplásicas/efectos de los fármacos , Células Madre Neoplásicas/metabolismo , Fenotipo , Pirazinas/farmacología , Pirazinas/uso terapéutico , Sulfonamidas/farmacología , Sulfonamidas/uso terapéutico , Neoplasias de la Mama Triple Negativas/tratamiento farmacológico , Neoplasias de la Mama Triple Negativas/patología , Ensayos Antitumor por Modelo de Xenoinjerto
10.
J Hematol Oncol ; 11(1): 66, 2018 05 16.
Artículo en Inglés | MEDLINE | ID: mdl-29769142

RESUMEN

Multiple myeloma (MM) remains an incurable malignancy despite the recent advancements in its treatment. The protective effects of the niche in which it develops has been well documented; however, little has been done to investigate the MM cell's ability to 're-program' cells within its environment to benefit disease progression. Here, we show that MM-derived macrophage migratory inhibitory factor (MIF) stimulates bone marrow stromal cells to produce the disease critical cytokines IL-6 and IL-8, prior to any cell-cell contact. Furthermore, we provide evidence that this IL-6/8 production is mediated by the transcription factor cMYC. Pharmacological inhibition of cMYC in vivo using JQ1 led to significantly decreased levels of serum IL-6-a highly positive prognostic marker in MM patients. CONCLUSIONS: Our presented findings show that MM-derived MIF causes BMSC secretion of IL-6 and IL-8 via BMSC cMYC. Furthermore, we show that the cMYC inhibitor JQ1 can reduce BMSC secreted IL-6 in vivo, irrespective of tumor burden. These data provide evidence for the clinical evaluation of both MIF and cMYC inhibitors in the treatment of MM.


Asunto(s)
Células de la Médula Ósea/patología , Interleucina-6/metabolismo , Oxidorreductasas Intramoleculares/fisiología , Factores Inhibidores de la Migración de Macrófagos/fisiología , Mieloma Múltiple/química , Células del Estroma/patología , Humanos , Interleucina-8/metabolismo , Proteínas Proto-Oncogénicas c-myc/metabolismo
11.
Oncogene ; 37(20): 2676-2686, 2018 05.
Artículo en Inglés | MEDLINE | ID: mdl-29487418

RESUMEN

Approximately 80% of patients diagnosed with acute myeloid leukemia (AML) die as a consequence of failure to eradicate the tumor from the bone marrow microenvironment. We have recently shown that stroma-derived interleukin-8 (IL-8) promotes AML growth and survival in the bone marrow in response to AML-derived macrophage migration inhibitory factor (MIF). In the present study we show that high constitutive expression of MIF in AML blasts in the bone marrow is hypoxia-driven and, through knockdown of MIF, HIF1α and HIF2α, establish that hypoxia supports AML tumor proliferation through HIF1α signaling. In vivo targeting of leukemic cell HIF1α inhibits AML proliferation in the tumor microenvironment through transcriptional regulation of MIF, but inhibition of HIF2α had no measurable effect on AML blast survival. Functionally, targeted inhibition of MIF in vivo improves survival in models of AML. Here we present a mechanism linking HIF1α to a pro-tumoral chemokine factor signaling pathway and in doing so, we establish a potential strategy to target AML.


Asunto(s)
Médula Ósea/metabolismo , Subunidad alfa del Factor 1 Inducible por Hipoxia/genética , Oxidorreductasas Intramoleculares/genética , Leucemia Mieloide Aguda/genética , Factores Inhibidores de la Migración de Macrófagos/genética , Regulación hacia Arriba , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Hipoxia de la Célula , Línea Celular Tumoral , Proliferación Celular , Supervivencia Celular , Femenino , Técnicas de Silenciamiento del Gen , Humanos , Oxidorreductasas Intramoleculares/metabolismo , Leucemia Mieloide Aguda/metabolismo , Leucemia Mieloide Aguda/patología , Factores Inhibidores de la Migración de Macrófagos/metabolismo , Ratones , Trasplante de Neoplasias , Transducción de Señal , Microambiente Tumoral
12.
J Am Heart Assoc ; 6(5)2017 May 03.
Artículo en Inglés | MEDLINE | ID: mdl-28468787

