Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 2.109
Filtrar
1.
Naunyn Schmiedebergs Arch Pharmacol ; 397(4): 2389-2400, 2024 04.
Artículo en Inglés | MEDLINE | ID: mdl-37837474

RESUMEN

BACKGROUND: 7,12-Dimethylbenzanthracene (DMBA) is a member of the polycyclic aromatic hydrocarbon family. It is a member of the polycyclic aromatic hydrocarbon family. It is a mutagenic, carcinogenic, and immunosuppressor agent. Cannabidiol (CBD) is a phytocannabinoid. It has anticonvulsant, anti-inflammatory, anti-anxiety, antioxidant, and anti-cancer properties. The purpose of this study was to investigate the possible protective and therapeutic benefits of CBD oil in DMBA-induced leukemia in rats. METHOD: Experimental animals were divided into six groups of five rats each. Group 1 (normal control) included healthy rats. Group 2 included normal rats that received olive oil. Group 3 included normal rats that received CBD. Group 4 included the DMBA-induced leukemic group. Group 5 (prophylactic group) included rats that received CBD as a prophylaxis before IV injection with DMBA. Group 6 (treated group) included DMBA-induced leukemic rats that received CBD as treatment. Liver functions (total, direct and indirect bilirubin, alkaline phosphatase (ALP), alanine transaminase (ALT), aspartate aminotransferase (AST), albumin, globulin, and albumin globulin ratio) were measured. Superoxide dismutase (SOD) and catalase (CAT) were also measured. Total RNA extraction followed by-real time qRT-PCR gene expression of LC3-II, Beclin, mTOR, and P62 was performed. Histopathological examination of liver and spleen tissues was performed. RESULTS: Administration of CBD in groups 5 and 6 resulted in a significant improvement of the levels of liver functions compared to the leukemic untreated rats. Also, the levels of catalase and SOD significantly increased after treatment with CBD compared to the leukemic group. After treatment with CBD in groups 5 and 6, there were downregulations in the expression of all studied genes compared to leukemic untreated rats. Treatment with CBD was more statistically effective than prophylactic use. CONCLUSION: Administration of CBD resulted in a significant improvement in the biochemical, antioxidant status, morphological, and molecular measures in DMBA-induced leukemia in adult male rats. The therapeutic use was more effective than the prophylactic one.


Asunto(s)
Cannabidiol , Globulinas , Leucemia Experimental , Ratas , Masculino , Animales , Antioxidantes/farmacología , Catalasa/metabolismo , 9,10-Dimetil-1,2-benzantraceno/metabolismo , 9,10-Dimetil-1,2-benzantraceno/farmacología , Leucemia Experimental/tratamiento farmacológico , Leucemia Experimental/metabolismo , Leucemia Experimental/patología , Hígado , Globulinas/metabolismo , Globulinas/farmacología , Superóxido Dismutasa/metabolismo , Albúminas/metabolismo
2.
J Mol Biol ; 433(18): 167111, 2021 09 03.
Artículo en Inglés | MEDLINE | ID: mdl-34153286

RESUMEN

5-aza-cytidine (5-aza-C) has been shown to be a potent human immunodeficiency virus type 1 (HIV-1) mutagen that induces G-to-C hypermutagenesis by incorporation of the reduced form (i.e., 5-aza-dC, 5-aza-dCTP). Evidence to date suggests that this lethal mutagenesis is the primary antiretroviral mechanism for 5-aza-C. To investigate the breadth of application of 5-aza-C as an antiretroviral mutagen, we have conducted a comparative, parallel analysis of the antiviral mechanism of 5-aza-C between HIV-1 and gammaretroviruses - i.e., murine leukemia virus (MuLV) and feline leukemia virus (FeLV). Intriguingly, in contrast to the hallmark G-to-C hypermutagenesis observed with HIV-1, MuLV and FeLV did not reveal the presence of a significant increase in mutational burden, particularly that of G-to-C transversion mutations. The effect of 5-aza-dCTP on DNA synthesis revealed that while HIV-1 RT was not inhibited by 5-aza-dCTP even at 100 µM, 5-aza-dCTP was incorporated and significantly inhibited MuLV RT, generating pause sites and reducing the fully extended product. 5-aza-dCTP was found to be incorporated into DNA by MuLV RT or HIV-1 RT, but only acted as a non-obligate chain terminator for MuLV RT. This biochemical data provides an independent line of experimental evidence in support of the conclusion that HIV-1 and MuLV have distinct primary mechanisms of antiretroviral action with 5-aza-C. Taken together, our data provides striking evidence that an antiretroviral mutagen can have strong potency via distinct mechanisms of action among closely related viruses, unlinking antiviral activity from antiviral mechanism of action.


Asunto(s)
Antivirales/farmacología , Azacitidina/análogos & derivados , Citidina Trifosfato/análogos & derivados , Infecciones por VIH/tratamiento farmacológico , Leucemia Experimental/tratamiento farmacológico , Mutación/efectos de los fármacos , Infecciones por Retroviridae/tratamiento farmacológico , Infecciones Tumorales por Virus/tratamiento farmacológico , Animales , Azacitidina/farmacología , Gatos , Citidina Trifosfato/farmacología , VIH/efectos de los fármacos , Infecciones por VIH/virología , Humanos , Virus de la Leucemia Felina/efectos de los fármacos , Virus de la Leucemia Murina/efectos de los fármacos , Leucemia Experimental/virología , Ratones , Mutagénesis , Mutágenos , Infecciones por Retroviridae/virología , Infecciones Tumorales por Virus/virología , Replicación Viral
3.
Sci Rep ; 11(1): 9103, 2021 04 27.
Artículo en Inglés | MEDLINE | ID: mdl-33907248

RESUMEN

(-)-Epigallocatechin-3-gallate (EGCG), the major active polyphenol extracted from green tea, has been shown to induce apoptosis and inhibit cell proliferation, cell invasion, angiogenesis and metastasis. Herein, we evaluated the in vivo effects of EGCG in acute myeloid leukaemia (AML) using an acute promyelocytic leukaemia (APL) experimental model (PML/RARα). Haematological analysis revealed that EGCG treatment reversed leucocytosis, anaemia and thrombocytopenia, and prolonged survival of PML/RARα mice. Notably, EGCG reduced leukaemia immature cells and promyelocytes in the bone marrow while increasing mature myeloid cells, possibly due to apoptosis increase and cell differentiation. The reduction of promyelocytes and neutrophils/monocytes increase detected in the peripheral blood, in addition to the increased percentage of bone marrow cells with aggregated promyelocytic leukaemia (PML) bodies staining and decreased expression of PML-RAR oncoprotein corroborates our results. In addition, EGCG increased expression of neutrophil differentiation markers such as CD11b, CD14, CD15 and CD66 in NB4 cells; and the combination of all-trans retinoic acid (ATRA) plus EGCG yield higher increase the expression of CD15 marker. These findings could be explained by a decrease of peptidyl-prolyl isomerase NIMA-interacting 1 (PIN1) expression and reactive oxygen species (ROS) increase. EGCG also decreased expression of substrate oncoproteins for PIN1 (including cyclin D1, NF-κB p65, c-MYC, and AKT) and 67 kDa laminin receptor (67LR) in the bone marrow cells. Moreover, EGCG showed inhibition of ROS production in NB4 cells in the presence of N-acetyl-L-cysteine (NAC), as well as a partial blockage of neutrophil differentiation and apoptosis, indicating that EGCG-activities involve/or are in response of oxidative stress. Furthermore, apoptosis of spleen cells was supported by increasing expression of BAD and BAX, parallel to BCL-2 and c-MYC decrease. The reduction of spleen weights of PML/RARα mice, as well as apoptosis induced by EGCG in NB4 cells in a dose-dependent manner confirms this assumption. Our results support further evaluation of EGCG in clinical trials for AML, since EGCG could represent a promising option for AML patient ineligible for current mainstay treatments.


Asunto(s)
Antineoplásicos Fitogénicos/farmacología , Catequina/análogos & derivados , Leucemia Promielocítica Aguda/tratamiento farmacológico , Peptidilprolil Isomerasa de Interacción con NIMA/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Animales , Apoptosis/efectos de los fármacos , Catequina/farmacología , Diferenciación Celular/efectos de los fármacos , Humanos , Leucemia Experimental/tratamiento farmacológico , Leucemia Experimental/mortalidad , Leucemia Experimental/patología , Leucemia Promielocítica Aguda/metabolismo , Leucemia Promielocítica Aguda/patología , Ratones Transgénicos , Receptor alfa de Ácido Retinoico/genética , Bazo/efectos de los fármacos , Bazo/patología , Proteína X Asociada a bcl-2/metabolismo , Proteína Letal Asociada a bcl/metabolismo
4.
Leukemia ; 35(8): 2274-2284, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-33526858

RESUMEN

Despite significant progress over the last few decades in the treatment of acute myeloid leukemia (AML), there still remains a major unmet medical need for this disease. Immunotherapy approaches for redirecting pan CD3+ T cells to target leukemia blasts have shown limited efficacy in clinical trials and often accompanied with severe toxicity in AML patients. We designed an alternative engager molecule (Anti-TRGV9/anti-CD123), a bispecific antibody that can simultaneously bind to the Vγ9 chain of the Vγ9Vδ2+ γδ T cell receptor and to AML target antigen, CD123, to selectively recruit Vγ9+ γδ T cells rather than pan T cells to target AML blasts. Our results suggest that prototypic bispecific antibodies (a) selectively activate Vγ9+ γδ T cells as judged by CD69 and CD25 surface expression, and intracellular Granzyme B expression, (b) selectively recruit Vγ9+ γδ T cells into cell-cell conjugate formation of γδ T cells with tumor cells indicating selective and effective engagement of effector and target tumor cells, and (c) mediate γδ T cell cytotoxicity (in vitro and in vivo) against tumor antigen-expressing cells. Collectively, these findings suggest that selectively redirecting Vγ9+ γδ T cells to target AML blasts has a potential for immunotherapy for AML patients and favors further exploration of this concept.


Asunto(s)
Anticuerpos Biespecíficos/inmunología , Antineoplásicos Inmunológicos/farmacología , Inmunoterapia/métodos , Leucemia Experimental/tratamiento farmacológico , Leucemia Mieloide Aguda/tratamiento farmacológico , Receptores de Antígenos de Linfocitos T gamma-delta/inmunología , Animales , Citotoxicidad Inmunológica , Humanos , Leucemia Experimental/inmunología , Leucemia Experimental/patología , Leucemia Mieloide Aguda/inmunología , Leucemia Mieloide Aguda/patología , Ratones , Ratones Endogámicos NOD , Ratones SCID , Células Tumorales Cultivadas , Ensayos Antitumor por Modelo de Xenoinjerto
5.
Anticancer Drugs ; 32(1): 61-65, 2021 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-32701559

RESUMEN

The pharmacologically active metabolite of cyclophosphamide is aldophosphamide. With cysteine, aldophosphamide forms stable aldophosphamide-thiazolidine which under physiological pH and temperature conditions hydrolyzes to aldophosphamide and cysteine. Aldophosphamide-thiazolidine was synthesized and tested for its ability as a cytostatic. The LD50 after a single intraperitoneal injection in mice was determined to be 2162 mg/kg, but after intravenous bolus administration of 500 mg/kg or in chronic toxicity tests with daily intraperitoneal injections, neurological side effects were observed. Antitumor activity was determined in therapy experiments in CD2F1 mice bearing subcutaneously transplanted P388 mouse leukemia cells. Administration of 100 mg/kg (less than 5% LD50) days 1-5 after tumor transplantation yielded an ILS of 100%. Organ distribution studies showed that aldophosphamide-thiazolidine is evenly distributed in all tissues examined, including brain tissue. The possibilities to increase the antitumor activity of aldophosphamide-thiazolidine by modulating the alkylating function are discussed.