RESUMEN

BACKGROUND: Plaque erosion causes 30% of ST-segment elevation myocardial infarctions, but the underlying cause is unknown. Inflammatory infiltrates are less abundant in erosion compared with rupture in autopsy studies. We hypothesized that erosion and rupture are associated with significant differences in intracoronary cytokines in vivo. METHODS AND RESULTS: Forty ST-segment elevation myocardial infarction patients with <6 hours of chest pain were classified as ruptured fibrous cap (RFC) or intact fibrous cap (IFC) using optical coherence tomography. Plasma samples from the infarct-related artery and a peripheral artery were analyzed for expression of 102 cytokines using arrays; results were confirmed with ELISA. Thrombectomy samples were analyzed for differential mRNA expression using quantitative real-time polymerase chain reaction. Twenty-three lesions were classified as RFC (58%), 15 as IFC (38%), and 2 were undefined (4%). In addition, 12% (12 of 102) of cytokines were differentially expressed in both coronary and peripheral plasma. I-TAC was preferentially expressed in RFC (significance analysis of microarrays adjusted P<0.001; ELISA IFC 10.2 versus RFC 10.8 log2 pg/mL; P=0.042). IFC was associated with preferential expression of epidermal growth factor (significance analysis of microarrays adjusted P<0.001; ELISA IFC 7.42 versus RFC 6.63 log2 pg/mL, P=0.036) and thrombospondin 1 (significance analysis of microarrays adjusted P=0.03; ELISA IFC 10.4 versus RFC 8.65 log2 ng/mL, P=0.0041). Thrombectomy mRNA showed elevated I-TAC in RFC (P=0.0007) epidermal growth factor expression in IFC (P=0.0264) but no differences in expression of thrombospondin 1. CONCLUSIONS: These results demonstrate differential intracoronary cytokine expression in RFC and IFC. Elevated thrombospondin 1 and epidermal growth factor may play an etiological role in erosion.


Asunto(s)
Enfermedad de la Arteria Coronaria/complicaciones , Citocinas/sangre , Mediadores de Inflamación/sangre , Placa Aterosclerótica , Infarto del Miocardio con Elevación del ST/etiología , Anciano , Biomarcadores/sangre , Angiografía Coronaria , Enfermedad de la Arteria Coronaria/sangre , Enfermedad de la Arteria Coronaria/diagnóstico por imagen , Enfermedad de la Arteria Coronaria/terapia , Citocinas/genética , Ensayo de Inmunoadsorción Enzimática , Factor de Crecimiento Epidérmico/sangre , Femenino , Fibrosis , Perfilación de la Expresión Génica/métodos , Humanos , Masculino , Persona de Mediana Edad , Análisis de Secuencia por Matrices de Oligonucleótidos , Intervención Coronaria Percutánea , Estudios Prospectivos , Reacción en Cadena en Tiempo Real de la Polimerasa , Factores de Riesgo , Rotura Espontánea , Infarto del Miocardio con Elevación del ST/sangre , Infarto del Miocardio con Elevación del ST/diagnóstico por imagen , Infarto del Miocardio con Elevación del ST/terapia , Trombectomía , Trombospondina 1/sangre , Tomografía de Coherencia Óptica
13.
Cancer Res ; 77(2): 303-311, 2017 01 15.
Artículo en Inglés | MEDLINE | ID: mdl-27872094

RESUMEN

Acute myeloid leukemia (AML) cells exhibit a high level of spontaneous apoptosis when cultured in vitro but have a prolonged survival time in vivo, indicating that tissue microenvironment plays a critical role in promoting AML cell survival. In vitro studies have shown that bone marrow mesenchymal stromal cells (BM-MSC) protect AML blasts from spontaneous and chemotherapy-induced apoptosis. Here, we report a novel interaction between AML blasts and BM-MSCs, which benefits AML proliferation and survival. We initially examined the cytokine profile in cultured human AML compared with AML cultured with BM-MSCs and found that macrophage migration inhibitory factor (MIF) was highly expressed by primary AML, and that IL8 was increased in AML/BM-MSC cocultures. Recombinant MIF increased IL8 expression in BM-MSCs via its receptor CD74. Moreover, the MIF inhibitor ISO-1 inhibited AML-induced IL8 expression by BM-MSCs as well as BM-MSC-induced AML survival. Protein kinase C ß (PKCß) regulated MIF-induced IL8 in BM-MSCs. Finally, targeted IL8 shRNA inhibited BM-MSC-induced AML survival. These results describe a novel, bidirectional, prosurvival mechanism between AML blasts and BM-MSCs. Furthermore, they provide biologic rationale for therapeutic strategies in AML targeting the microenvironment, specifically MIF and IL8. Cancer Res; 77(2); 303-11. ©2016 AACR.