Asunto(s)
Barrera Hematoencefálica/metabolismo , Citostáticos/farmacología , Leucemia Experimental/tratamiento farmacológico , Compuestos de Mostaza Nitrogenada/farmacología , Tiazolidinas/farmacología , Animales , Apoptosis , Barrera Hematoencefálica/efectos de los fármacos , Proliferación Celular , Citostáticos/farmacocinética , Femenino , Leucemia Experimental/metabolismo , Leucemia Experimental/patología , Ratones , Compuestos de Mostaza Nitrogenada/farmacocinética , Tiazolidinas/farmacocinética , Distribución Tisular , Células Tumorales Cultivadas
6.
J Ethnopharmacol ; 267: 113519, 2021 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-33137432

RESUMEN

ETHNOPHARMACOLOGICAL RELEVANCE: Astragalus glycyphyllos L. has been extensively used in Bulgarian folk medicine as an antihypertensive, diuretic, anti-inflammatory, anti-tumour, in cases of cardiac insufficiency, renal inflammation, calculosis, etc. AIM OF THE STUDY: To evaluate the possible in vitro/in vivo anti-proliferative/anti-tumour activity of a purified saponins' mixture (PSM) obtained from the plant. MATERIALS AND METHODS: Viability and proliferative activity of the Graffi myeloid tumour cells was assessed by MTT test. The morphological alterations were visualized and analysed by fluorescent microscopy after intravital double staining. An in vivo model of Graffi tumour bearing hamsters was used to examine the influence of PSM on transplantability, tumour growth, survival and mortality as well as to observe pathomorphological changes. RESULTS: Graffi tumour cells were sensitive to purified saponins' mixture after 24 and 48 h treatment. The treatment induced a statistically significant decrease of the viability/proliferation of the Graffi tumour cells. These effects were concentration- and time-dependent. Fluorescent microscopy studies showed that these antiproliferative effects were connected to the induction of apoptosis. The in vivo study showed the presence of a stromal component, single mononuclear cells in the stroma. Multiple incorrect mitotic figures were observed in the tumour tissue from the control group. Well-formed stroma with accumulation of mononuclear cells and mitotic cells were found in the group, treated with PSM. The tumour weight was decreased in the group, treated with PMS. CONCLUSION: The results indicate that PSM exhibited in vitro/in vivo antiproliferative/anti-tumour effects.


Asunto(s)
Antineoplásicos Fitogénicos/farmacología , Planta del Astrágalo , Proliferación Celular/efectos de los fármacos , Leucemia Experimental/tratamiento farmacológico , Extractos Vegetales/farmacología , Saponinas/farmacología , Animales , Antineoplásicos Fitogénicos/aislamiento & purificación , Apoptosis/efectos de los fármacos , Planta del Astrágalo/química , Cricetinae , Femenino , Virus de la Leucemia Murina/patogenicidad , Leucemia Experimental/patología , Leucemia Experimental/virología , Masculino , Mesocricetus , Extractos Vegetales/aislamiento & purificación , Cultivo Primario de Células , Infecciones por Retroviridae/virología , Saponinas/aislamiento & purificación , Carga Tumoral/efectos de los fármacos , Células Tumorales Cultivadas , Infecciones Tumorales por Virus/virología
7.
Biochim Biophys Acta Mol Basis Dis ; 1866(12): 165922, 2020 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-32800945

RESUMEN

Excessive production of immunoglobulins (Ig) causes endoplasmic reticulum (ER) stress and triggers the unfolded protein response (UPR). Hypergammaglobulinemia and lymphadenopathy are hallmarks of murine AIDS that develops in mice infected with the LP-BM5 murine leukemia retrovirus complex. In these mice, Th2 polarization and aberrant humoral response have been previously correlated to altered intracellular redox homeostasis. Our goal was to understand the role of the cell's redox state in Ig secretion and plasma cell (PC) maturation. To this aim, LP-BM5-infected mice were treated with I-152, an N-acetyl-cysteine and cysteamine supplier. Intraperitoneal I-152 administration (30 µmol/mouse three times a week for 9 weeks) decreased plasma IgG and increased IgG/Syndecan 1 ratio in the lymph nodes where IgG were in part accumulated within the ER. PC containing cytoplasmic inclusions filled with IgG were present in all animals, with fewer mature PC in those treated with I-152. Infection induced up-regulation of signaling molecules involved in the UPR, i.e. CHAC1, BiP, sXBP-1 and PDI, that were generally unaffected by I-152 treatment except for PDI and sXBP-1, which have a key role in protein folding and PC maturation, respectively. Our data suggest that one of the mechanisms through which I-152 can limit hypergammaglobulinemia in LP-BM5-infected mice is by influencing IgG folding/assembly as well as secretion and affecting PC maturation.


Asunto(s)
Acetilcisteína/análogos & derivados , Antivirales/farmacología , Cisteamina/análogos & derivados , Inmunoglobulinas/metabolismo , Células Plasmáticas/efectos de los fármacos , Infecciones por Retroviridae/tratamiento farmacológico , Infecciones Tumorales por Virus/tratamiento farmacológico , Respuesta de Proteína Desplegada/efectos de los fármacos , Acetilcisteína/administración & dosificación , Acetilcisteína/farmacología , Animales , Antivirales/administración & dosificación , Cisteamina/administración & dosificación , Cisteamina/farmacología , Modelos Animales de Enfermedad , Femenino , Inmunoglobulinas/sangre , Inyecciones Intraperitoneales , Leucemia Experimental/tratamiento farmacológico , Leucemia Experimental/metabolismo , Leucemia Experimental/virología , Ratones , Ratones Endogámicos C57BL , Células Plasmáticas/metabolismo , Células Plasmáticas/virología , Desplegamiento Proteico/efectos de los fármacos , Infecciones por Retroviridae/metabolismo , Infecciones por Retroviridae/virología , Infecciones Tumorales por Virus/metabolismo , Infecciones Tumorales por Virus/virología
8.
FASEB J ; 34(7): 8843-8857, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-32433826

RESUMEN

Drug resistance is a common obstacle in leukemia treatment and failing to eradicate leukemia stem cells is the main cause of leukemia relapse. Previous studies have demonstrated that telomerase activity is associated with deregulated self-renewal of leukemia stem cells (LSCs). Here, we identified a novel compound IX, an imatinib derivative with a replacement fragment of a telomerase inhibitor, which can effectively eradicate LSCs but had no influence on normal hematopoietic stem cells (HSCs) survival. We showed that compound IX can decrease the viability of drug-resistant K562/G cells and blast crisis CML primary patient cells. Besides, IX can affect LSC survival, inhibit the colony-forming ability, and reduce LSC frequency. In vivo results showed that IX can relieve the tumor burden in patient-derived xenograft (PDX) model and prolong the lifespan. We observed that compound IX can not only decrease telomerase activity, but also affect the alternative lengthening of telomeres. In addition, IX can inhibit both the canonical and non-canonical Wnt pathways. Our data suggested this novel compound IX as a promising candidate for drug-resistant leukemia therapy.


Asunto(s)
Carcinogénesis/efectos de los fármacos , Resistencia a Antineoplásicos , Leucemia Experimental/tratamiento farmacológico , Leucemia Mielógena Crónica BCR-ABL Positiva/tratamiento farmacológico , Leucemia Mieloide Aguda/tratamiento farmacológico , Bibliotecas de Moléculas Pequeñas/farmacología , Telómero/efectos de los fármacos , Apoptosis , Carcinogénesis/metabolismo , Carcinogénesis/patología , Ciclo Celular , Movimiento Celular , Proliferación Celular , Humanos , Leucemia Experimental/metabolismo , Leucemia Experimental/patología , Leucemia Mielógena Crónica BCR-ABL Positiva/metabolismo , Leucemia Mielógena Crónica BCR-ABL Positiva/patología , Leucemia Mieloide Aguda/metabolismo , Leucemia Mieloide Aguda/patología , Células Madre Neoplásicas/efectos de los fármacos , Células Madre Neoplásicas/metabolismo , Células Madre Neoplásicas/patología , Preparaciones Farmacéuticas/administración & dosificación , Telómero/metabolismo , Células Tumorales Cultivadas
9.
Nat Commun ; 11(1): 740, 2020 02 06.
Artículo en Inglés | MEDLINE | ID: mdl-32029739

RESUMEN

Primary and acquired drug resistance imposes a major threat to achieving optimized clinical outcomes during cancer treatment. Aberrant changes in epigenetic modifications are closely involved in drug resistance of tumor cells. Using BET inhibitor (BETi) resistant leukemia cells as a model system, we demonstrated herein that genome-wide enhancer remodeling played a pivotal role in driving therapeutic resistance via compensational re-expression of pro-survival genes. Capitalizing on the CRISPR interference technology, we identified the second intron of IncRNA, PVT1, as a unique bona fide gained enhancer that restored MYC transcription independent of BRD4 recruitment in leukemia. A combined BETi and CDK7 inhibitor treatment abolished MYC transcription by impeding RNAPII loading without affecting PVT1-mediated chromatin looping at the MYC locus in BETi-resistant leukemia cells. Together, our findings have established the feasibility of targeting enhancer plasticity to overcome drug resistance associated with epigenetic therapies.


Asunto(s)
Leucemia Experimental/tratamiento farmacológico , Leucemia Experimental/genética , Proteínas Nucleares/antagonistas & inhibidores , Factores de Transcripción/antagonistas & inhibidores , Animales , Proteínas de Ciclo Celular/antagonistas & inhibidores , Línea Celular Tumoral , Quinasas Ciclina-Dependientes/antagonistas & inhibidores , Resistencia a Antineoplásicos/genética , Sinergismo Farmacológico , Elementos de Facilitación Genéticos , Femenino , Genes myc/efectos de los fármacos , Compuestos Heterocíclicos de 4 o más Anillos/administración & dosificación , Humanos , Células Jurkat , Células K562 , Leucemia Experimental/metabolismo , Ratones , Modelos Genéticos , Fenilendiaminas/administración & dosificación , Pirimidinas/administración & dosificación , ARN Polimerasa II/metabolismo , ARN Largo no Codificante/genética , Quinasa Activadora de Quinasas Ciclina-Dependientes
10.
Eur J Med Chem ; 189: 112023, 2020 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-31978781

RESUMEN

Disruptor of Telomeric Silencing 1-Like (DOT1L), the sole histone H3 lysine 79 (H3K79) methyltransferase, is required for leukemogenic transformation in a subset of leukemias bearing chromosomal translocations of the Mixed Lineage Leukemia (MLL) gene, as well as other cancers. Thus, DOT1L is an attractive therapeutic target and discovery of small molecule inhibitors remain of high interest. Herein, we are presenting screening results for a unique focused library of 1200 nucleoside analogs originally produced under the aegis of the NIH Pilot Scale Library Program. The complete nucleoside set was screened virtually against DOT1L, resulting in 210 putative hits. In vitro screening of the virtual hits resulted in validation of 11 compounds as DOT1L inhibitors clustered into two distinct chemical classes, adenosine-based inhibitors and a new chemotype that lacks adenosine. Based on the developed DOT1L ligand binding model, a structure-based design strategy was applied and a second-generation of non-nucleoside DOT1L inhibitors was developed. Newly synthesized compound 25 was the most potent DOT1L inhibitor in the new series with an IC50 of 1.0 µM, showing 40-fold improvement in comparison with hit 9 and exhibiting reasonable on target effects in a DOT1L dependent murine cell line. These compounds represent novel chemical probes with a unique non-nucleoside scaffold that bind and compete with the SAM binding site of DOT1L, thus providing foundation for further medicinal chemistry efforts to develop more potent compounds.