Asunto(s)
Interleucina-8/metabolismo , Oxidorreductasas Intramoleculares/metabolismo , Leucemia Mieloide Aguda/patología , Factores Inhibidores de la Migración de Macrófagos/metabolismo , Células Madre Mesenquimatosas/metabolismo , Proteína Quinasa C beta/metabolismo , Western Blotting , Supervivencia Celular , Técnicas de Cocultivo , Ensayo de Inmunoadsorción Enzimática , Citometría de Flujo , Humanos , Leucemia Mieloide Aguda/metabolismo , Reacción en Cadena en Tiempo Real de la Polimerasa , Transducción de Señal/fisiología , Microambiente Tumoral/fisiología
14.
Oncotarget ; 8(10): 16220-16232, 2017 Mar 07.
Artículo en Inglés | MEDLINE | ID: mdl-27092880

RESUMEN

Direct co-operation between sensitiser molecules BAD and NOXA in mediating apoptosis suggests that therapeutic agents which sensitise to BAD may complement agents which sensitise to NOXA. Dynamic BH3 profiling is a novel methodology that we have applied to the measurement of complementarity between sensitiser BH3 peptide mimetics and therapeutic agents. Using dynamic BH3 profiling, we show that the agent TG02, which downregulates MCL-1, sensitises to the BCL-2-inhibitory BAD-BH3 peptide, whereas the BCL-2 antagonist ABT-199 sensitises to MCL-1 inhibitory NOXA-BH3 peptide in acute myeloid leukaemia (AML) cells. At the concentrations used, the peptides did not trigger mitochondrial outer membrane permeabilisation in their own right, but primed cells to release Cytochrome C in the presence of an appropriate trigger of a complementary pathway. In KG-1a cells TG02 and ABT-199 synergised to induce apoptosis. In heterogeneous AML patient samples we noted a range of sensitivities to the two agents. Although some individual samples markedly favoured one agent or the other, in the group as a whole the combination of TG02 + ABT-199 was significantly more cytotoxic than either agent individually. We conclude that dynamic NOXA and BAD BH3 profiling is a sensitive methodology for investigating molecular pathways of drug action and complementary mechanisms of chemoresponsiveness.


Asunto(s)
Apoptosis/efectos de los fármacos , Compuestos Bicíclicos Heterocíclicos con Puentes/farmacología , Compuestos Heterocíclicos de 4 o más Anillos/farmacología , Leucemia Mieloide/tratamiento farmacológico , Sulfonamidas/farmacología , Enfermedad Aguda , Antineoplásicos/farmacología , Apoptosis/genética , Materiales Biomiméticos/farmacología , Western Blotting , Línea Celular Tumoral , Ciclina D1/antagonistas & inhibidores , Ciclina D1/genética , Ciclina D1/metabolismo , Citocromos c/metabolismo , Sinergismo Farmacológico , Regulación Leucémica de la Expresión Génica/efectos de los fármacos , Humanos , Leucemia Mieloide/genética , Leucemia Mieloide/metabolismo , Proteína 1 de la Secuencia de Leucemia de Células Mieloides/genética , Proteína 1 de la Secuencia de Leucemia de Células Mieloides/metabolismo , Fragmentos de Péptidos/farmacología , Proteínas Proto-Oncogénicas/farmacología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
15.
Oncotarget ; 7(26): 39784-39795, 2016 Jun 28.
Artículo en Inglés | MEDLINE | ID: mdl-27174919