Asunto(s)
Médula Ósea/efectos de los fármacos , Proliferación Celular , Inhibidores Enzimáticos/farmacología , Ensayos Analíticos de Alto Rendimiento/métodos , N-Metiltransferasa de Histona-Lisina/antagonistas & inhibidores , Leucemia Experimental/tratamiento farmacológico , Nucleósidos/farmacología , Triazoles/farmacología , Animales , Médula Ósea/enzimología , Simulación por Computador , Inhibidores Enzimáticos/química , Leucemia Experimental/enzimología , Ratones , Nucleósidos/química , Relación Estructura-Actividad , Triazoles/química
11.
Environ Toxicol ; 35(4): 457-467, 2020 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-31793136

RESUMEN

Cardamonin, a monomeric alkaloid, is isolated from Alpinia conchigera Griff and other natural plants. Recently, it has been focused on its anticancer activities, and no information showing its immune effects on leukemia mice was reported. In this study, we investigated the immune effects of cardamonin on WEHI-3 cell-generated leukemia mice. Forty BALB/c mice were randomly divided into four groups: Group I mice were normal animals and groups II-IV were leukemia. Group II mice, as a positive control, were administered with normal diet, and group III and IV mice were treated with 1 and 5 mg/kg of cardamonin, respectively, by intraperitoneal injection every 2 days for 14 days. The population of white blood cells, macrophage phagocytosis, and the proliferations of T and B cells were analyzed by flow cytometry. Another forty mice were also separated randomly into four groups for the determination of survival rate. Results showed that cardamonin did not affect body weight. Cardamonin decreased CD3, CD11b, and Mac-3 cell populations but increased CD19 number. Cardamonin enhanced phagocytic abilities of macrophages from the peripheral blood mononuclear cells of leukemia mice. Furthermore, cardamonin at 1 mg/kg treatment improved the survival rate of leukemia mice in vivo. Therefore, cardamonin could be applied for a leukemia therapeutic reagent at a defined dose.


Asunto(s)
Antineoplásicos Fitogénicos/farmacología , Chalconas/farmacología , Leucemia Experimental/tratamiento farmacológico , Leucemia Experimental/inmunología , Leucocitos Mononucleares/efectos de los fármacos , Animales , Antígenos CD19/sangre , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Leucocitos Mononucleares/inmunología , Macrófagos/efectos de los fármacos , Macrófagos/inmunología , Masculino , Ratones , Ratones Endogámicos BALB C , Fagocitosis/efectos de los fármacos , Fagocitosis/inmunología , Tasa de Supervivencia
12.
Life Sci ; 242: 117228, 2020 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-31881227

RESUMEN

AIMS: Berberine (BBR) is reported to induce apoptosis and inhibit migration of leukemic cells, but the underlying pharmacological mechanisms have not been fully revealed. This study aims to investigate the possible mechanisms from the perspective of autophagy. MAIN METHODS: P-53-null leukemic cell lines Jurkat and U937 were used for the in vitro study. MDC staining was used for observation of autophagy in leukemic cells, and Western blot analysis was for determination of the expression levels of autophagy-associated proteins. Apoptosis of the leukemic cells was detected by flow cytometry. Cellular location of MDM2 was observed with immunofluorescence staining. Ubiquitination of MDM2 was assessed by immunoprecipitation. Male 6-8-week-old NOD/SCID mice were used for evaluating the effect of BBR on chemotherapy sensitivity in vivo. KEY FINDINGS: BBR induced autophagy in p53-null leukemic cells, which was inhibited by autophagy inhibitors 3-methyladenine. 3-methyladenine also inhibited BBR-induced apoptosis in leukemic cells. In addition, BBR not only decreased MDM2 mRNA expression, but also enhanced MDM2 self-ubiquitination in leukemic cells. Forced overexpression of MDM2 reversed the effect of BBR on autophagy and apoptosis. Furthermore, BBR promoted doxorubicin-induced autophagy and cell death in the leukemic cells and overexpression of MDM2 suppressed these effects. In vivo, BBR combined with doxorubicin achieved better therapeutic effect than doxorubicin alone. SIGNIFICANCE: MDM2 inhibits autophagy and apoptosis in leukemic cells in a p53-independent manner. BBR induces autophagy in p53-null leukemic cells through downregulating MDM2 expression at both transcriptional and post-transcriptional levels, which may contribute to the anti-cancer effect of BBR in leukemia.


Asunto(s)
Antineoplásicos Fitogénicos/farmacología , Apoptosis/efectos de los fármacos , Autofagia/efectos de los fármacos , Berberina/farmacología , Células Jurkat/efectos de los fármacos , Leucemia Experimental/tratamiento farmacológico , Proteínas Proto-Oncogénicas c-mdm2/metabolismo , Proteína p53 Supresora de Tumor/metabolismo , Células U937/efectos de los fármacos , Animales , Western Blotting , Citometría de Flujo , Técnica del Anticuerpo Fluorescente , Humanos , Células Jurkat/metabolismo , Leucemia Experimental/metabolismo , Masculino , Ratones Endogámicos NOD , Ratones SCID , Trasplante de Neoplasias , Reacción en Cadena en Tiempo Real de la Polimerasa , Células U937/metabolismo , Ubiquitinación
13.
Nat Commun ; 10(1): 2691, 2019 06 19.
Artículo en Inglés | MEDLINE | ID: mdl-31217428

RESUMEN

The MUSASHI (MSI) family of RNA binding proteins (MSI1 and MSI2) contribute to a wide spectrum of cancers including acute myeloid leukemia. We find that the small molecule Ro 08-2750 (Ro) binds directly and selectively to MSI2 and competes for its RNA binding in biochemical assays. Ro treatment in mouse and human myeloid leukemia cells results in an increase in differentiation and apoptosis, inhibition of known MSI-targets, and a shared global gene expression signature similar to shRNA depletion of MSI2. Ro demonstrates in vivo inhibition of c-MYC and reduces disease burden in a murine AML leukemia model. Thus, we identify a small molecule that targets MSI's oncogenic activity. Our study provides a framework for targeting RNA binding proteins in cancer.


Asunto(s)
Regulación Leucémica de la Expresión Génica/efectos de los fármacos , Leucemia Experimental/tratamiento farmacológico , Leucemia Mieloide Aguda/tratamiento farmacológico , Pteridinas/farmacología , Proteínas de Unión al ARN/antagonistas & inhibidores , Animales , Apoptosis/efectos de los fármacos , Flavinas , Perfilación de la Expresión Génica , Humanos , Leucemia Experimental/sangre , Leucemia Mieloide Aguda/sangre , Masculino , Ratones , Cultivo Primario de Células , Proteínas Proto-Oncogénicas c-myc/metabolismo , Pteridinas/uso terapéutico , ARN/metabolismo , Motivo de Reconocimiento de ARN/efectos de los fármacos , ARN Interferente Pequeño/metabolismo , Proteínas de Unión al ARN/genética , Proteínas de Unión al ARN/metabolismo , Transcriptoma/efectos de los fármacos , Células Tumorales Cultivadas
14.
J Assist Reprod Genet ; 36(9): 1793-1803, 2019 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-31250176

RESUMEN

PURPOSE: To determine whether pharmacological administration of recombinant human anti-Mullerian hormone (rAMH) protects the ovarian reserve and preserves fertility without interfering with anti-tumoural cytotoxic action of chemotherapy. METHODS: Intraperitoneal delivery of rAMH and ovarian post-receptor activity were assessed with immunohistochemistry and western blot. Differential follicle counts and reproductive outcomes were assessed after cyclophosphamide (Cy) administration, with/without concurrent administration of rAMH. Interference of rAMH with Cy chemotoxicity was assessed on a human breast cancer cell line and an in vivo mouse model of human leukaemia. RESULTS: rAMH reached the ovary after intraperitoneal injection and demonstrated post-receptor bioactivity. Cy administration in mice caused primordial follicle activation, as shown by a decrease in primordial follicle population accompanied by an increase in early growing follicles and granulosa cell proliferation. Co-administration of rAMH reduced follicle activation, thereby protecting the primordial follicle reserve, and improving long-term fertility and reproductive outcomes. rAMH co-administration did not interfere with the cytotoxic actions of Cy in vitro on breast cancer cell line or in vivo in a model of human leukaemia. CONCLUSION: This study demonstrates that rAMH is bioactive in the ovary for a limited time, and that pharmacological administration of rAMH during chemotherapy treatment reduces follicle activation and primordial follicle loss and significantly improves reproductive outcomes in a mouse model, and does not interfere with the therapeutic actions of the treatment. Further investigation is necessary to determine whether it has similar protective effects in the human ovary.


Asunto(s)
Hormona Antimülleriana/farmacología , Ciclofosfamida/farmacología , Preservación de la Fertilidad/métodos , Reserva Ovárica/efectos de los fármacos , Animales , Hormona Antimülleriana/genética , Antineoplásicos Alquilantes/efectos adversos , Antineoplásicos Alquilantes/farmacología , Línea Celular Tumoral , Ciclofosfamida/efectos adversos , Femenino , Humanos , Leucemia Experimental/tratamiento farmacológico , Masculino , Ratones Endogámicos BALB C , Ratones Endogámicos NOD , Folículo Ovárico/efectos de los fármacos , Folículo Ovárico/patología , Reserva Ovárica/fisiología , Embarazo , Índice de Embarazo , Proteínas Recombinantes/genética , Proteínas Recombinantes/farmacología
15.
Toxicol Appl Pharmacol ; 379: 114638, 2019 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-31254567

RESUMEN

Our previous study has demonstrated that 4-amino-2-trifluoromethyl-phenyl Retinate (ATPR) can induce human leukemia NB4 cells differentiation and G0/G1 phase arrest, but the underlying mechanism is still unclear. In this study, we used proteomics to screen differentially expressed protein profiles in NB4 cells before and after ATPR treatment in vitro. We analyzed the peptides digested from total cellular proteins by reverse phase LC-MS/MS and then performed label-free quantitative analysis. We found 27 significantly up-regulated proteins in the ATPR group compared to the control group. NCF1 was the most significantly changed protein. Immunoprecipitation and double immunofluorescent staining showed that EBP50 bind to NCF1. We further explored the potential molecular mechanism of EBP50/NCF1 complex in ATPR-induced differentiation and G0/G1 phase arrest. The results showed that ATPR remarkably reduced the expression of EBP50 in vivo and in vitro. Interestingly, the reduction of EBP50 contributed to ROS release by modulating the subcellular localization of NCF1. The reduction of EBP50 also contributed to G0/G1 phase arrest by inhibiting CyclinD1, CyclinA2 and CDK4, as well as promoting the differentiation of NB4 cells by increasing the expression of CD11b. Furthermore, we found that the overexpression of EBP50 restrained the effects of ATPR on differentiation and G0/G1 phase arrest in NB4 cells. These results suggest that ATPR-induced differentiation and G0/G1 phase arrest in acute promyelocytic leukemia (APL) by repressing EBP50/NCF1 complex to promote the production of ROS, and the results from in vivo experiments were consistent with those from in vitro studies. Therefore, our finding results suggest that EBP50 may be a new target for ATPR in the treatment of APL.


Asunto(s)
Fase G1/efectos de los fármacos , Leucemia Promielocítica Aguda/tratamiento farmacológico , NADPH Oxidasas/metabolismo , Fosfoproteínas/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Fase de Descanso del Ciclo Celular/efectos de los fármacos , Retinoides/uso terapéutico , Intercambiadores de Sodio-Hidrógeno/metabolismo , Animales , Western Blotting , Línea Celular Tumoral , Técnica del Anticuerpo Fluorescente , Humanos , Leucemia Experimental/tratamiento farmacológico , Leucemia Promielocítica Aguda/metabolismo , Ratones , Trasplante de Neoplasias , Reacción en Cadena en Tiempo Real de la Polimerasa , Retinoides/farmacología
16.
Proc Natl Acad Sci U S A ; 116(21): 10482-10487, 2019 05 21.
Artículo en Inglés | MEDLINE | ID: mdl-31068472

RESUMEN

A major obstacle to curing chronic myeloid leukemia (CML) is the intrinsic resistance of CML stem cells (CMLSCs) to the drug imatinib mesylate (IM). Prosurvival genes that are preferentially expressed in CMLSCs compared with normal hematopoietic stem cells (HSCs) represent potential therapeutic targets for selectively eradicating CMLSCs. However, the discovery of such preferentially expressed genes has been hampered by the inability to completely separate CMLSCs from HSCs, which display a very similar set of surface markers. To overcome this challenge, and to minimize confounding effects of individual differences in gene expression profiles, we performed single-cell RNA-seq on CMLSCs and HSCs that were isolated from the same patient and distinguished based on the presence or absence of BCR-ABL. Among genes preferentially expressed in CMLSCs is PIM2, which encodes a prosurvival serine-threonine kinase that phosphorylates and inhibits the proapoptotic protein BAD. We show that IM resistance of CMLSCs is due, at least in part, to maintenance of BAD phosphorylation by PIM2. We find that in CMLSCs, PIM2 expression is promoted by both a BCR-ABL-dependent (IM-sensitive) STAT5-mediated pathway and a BCR-ABL-independent (IM-resistant) STAT4-mediated pathway. Combined treatment with IM and a PIM inhibitor synergistically increases apoptosis of CMLSCs, suppresses colony formation, and significantly prolongs survival in a mouse CML model, with a negligible effect on HSCs. Our results reveal a therapeutically targetable mechanism of IM resistance in CMLSCs. The experimental approach that we describe can be generally applied to other malignancies that harbor oncogenic fusion proteins or other characteristic genetic markers.


Asunto(s)
Compuestos de Bifenilo/uso terapéutico , Resistencia a Antineoplásicos , Leucemia Mielógena Crónica BCR-ABL Positiva/tratamiento farmacológico , Células Madre Neoplásicas/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Tiazolidinas/uso terapéutico , Animales , Ensayos de Selección de Medicamentos Antitumorales , Proteínas de Fusión bcr-abl/metabolismo , Humanos , Mesilato de Imatinib , Leucemia Experimental/tratamiento farmacológico , Leucemia Mielógena Crónica BCR-ABL Positiva/metabolismo , Ratones , Terapia Molecular Dirigida , Fosforilación , Inhibidores de Proteínas Quinasas , Factores de Transcripción STAT/metabolismo , Proteína Letal Asociada a bcl/metabolismo
17.
Nat Commun ; 10(1): 2189, 2019 05 16.
Artículo en Inglés | MEDLINE | ID: mdl-31097698

RESUMEN

Improvement in survival has been achieved for children and adolescents with AML but is largely attributed to enhanced supportive care as opposed to the development of better treatment regimens. High risk subtypes continue to have poor outcomes with event free survival rates <40% despite the use of high intensity chemotherapy in combination with hematopoietic stem cell transplant. Here we combine high-throughput screening, intracellular accumulation assays, and in vivo efficacy studies to identify therapeutic strategies for pediatric AML. We report therapeutics not currently used to treat AML, gemcitabine and cabazitaxel, have broad anti-leukemic activity across subtypes and are more effective relative to the AML standard of care, cytarabine, both in vitro and in vivo. JAK inhibitors are selective for acute megakaryoblastic leukemia and significantly prolong survival in multiple preclinical models. Our approach provides advances in the development of treatment strategies for pediatric AML.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Desoxicitidina/análogos & derivados , Inhibidores de las Cinasas Janus/farmacología , Leucemia Experimental/tratamiento farmacológico , Leucemia Mieloide Aguda/tratamiento farmacológico , Adulto , Animales , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Médula Ósea/patología , Médula Ósea/efectos de la radiación , Trasplante de Médula Ósea , Línea Celular Tumoral , Niño , Preescolar , Citarabina/farmacología , Citarabina/uso terapéutico , Desoxicitidina/farmacología , Desoxicitidina/uso terapéutico , Supervivencia sin Enfermedad , Femenino , Ensayos Analíticos de Alto Rendimiento/métodos , Humanos , Lactante , Inhibidores de las Cinasas Janus/uso terapéutico , Leucemia Experimental/etiología , Leucemia Experimental/mortalidad , Leucemia Experimental/patología , Leucemia Mieloide Aguda/mortalidad , Leucemia Mieloide Aguda/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Taxoides/farmacología , Taxoides/uso terapéutico , Irradiación Corporal Total/efectos adversos , Ensayos Antitumor por Modelo de Xenoinjerto , Adulto Joven , Gemcitabina
18.
Cancer ; 125(16): 2818-2828, 2019 08 15.
Artículo en Inglés | MEDLINE | ID: mdl-31034603

RESUMEN

BACKGROUND: Tyrosine kinase inhibitors (TKIs) are the standard treatment for chronic myeloid leukemia (CML). Despite their clinical success, TKIs are faced with challenges such as treatment resistance, which may be driven by kinase domain mutations, and frequent disease relapse upon the cessation of treatment. The combination of arsenic trioxide (ATO) and interferon-α (IFN) was previously demonstrated to inhibit proliferation and induce apoptosis in CML cell lines, prolong the survival of primary wild-type CML mice, and dramatically decrease the activity of leukemia-initiating cells (LICs). METHODS: The ATO/IFN combination was tested in vitro on imatinib (IMN)-resistant K562-R and Ar230-R cells. 3-(4,5-Dimethylthiazol-2-yl)-2,5-diphenyl tetrazolium bromide and terminal deoxynucleotidyl transferase-mediated deoxyuridine triphosphate nick-end labeling assays were used to evaluate proliferation and apoptosis, respectively. The acridine orange assay was used to assess autophagy, and quantitative reverse transcription-polymerase chain reaction was used to assess the involvement of the hedgehog (Hh) pathway. In vivo, a retroviral transduction/transplantation T315I BCR-ABL CML mouse model was used to assay the effect of the treatment on survival, tumor burden (histopathology and blood counts), and LIC activity (secondary transplantation). RESULTS: In vitro, ATO/IFN synergized to inhibit proliferation and induce apoptosis of IMN-resistant cells with variant modes of resistance. Furthermore, the preclinical effects of ATO/IFN were associated with induction of autophagy along with inhibition of the Hh pathway. Most remarkably, ATO/IFN significantly prolonged the survival of primary T315I-CML mice and displayed a dramatic impairment of disease engraftment in secondary mice, which reflected decreased LIC activity. CONCLUSIONS: Collectively, the ATO/IFN strategy has been demonstrated to have the potential to lead to durable remissions in TKI-resistant CML preclinical models and to overcome various TKI-specific mechanisms of resistance.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Resistencia a Antineoplásicos/efectos de los fármacos , Leucemia Experimental/tratamiento farmacológico , Leucemia Mielógena Crónica BCR-ABL Positiva/tratamiento farmacológico , Inhibidores de Proteínas Quinasas/farmacología , Animales , Apoptosis/efectos de los fármacos , Trióxido de Arsénico/administración & dosificación , Autofagia/efectos de los fármacos , Proteínas de Fusión bcr-abl/metabolismo , Proteínas Hedgehog/metabolismo , Humanos , Mesilato de Imatinib/farmacología , Interferón-alfa/administración & dosificación , Leucemia Experimental/patología , Leucemia Mielógena Crónica BCR-ABL Positiva/metabolismo , Ratones Endogámicos BALB C , Ratones Transgénicos
19.
Cancer Res ; 79(9): 2339-2351, 2019 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-30862722

RESUMEN

Acute lymphoblastic leukemia (ALL) is the most common cancer in children. The highest rates of treatment failure occur in specific genetic subsets of ALL, including hypodiploid B-cell ALL (B-ALL), for which effective alternative therapies to current intensive chemotherapy treatments have yet to be developed. Here, we integrated biochemical and genomic profiling with functional drug assays to select effective agents with therapeutic potential against hypodiploid B-ALL. ABT-199, a selective Bcl-2 inhibitor, was effective in reducing leukemic burden in vitro and in vivo in patient-derived xenograft models of hypodiploid B-ALL. Daily oral treatment with ABT-199 significantly increased survival in xenografted mice. The unexpected efficacy of ABT-199 observed in hypodiploid leukemias lacking BIM expression (the major reported mediator of ABT-199-induced apoptosis) led us to investigate the mechanism of action of ABT-199 in the absence of BIM. Treatment with ABT-199 elicited responses in a dose-dependent manner, from cell-cycle arrest at low nanomolar concentrations to cell death at concentrations above 100 nmol/L. Collectively, these results demonstrate the efficacy of Bcl-2 inhibition and potential therapeutic strategy in hypodiploid B-ALL. SIGNIFICANCE: These results demonstrate the efficacy of ABT-199 in vivo and provide encouraging preclinical data of Bcl-2 as a potential target for the treatment of hypodiploid B-ALL.


Asunto(s)
Antineoplásicos/farmacología , Diploidia , Leucemia Experimental/tratamiento farmacológico , Leucemia-Linfoma Linfoblástico de Células Precursoras B/tratamiento farmacológico , Proteínas Proto-Oncogénicas c-bcl-2/antagonistas & inhibidores , Bibliotecas de Moléculas Pequeñas/farmacología , Animales , Apoptosis , Compuestos Bicíclicos Heterocíclicos con Puentes/farmacología , Linaje de la Célula , Proliferación Celular , Humanos , Leucemia Experimental/metabolismo , Leucemia Experimental/patología , Ratones , Ratones Endogámicos NOD , Ratones SCID , Leucemia-Linfoma Linfoblástico de Células Precursoras B/metabolismo , Leucemia-Linfoma Linfoblástico de Células Precursoras B/patología , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Sulfonamidas/farmacología , Células Tumorales Cultivadas , Ensayos Antitumor por Modelo de Xenoinjerto
20.
Environ Toxicol ; 34(5): 659-665, 2019 May.
Artículo en Inglés | MEDLINE | ID: mdl-30761740

RESUMEN

Ouabain, a cardiotonic steroid, was used for the treatment of heart failure and atrial fibrillation and induces cancer cell apoptosis in many human cancer cells including human leukemia cells. However, there are no reports to show the effects on immune responses in a leukemia mouse model. In this study, WEHI-3 cell generated leukemia mice were developed and treated by oral ouabain at 0, 0.75, 1.5, and 3 mg/kg for 15 days. Results indicated that ouabain did not affect body appearance, but decreased liver and spleen weights, B- and T-cell proliferation at all three doses treatment and increased CD19 cells at 3.0 mg/kg treatment, decreased CD3, CD11b, and Mac-3 cells levels compared with positive control. Furthermore, ouabain increased the macrophage phagocytosis from peripheral blood mononuclear cell and peritoneal cavity at all three doses treatment and increased NK cell activities. Ouabain restored GOT, GPT and LDH levels in WEHI-3 leukemia mice in vivo.


Asunto(s)
Antineoplásicos Fitogénicos/uso terapéutico , Citotoxicidad Inmunológica/efectos de los fármacos , Células Asesinas Naturales/efectos de los fármacos , Leucemia Experimental/tratamiento farmacológico , Activación de Linfocitos/efectos de los fármacos , Ouabaína/uso terapéutico , Fagocitosis/efectos de los fármacos , Animales , Línea Celular Tumoral , Células Asesinas Naturales/inmunología , Leucemia Experimental/inmunología , Leucemia Experimental/patología , Activación de Linfocitos/inmunología , Ratones , Ratones Endogámicos BALB C , Trasplante de Neoplasias , Fagocitosis/inmunología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...