RESUMEN

Phosphoinositide-3-kinase (PI3K) is an enzyme group, known to regulate key survival pathways in acute myeloid leukaemia (AML). It generates phosphatidylinositol-3,4,5-triphosphate, which provides a membrane docking site for protein kinaseB activation. PI3K catalytic p110 subunits are divided into 4 isoforms; α,ß,δ and γ. The PI3Kδ isoform is always expressed in AML cells, whereas the frequency of PI3Kγ expression is highly variable. The functions of these individual catalytic enzymes have not been fully resolved in AML, therefore using the PI3K p110δ and p110γ-targeted inhibitor IPI-145 (duvelisib) and specific p110δ and p110γ shRNA, we analysed the role of these two p110 subunits in human AML blast survival. The results show that PI3Kδ and PI3Kγ inhibition with IPI-145 has anti-proliferative activity in primary AML cells by inhibiting the activity of AKT and MAPK. Pre-treatment of AML cells with IPI-145 inhibits both adhesion and migration of AML blasts to bone marrow stromal cells. Using shRNA targeted to the individual isoforms we demonstrated that p110δ-knockdown had a more significant anti-proliferative effect on AML cells, whereas targeting p110γ-knockdown significantly inhibited AML migration. The results demonstrate that targeting both PI3Kδ and PI3Kγ to inhibit AML-BMSC interactions provides a biologic rationale for the pre-clinical evaluation of IPI-145 in AML.


Asunto(s)
Células de la Médula Ósea/citología , Fosfatidilinositol 3-Quinasa Clase I/metabolismo , Fosfatidilinositol 3-Quinasa Clase Ib/metabolismo , Leucemia Mieloide Aguda/metabolismo , Células Madre Mesenquimatosas/citología , Adhesión Celular , Movimiento Celular , Proliferación Celular , Supervivencia Celular , Dermatoglifia del ADN , Regulación Leucémica de la Expresión Génica , Humanos , Isoquinolinas/farmacología , Leucemia Mieloide Aguda/genética , Fosforilación , Purinas/farmacología , ARN Interferente Pequeño/metabolismo , Transducción de Señal , Resultado del Tratamiento
16.
Oxid Med Cell Longev ; 2015: 454659, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25918581

RESUMEN

Oxidative stress as a result of elevated levels of reactive oxygen species (ROS) has been observed in almost all cancers, including leukaemia, where they contribute to disease development and progression. However, cancer cells also express increased levels of antioxidant proteins which detoxify ROS. This includes glutathione, the major antioxidant in human cells, which has recently been identified to have dysregulated metabolism in human leukaemia. This suggests that critical balance of intracellular ROS levels is required for cancer cell function, growth, and survival. Nuclear factor (erythroid-derived 2)-like 2 (NRF2) transcription factor plays a dual role in cancer. Primarily, NRF2 is a transcription factor functioning to protect nonmalignant cells from malignant transformation and oxidative stress through transcriptional activation of detoxifying and antioxidant enzymes. However, once malignant transformation has occurred within a cell, NRF2 functions to protect the tumour from oxidative stress and chemotherapy-induced cytotoxicity. Moreover, inhibition of the NRF2 oxidative stress pathway in leukaemia cells renders them more sensitive to cytotoxic chemotherapy. Our improved understanding of NRF2 biology in human leukaemia may permit mechanisms by which we could potentially improve future cancer therapies. This review highlights the mechanisms by which leukaemic cells exploit the NRF2/ROS response to promote their growth and survival.


Asunto(s)
Leucemia/patología , Factor 2 Relacionado con NF-E2/metabolismo , Estrés Oxidativo , Antioxidantes/metabolismo , Supervivencia Celular , Daño del ADN , Glutatión/metabolismo , Humanos , Leucemia/metabolismo , Factor 2 Relacionado con NF-E2/genética , Especies Reactivas de Oxígeno/metabolismo
17.
Sci Rep ; 5: 12949, 2015 Aug 21.
Artículo en Inglés | MEDLINE | ID: mdl-26292723

RESUMEN

Approximately 20% of patients with acute myeloid leukaemia (AML) have a mutation in FMS-like-tyrosine-kinase-3 (FLT3). FLT3 is a trans-membrane receptor with a tyrosine kinase domain which, when activated, initiates a cascade of phosphorylated proteins including the SRC family of kinases. Recently our group and others have shown that pharmacologic inhibition and genetic knockdown of Bruton's tyrosine kinase (BTK) blocks AML blast proliferation, leukaemic cell adhesion to bone marrow stromal cells as well as migration of AML blasts. The anti-proliferative effects of BTK inhibition in human AML are mediated via inhibition of downstream NF-κB pro-survival signalling however the upstream drivers of BTK activation in human AML have yet to be fully characterised. Here we place the FLT3-ITD upstream of BTK in AML and show that the BTK inhibitor ibrutinib inhibits the survival and proliferation of FLT3-ITD primary AML blasts and AML cell lines. Furthermore ibrutinib inhibits the activation of downstream kinases including MAPK, AKT and STAT5. In addition we show that BTK RNAi inhibits proliferation of FLT3-ITD AML cells. Finally we report that ibrutinib reverses the cyto-protective role of BMSC on FLT3-ITD AML survival. These results argue for the evaluation of ibrutinib in patients with FLT3-ITD mutated AML.


Asunto(s)
Leucemia Mieloide Aguda/metabolismo , Proteínas Tirosina Quinasas/metabolismo , Tirosina Quinasa 3 Similar a fms/química , Tirosina Quinasa 3 Similar a fms/metabolismo , Adenina/análogos & derivados , Agammaglobulinemia Tirosina Quinasa , Apoptosis/efectos de los fármacos , Crisis Blástica/patología , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Daunorrubicina/farmacología , Humanos , Mutación/genética , Piperidinas , Estructura Terciaria de Proteína , Pirazoles/farmacología , Pirimidinas/farmacología , ARN Interferente Pequeño/metabolismo , Transducción de Señal/efectos de los fármacos
18.
Lancet Haematol ; 2(5): e204-11, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-26688095

RESUMEN

BACKGROUND: Roughly 80% of patients with acute myeloid leukaemia have high activity of Bruton's tyrosine-kinase (BTK) in their blast cells compared with normal haemopoietic cells, rendering the cells sensitive to the oral BTK inhibitor ibrutinib in vitro. We aimed to develop the biological understanding of the BTK pathway in acute myeloid leukaemia to identify clinically relevant diagnostic information that might define a subset of patients that should respond to ibrutinib treatment. METHODS: We obtained acute myeloid leukaemia blast cells from unselected patients attending our UK hospital between Feb 19, 2010, and Jan 20, 2014. We isolated primary acute myeloid leukaemia blast cells from heparinised blood and human peripheral blood mononuclear cells to establish the activity of BTK in response to CD117 activation. Furthermore, we investigated the effects of ibrutinib on CD117-induced BTK activation, downstream signalling, adhesion to primary bone-marrow mesenchymal stromal cells, and proliferation of primary acute myeloid leukaemia blast cells. We used the Mann-Whitney U test to compare results between groups. FINDINGS: We obtained acute myeloid leukaemia blast cells from 29 patients. Ibrutinib significantly inhibited CD117-mediated proliferation of primary acute myeloid leukaemia blast cells (p=0·028). CD117 activation increased BTK activity by inducing phosphorylated BTK in patients with CD117-positive acute myeloid leukaemia. Furthermore, ibrutinib inhibited CD117-induced activity of BTK and downstream kinases at a concentration of 100 nM or more. CD117-mediated adhesion of CD117-expressing blast cells to bone-marrow stromal cells was significantly inhibited by Ibrutinib at 500 nM (p=0·028) INTERPRETATION: As first-in-man clinical trials of ibrutinib in patients with acute myeloid leukaemia commence, the data suggest not all patients will respond. Our findings show that BTK has specific pro-tumoural biological actions downstream of surface CD117 activation, which are inhibited by ibrutinib. Accordingly, we propose that patients with acute myeloid leukaemia whose blast cells express CD117 should be considered for forthcoming clinical trials of ibrutinib. FUNDING: Worldwide Cancer Research, The Big C, UK National Institutes for Health Research, the Humane Research Trust, the Department of Higher Education and Research of the Libyan Government, and Norwich Research Park.


Asunto(s)
Leucemia Mieloide Aguda/tratamiento farmacológico , Proteínas Tirosina Quinasas/antagonistas & inhibidores , Pirazoles/uso terapéutico , Pirimidinas/uso terapéutico , Transducción de Señal , Adenina/análogos & derivados , Adulto , Agammaglobulinemia Tirosina Quinasa , Anciano , Anciano de 80 o más Años , Linfocitos B/citología , Femenino , Humanos , Leucocitos Mononucleares/citología , Masculino , Persona de Mediana Edad , Piperidinas , Proteínas Proto-Oncogénicas c-kit
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA