Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 479
Filtrar
1.
Int J Mol Sci ; 24(9)2023 Apr 30.
Artigo em Inglês | MEDLINE | ID: mdl-37175820

RESUMO

Sanguinarine (1) is a natural product with significant pharmacological effects. However, the application of sanguinarine has been limited due to its toxic side effects and a lack of clarity regarding its molecular mechanisms. To reduce the toxic side effects of sanguinarine, its cyanide derivative (1a) was first designed and synthesized in our previous research. In this study, we confirmed that 1a presents lower toxicity than sanguinarine but shows comparable anti-leukemia activity. Further biological studies using RNA-seq, lentiviral transfection, Western blotting, and flow cytometry analysis first revealed that both compounds 1 and 1a inhibited the proliferation and induced the apoptosis of leukemic cells by regulating the transcription of c-MET and then suppressing downstream pathways, including the MAPK, PI3K/AKT and JAK/STAT pathways. Collectively, the data indicate that 1a, as a potential anti-leukemia lead compound regulating c-MET transcription, exhibits better safety than 1 while maintaining cytostatic activity through the same mechanism as 1.


Assuntos
Citostáticos , Leucemia Eritroblástica Aguda , Leucemia , Humanos , Citostáticos/farmacologia , Leucemia Eritroblástica Aguda/tratamento farmacológico , Fosfatidilinositol 3-Quinases/metabolismo , Cianetos , Apoptose , Leucemia/tratamento farmacológico , Proteínas Proto-Oncogênicas c-akt/metabolismo , Linhagem Celular Tumoral
2.
BMC Cancer ; 23(1): 306, 2023 Apr 04.
Artigo em Inglês | MEDLINE | ID: mdl-37016335

RESUMO

BACKGROUND: Lovastatin, an HMG-CoA inhibitor and an effective cholesterol lowering drug, exhibits anti-neoplastic activity towards several types of cancer, although the underlying mechanism is still not fully understood. Herein, we investigated mechanism of growth inhibition of leukemic cells by lovastatin. METHODS: RNAseq analysis was used to explore the effect of lovastatin on gene expression in leukemic cells. An animal model of leukemia was used to test the effect of this statin in vivo. FAM83A and DDIT4 expression was knocked-downed in leukemia cells via lentivirus-shRNA. Western blotting, RT-qPCR, cell cycle analysis and apoptosis assays were used to determine the effect of lovastatin-induced growth suppression in leukemic cells in vitro. RESULTS: Lovastatin treatment strongly inhibited cancer progression in a mouse model of erythroleukemia induced by Friend virus. In tissue culture, lovastatin inhibited cell proliferation through induction of G1 phase cell cycle arrest and apoptosis. Interestingly, lovastatin induced most known genes associated with cholesterol biosynthesis in leukemic cells. Moreover, it suppressed ERK1/2 phosphorylation by downregulating FAM83A and DDIT4, two mediators of MAP-Kinase signaling. RNAseq analysis of lovastatin treated leukemic cells revealed a strong induction of the tumor suppressor gene KLF2. Accordingly, lentivirus-mediated knockdown of KLF2 antagonized leukemia cell suppression induced by lovastatin, associated with higher ERK1/2 phosphorylation compared to control. We further show that KLF2 induction by lovastatin is responsible for lower expression of the FAM83A and DDIT4 oncogenes, involved in the activation of ERK1/2. KLF2 activation by lovastatin also activated a subset of cholesterol biosynthesis genes that may further contribute to leukemia suppression. CONCLUSIONS: These results implicate KLF2-mediated FAM83A/DDIT4/MAPK suppression and activation of cholesterol biosynthesis as the mechanism of leukemia cell growth inhibition by lovastatin.


Assuntos
Inibidores de Hidroximetilglutaril-CoA Redutases , Leucemia Eritroblástica Aguda , Neoplasias , Animais , Camundongos , Lovastatina/farmacologia , Leucemia Eritroblástica Aguda/tratamento farmacológico , Leucemia Eritroblástica Aguda/genética , Inibidores de Hidroximetilglutaril-CoA Redutases/farmacologia , Inibidores de Hidroximetilglutaril-CoA Redutases/uso terapêutico , Colesterol , Apoptose , Fatores de Transcrição Kruppel-Like/genética
3.
Arch Pharm (Weinheim) ; 355(12): e2200367, 2022 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-36216575

RESUMO

Chronic myeloid leukemia accounts for human deaths worldwide and could enhance sevenfold by 2050. Thus, the treatment regimen for this disorder is highly crucial at this time. Flavaglines are a natural class of cyclopentane benzofurans exhibiting various bioactivities like anticancer action. Despite the antiproliferative activity of flavaglines against diverse cancer cells, their roles and mechanism of action in chronic myeloid leukemia (CML) remain poorly understood. Thus, this study examines the antiproliferative effect of a newly synthesized flavagline derivative, 1-chloracetylrocaglaol (A2074), on erythroleukemia K562 cells and the zebrafish xenograft model. The study revealed that A2074 could inhibit proliferation, promote apoptosis, and boost megakaryocyte differentiation of K562 cells. This flavagline downregulated c-MYC and miR-17-92 cluster genes, targeting upregulation of the apoptotic protein Bcl-2-like protein 11 (BIM). The work uncovered a critical role of the c-MYC-miR-17-92-BIM axis in the growth and survival of CML cells.


Assuntos
Leucemia Eritroblástica Aguda , Leucemia Mielogênica Crônica BCR-ABL Positiva , MicroRNAs , Animais , Humanos , Células K562 , Leucemia Eritroblástica Aguda/tratamento farmacológico , Leucemia Eritroblástica Aguda/genética , Peixe-Zebra/genética , Peixe-Zebra/metabolismo , MicroRNAs/genética , MicroRNAs/metabolismo , MicroRNAs/farmacologia , Relação Estrutura-Atividade , Apoptose , Leucemia Mielogênica Crônica BCR-ABL Positiva/tratamento farmacológico , Proliferação de Células
4.
Food Chem Toxicol ; 168: 113336, 2022 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-35963475

RESUMO

The novel di-and triphenyltin(IV) dithiocarbamate compounds represented as RnSnL2 (where R = C4H9, C6H5; n = 2,3; L = N,N-dithiocarbamate), Ph2Sn(N,N-diisopropyldithiocarbamate) (OC1), Ph3Sn(N,N-diisopropyldithiocarbamate) (OC2), Ph2Sn(N,N-diallyldithiocarbamate) (OC3), Ph3Sn(N,N-diallyldithiocarbamate) (OC4), and Ph2Sn(N,N-diethyldithiocarbamate) (OC5) were assessed for their cytotoxicity in K562 human erythroleukemia cells. All compounds inhibited the growth of cells at low micromolar concentrations (<10 µM), and the mechanism underlying their antiproliferative effects on K562 cells was apoptosis, as corroborated by the exposure of plasma membrane phosphatidylserine. OC2, which showed the most promising antiproliferative activity, was selected for further analyses. The results demonstrated that OC2 induced apoptosis in K562 cells via an intrinsic mitochondrial pathway triggered upon DNA damage, an early apoptotic signal. Subsequently, OC2 produced excessive intracellular reactive oxygen species. The role of oxidative stress was corroborated by the significant reduction in GSH levels and percentage of apoptosis in NAC-pretreated cells. OC2 could arrest the cell cycle progression in the S phase. These new findings elucidate the antiproliferative potential of OC2 in the K562 human erythroleukemia cells and warrant further investigation, specifically to determine the exact signaling pathway underlying its antileukemic efficacy.


Assuntos
Leucemia Eritroblástica Aguda , Apoptose , Dano ao DNA , Ditiocarb/análogos & derivados , Ditiocarb/metabolismo , Ditiocarb/farmacologia , Humanos , Células K562 , Leucemia Eritroblástica Aguda/tratamento farmacológico , Leucemia Eritroblástica Aguda/metabolismo , Mitocôndrias , Compostos Orgânicos de Estanho , Fosfatidilserinas/metabolismo , Espécies Reativas de Oxigênio/metabolismo
5.
Blood Cancer Discov ; 3(5): 410-427, 2022 09 06.
Artigo em Inglês | MEDLINE | ID: mdl-35839275

RESUMO

Acute erythroid leukemia (AEL) is a unique subtype of acute myeloid leukemia characterized by prominent erythroid proliferation whose molecular basis is poorly understood. To elucidate the underlying mechanism of erythroid proliferation, we analyzed 121 AEL using whole-genome, whole-exome, and/or targeted-capture sequencing, together with transcriptome analysis of 21 AEL samples. Combining publicly available sequencing data, we found a high frequency of gains and amplifications involving EPOR/JAK2 in TP53-mutated cases, particularly those having >80% erythroblasts designated as pure erythroid leukemia (10/13). These cases were frequently accompanied by gains and amplifications of ERG/ETS2 and associated with a very poor prognosis, even compared with other TP53-mutated AEL. In addition to activation of the STAT5 pathway, a common feature across all AEL cases, these AEL cases exhibited enhanced cell proliferation and heme metabolism and often showed high sensitivity to ruxolitinib in vitro and in xenograft models, highlighting a potential role of JAK2 inhibition in therapeutics of AEL. SIGNIFICANCE: This study reveals the major role of gains, amplifications, and mutations of EPOR and JAK2 in the pathogenesis of pure erythroleukemia. Their frequent response to ruxolitinib in patient-derived xenograft and cell culture models highlights a possible therapeutic role of JAK2 inhibition for erythroleukemia with EPOR/JAK2-involving lesions. This article is highlighted in the In This Issue feature, p. 369.


Assuntos
Janus Quinase 2 , Leucemia Eritroblástica Aguda , Leucemia Mieloide Aguda , Receptores da Eritropoetina , Exoma , Humanos , Janus Quinase 2/genética , Leucemia Eritroblástica Aguda/tratamento farmacológico , Leucemia Mieloide Aguda/tratamento farmacológico , Mutação , Prognóstico , Receptores da Eritropoetina/genética
6.
Leuk Res ; 118: 106860, 2022 07.
Artigo em Inglês | MEDLINE | ID: mdl-35636055

RESUMO

Pure erythroid leukemia (PEL) is a rare acute leukemia with a dismal prognosis. TP53 mutations are a dominant feature of PEL, but the characteristics of TP53 alterations in PEL without prior exposure to cytotoxic therapy (d-PEL) or with such exposure (t-PEL) is unknown. We studied 25 patients with TP53-mutated PEL including 16 d-PEL and 9 t-PEL. Both groups had comparable clinical findings and overall survival. The TP53 mutation, commonly missense, was present in the dominant clone in all cases. In the d-PEL group, 10/16 (62.5%) had one TP53 mutation compared to 8/9 (89%) patients in the t-PEL group. In the d-PEL group, 9/16 (56.2%) patients had hotspot mutations compared to 2 (22.2%) patients in the t-PEL group. Notably, monosomy 17 or del(17p) were less common in the d-PEL group (26.6%) compared to the t-PEL group (71.4%), underscoring distinctive TP53 alterations in d-PEL versus t-PEL, possibly reflecting different fitness advantages.


Assuntos
Leucemia Eritroblástica Aguda , Leucemia Mieloide Aguda , Humanos , Leucemia Eritroblástica Aguda/tratamento farmacológico , Leucemia Eritroblástica Aguda/genética , Leucemia Mieloide Aguda/genética , Monossomia , Mutação , Prognóstico , Proteína Supressora de Tumor p53/genética
7.
Rinsho Ketsueki ; 63(2): 121-133, 2022.
Artigo em Japonês | MEDLINE | ID: mdl-35264503

RESUMO

Acute erythroid leukemia (AEL) is a unique subtype of acute myeloid leukemia characterized by erythroid predominance and dysplasia. It is classified into two subtypes: pure erythroid (PEL) and erythroid/myeloid (EML) phenotypes. To understand the mechanism of the erythroid dominant phenotype of AEL and identify potential therapeutic targets for AEL, we analyzed 105 AEL and 214 non-AEL cases using whole-genome/exome and/or targeted-capture sequencing, with SNP probes for detecting copy number abnormalities. We also performed a transcriptome analysis of 12 AEL samples. Combining publicly available sequencing data, AEL was genetically clustered into four groups according to mutational status in TP53, STAG2, and NPM1 genes. Conspicuously, highly recurrent gains and amplifications affecting EPOR, JAK2, and/or ERG/ETS2 were recurrently detected in AEL cases, almost exclusively found in TP53-mutated cases. Among these, gains/amplifications of EPOR/JAK2 were more highly enriched in PEL than EML cases. Along with the activated STAT5 pathway, a common feature across all AEL cases, these AEL cases exhibited enhanced cell proliferation and heme metabolism, and they showed high sensitivity to ruxolitinib in in vitro and in xenograft models, highlighting the potential role of JAK2 inhibition in AEL therapeutics.


Assuntos
Leucemia Eritroblástica Aguda , Leucemia Mieloide Aguda , Criança , Exoma , Humanos , Leucemia Eritroblástica Aguda/diagnóstico , Leucemia Eritroblástica Aguda/tratamento farmacológico , Leucemia Eritroblástica Aguda/genética , Leucemia Mieloide Aguda/genética , Mutação , Fenótipo
8.
Cancer Gene Ther ; 29(6): 784-792, 2022 06.
Artigo em Inglês | MEDLINE | ID: mdl-34117374

RESUMO

Cabozantinib is a potent tyrosine kinase inhibitor with multiple targets including MET, VEGFR2, RET, KIT, and FLT3. Cabozantinib is widely used for the treatment of medullary thyroid cancer and renal cell carcinoma. We recently suggested cabozantinib as a potential therapeutic alternative for acute myeloid leukemia (AML) patients with FLT3-internal tandem duplication (FLT3-ITD). Here, we report that cabozantinib can promote differentiation in erythroid leukemia cells. We found that K562 erythroid leukemia cells treated with 1 µM cabozantinib for 72 h underwent erythroid lineage differentiation. Transcriptomic analysis revealed that various pathways associated with heme biosynthesis, hemoglobin production, and GATA1 targets were upregulated, whereas cell survival pathways were downregulated. Further examination revealed that cabozantinib-induced erythroid differentiation is at least in part regulated by JNK activation and phosphorylation. Levels of phosphorylated BCR-ABL, AKT, STAT5, ERK, and p38 also decreased following cabozantinib treatment. Therefore, we indicate that cabozantinib has dual functions. First, it induces K562 cell differentiation toward the erythroid lineage by upregulating heme biosynthesis, globin synthesis, and erythroid-associated reactions. Second, cabozantinib inhibits K562 cell proliferation by inhibiting the phosphorylation of BCR-ABL and the downstream MAPK, PI3K-AKT, and JAK-STAT signaling pathways.


Assuntos
Leucemia Eritroblástica Aguda , Anilidas , Diferenciação Celular/fisiologia , Ativação Enzimática , Expressão Gênica , Heme , Humanos , Células K562 , Leucemia Eritroblástica Aguda/tratamento farmacológico , Leucemia Eritroblástica Aguda/genética , MAP Quinase Quinase 4/metabolismo , Fosfatidilinositol 3-Quinases , Proteínas Proto-Oncogênicas c-akt , Piridinas
9.
Vet Med Sci ; 8(1): 9-13, 2022 01.
Artigo em Inglês | MEDLINE | ID: mdl-34599794

RESUMO

Erythroleukaemia is a malignant neoplasm of the erythroid lineage that rarely occurs in cats. It is associated with the feline leukaemia virus (FeLV), and owing to the poor prognosis, treatment is rarely reported. A 4-year-old female Korean domestic shorthair cat was presented with hyporexia, fever, lethargy, severe anaemia and rubricytosis. An FeLV antigen test was positive, but a subsequent polymerase chain reaction test was negative. Serum biochemistry analysis results were normal, except for slightly elevated alanine aminotransferase. The patient was tentatively diagnosed with acute erythroleukaemia, and single high-dose (600 mg/m2 ) cytarabine chemotherapy was administered via constant rate infusion for 12 h a day for 5 days. After the first cytarabine administration, the clinical signs and anaemia improved, though no change was noted to other haematological parameters. The patient died of shock 16 days after the second cytarabine administration; the total survival time after diagnosis was 67 days. Post-mortem cytological evaluation of bone marrow aspiration revealed that the myeloid/erythroid ratio was 0.49, the erythroid progenitor cells were 64% of all nucleated cells and the blast cells were 84% of the non-erythroid cells. Histopathology images indicated that the spleen was diffusely expanded by atypical round cells, possibly erythroid precursors. This is the first case report on the prognosis and effects of high-dose cytarabine chemotherapy for acute feline erythroleukaemia with FeLV infection. Although the clinical signs improved, the treatment was not effective. Further studies on erythroleukaemia chemotherapy protocols are required.


Assuntos
Doenças do Gato , Leucemia Eritroblástica Aguda , Animais , Doenças do Gato/tratamento farmacológico , Gatos , Citarabina/uso terapêutico , Feminino , Vírus da Leucemia Felina , Leucemia Eritroblástica Aguda/tratamento farmacológico , Leucemia Eritroblástica Aguda/veterinária , Baço
10.
Int J Mol Sci ; 22(21)2021 Nov 05.
Artigo em Inglês | MEDLINE | ID: mdl-34769424

RESUMO

A series of heterocyclic compounds containing a spiro-fused pyrrolo[3,4-a]pyrrolizine and tryptanthrin framework have been synthesized and studied as potential antitumor agents. Cytotoxicity of products was screened against human erythroleukemia (K562) and human cervical carcinoma (HeLa) cell lines. Among the screened compounds. 4a, 4b and 5a were active against human erythroleukemia (K562) cell line, while 4a and 5a were active against cervical carcinoma (HeLa) cell line. In agreement with the DNA cytometry studies, the tested compounds have achieved significant cell-cycle perturbation with higher accumulation of cells in G2/M phase and induced apoptosis. Using confocal microscopy, we found that with 4a and 5a treatment of HeLa cells, actin filaments disappeared, and granular actin was distributed diffusely in the cytoplasm in 76-91% of cells. We discovered that HeLa cells after treatment with compounds 4a and 5a significantly reduced the number of cells with filopodium-like membrane protrusions (from 63 % in control cells to 29% after treatment) and a decrease in cell motility.


Assuntos
Antineoplásicos/farmacologia , Leucemia Eritroblástica Aguda/tratamento farmacológico , Pirrolidinas/síntese química , Pirrolidinas/farmacologia , Quinazolinas/farmacologia , Compostos de Espiro/farmacologia , Neoplasias do Colo do Útero/tratamento farmacológico , Antineoplásicos/síntese química , Apoptose/efeitos dos fármacos , Ciclo Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Movimento Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Ensaios de Seleção de Medicamentos Antitumorais , Feminino , Humanos , Leucemia Eritroblástica Aguda/metabolismo , Leucemia Eritroblástica Aguda/patologia , Quinazolinas/síntese química , Compostos de Espiro/síntese química , Neoplasias do Colo do Útero/metabolismo , Neoplasias do Colo do Útero/patologia
12.
Curr Oncol Rep ; 23(6): 69, 2021 04 20.
Artigo em Inglês | MEDLINE | ID: mdl-33876292

RESUMO

PURPOSE OF THE REVIEW: Acute erythroleukemia (AEL) is a rare form of acute myeloid leukemia recognized by erythroblastic proliferation. Many controversies remain around diagnosis influencing prognostic and therapeutic implications relating to this unique leukemia subset. RECENT FINDINGS: The 2016 WHO classification includes more clear and restrictive diagnostic criteria for AEL. Primary acute erythroid leukemia is associated with complex and high-risk karyotypes including chromosomes 5q and 7q abnormalities. Mutational data shows that AEL is characterized by far lower NPM1 and FLT3-ITD mutation rates and higher mutational rates in TP53 compared with other AML subtypes. Hypomethylating agents have shown therapeutic value in AEL. In this article, we discuss the evolving diagnostic concepts of erythroleukemia, genomics, clinical outcome, and promising therapeutic targets through an appraisal of the current literature.


Assuntos
Leucemia Eritroblástica Aguda/tratamento farmacológico , Aberrações Cromossômicas , Humanos , Imunofenotipagem , Leucemia Eritroblástica Aguda/diagnóstico , Leucemia Eritroblástica Aguda/genética , Leucemia Eritroblástica Aguda/imunologia , Mutação
13.
Pharmacol Res ; 166: 105491, 2021 04.
Artigo em Inglês | MEDLINE | ID: mdl-33582247

RESUMO

Acute erythroid leukemia (AEL) is a rare and aggressive hematologic malignancy with no specific treatment. Sanguisorba officinalis L. (S. officinalis), a well-known traditional Chinese medicine, possesses potent anticancer activity. However, the active components of S. officinalis against AEL and the associated molecular mechanisms remain unknown. In this study, we predicted the anti-AML effect of S. officinalis based on network pharmacology. Through the identification of active components of S. officinalis, we found that 3,8-Di-O-methylellagic acid 2-O-glucoside (DMAG) not only significantly inhibited the proliferation of erythroleukemic cell line HEL, but also induced their differentiation to megakaryocytes. Furthermore, we demonstrated that DMAG could prolong the survival of AEL mice model. Whole-transcriptome sequencing was performed to elucidate the underlying molecular mechanisms associated with anti-AEL effect of DMAG. The results showed that the total of 68 miRNAs, 595 lncRNAs, 4030 mRNAs and 35 circRNAs were significantly differentially expressed during DMAG induced proliferation inhibition and differentiation of HEL cells. Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analyses revealed that the differentially expressed miRNAs, lncRNAs, mRNAs and circRNAs were mainly involved in metabolic, HIF-1, MAPK, Notch pathway and apoptosis. The co-expression networks showed that miR-23a-5p, miR-92a-1-5p, miR-146b and miR-760 regulatory networks were crucial for megakaryocyte differentiation induced by DMAG. In conclusion, our results suggest that DMAG, derived from S. officinalis might be a potent differentiation inducer of AEL cells and provide important information on the underlying mechanisms associated with its anti-AEL activity.


Assuntos
Antineoplásicos Fitogênicos/farmacologia , Leucemia Eritroblástica Aguda/tratamento farmacológico , Sanguisorba , Antineoplásicos Fitogênicos/química , Diferenciação Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Regulação Leucêmica da Expressão Gênica/efeitos dos fármacos , Humanos , Leucemia Eritroblástica Aguda/genética , Leucemia Eritroblástica Aguda/patologia , Farmacologia em Rede , Sanguisorba/química , Transcriptoma/efeitos dos fármacos
14.
Curr Mol Med ; 21(10): 819-831, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33475071

RESUMO

Acute erythroid leukemia (AEL) is a subtype of acute myeloid leukemia (AML) with features such as accumulation of maturation-arrested erythroblasts. Compared with AML, the progression of AEL is faster and the prognosis to available therapy is worse. However, its categorization is still being updated and the pathophysiology of AEL is still under research, making diagnosis and chemotherapy challenging for physicians. To achieve better outcomes, therapies should be optimized and new drugs should be developed. In this review, we summarize current strategies of diagnosis and therapies of AEL, and discuss prospective targets for chemotherapeutic agents based on the biological characteristics of AEL neoplastic cells as well as transcriptional factors and pathways related to erythroid differentiation.


Assuntos
Diferenciação Celular/genética , Leucemia Eritroblástica Aguda , Mutação , Proteínas de Neoplasias , Fatores de Transcrição , Humanos , Leucemia Eritroblástica Aguda/tratamento farmacológico , Leucemia Eritroblástica Aguda/genética , Leucemia Eritroblástica Aguda/metabolismo , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo
15.
Anticancer Agents Med Chem ; 21(14): 1911-1920, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33397267

RESUMO

BACKGROUND: Quercetin has potential against the Multidrug Resistance (MDR) phenotype, but with low bioavailability. The increase in the bioavailability can be obtained with nanostructures. OBJECTIVE: To analyze the effects of quercetin and its nanoemulsion on MDR and non-MDR cells. METHODS: We used high-pressure homogenization for nanoemulsion production; Trypan Blue for cytostatic/cytotoxicity assays; Epifluorescence microscope (with specific probes) for apoptosis and DNA damage; Real-Time PCR for gene expression; AutoDock Vina for docking and Flow Cytometry for efflux analysis. Quercetin exerted antiproliferative impact, induced apoptosis, necrosis and DNA damage on cells. RESULTS: Quercetin combined with vincristine showed an effect similar to verapamil (an ABCB1 inhibitor), and docking showed that it binds to ABCB1 in a similar region. Quercetin was also capable of altering ABCB1 gene expression. Quercetin in nanoemulsion maintained the cytotoxic and cytostatic effects of quercetin, which may increase bioavailability. Besides, the unloaded nanoemulsion was able to inhibit per se the efflux activity of ABCB1, demonstrating pharmacological action of this structure. CONCLUSION: Quercetin may be considered as a prospective drug to overcome resistance in cancer cells and its nanoemulsion can be an alternative for in vivo application.


Assuntos
Antineoplásicos/farmacologia , Resistência a Múltiplos Medicamentos/efeitos dos fármacos , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Leucemia Eritroblástica Aguda/tratamento farmacológico , Nanopartículas/química , Quercetina/farmacologia , Antineoplásicos/química , Apoptose/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Dano ao DNA , Ensaios de Seleção de Medicamentos Antitumorais , Emulsões , Citometria de Fluxo , Humanos , Leucemia Eritroblástica Aguda/patologia , Simulação de Acoplamento Molecular , Quercetina/química , Células Tumorais Cultivadas
16.
Anticancer Agents Med Chem ; 21(4): 498-507, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-32538736

RESUMO

BACKGROUND AND PURPOSE: Leukemia is considered a top-listed ailment, according to WHO, which contributes to the death of a major population of the world every year. Paris Saponin VII (PS), a saponin which was isolated from the roots of Trillium kamtschaticum, from our group, was reported to provide hemostatic, cytotoxic and antimicrobial activities. However, its molecular mechanism underlying the anti-proliferative effects remains unclear. Thus, this study hypothesized to assess that mechanism in PS treated HEL cells. METHODS: The MTT assay was used to analyze the PS inhibited cell viability in the HEL cells. We further found that PS could induce S phase cell cycle arrest through flow cytometry as well as the western blot analysis of intrinsic and extrinsic apoptotic molecules. RESULTS: The MTT assay showed the IC50 concentration of PS as 0.667µM. The study revealed that PS treatment inhibits cell proliferation dose-dependently. It further caused mitochondrial membrane potential changes by PS treatment. Mechanistic protein expression revealed a dose-dependent upsurge for Bid and Bim molecules, while Bcl2 and PARP expression levels were significantly (P<0.05) down-regulated in PS treated HEL cells resulting in caspase -3 release and increased the Bim levels upon 24h of incubation. CONCLUSION: These findings indicate that PS possesses an excellent anti-leukemic activity via the regulation of the mitochondrial pathway, leading to S phase cell cycle arrest and caspase-dependent apoptosis, suggesting it as a potential alternative chemotherapeutic agent for leukemia patients.


Assuntos
Antineoplásicos Fitogênicos/farmacologia , Apoptose/efeitos dos fármacos , Leucemia Eritroblástica Aguda/tratamento farmacológico , Extratos Vegetais/farmacologia , Saponinas/farmacologia , Antineoplásicos Fitogênicos/química , Antineoplásicos Fitogênicos/isolamento & purificação , Pontos de Checagem do Ciclo Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Relação Dose-Resposta a Droga , Ensaios de Seleção de Medicamentos Antitumorais , Humanos , Leucemia Eritroblástica Aguda/metabolismo , Leucemia Eritroblástica Aguda/patologia , Membranas Mitocondriais/efeitos dos fármacos , Estrutura Molecular , Extratos Vegetais/química , Extratos Vegetais/isolamento & purificação , Saponinas/química , Saponinas/isolamento & purificação , Transdução de Sinais/efeitos dos fármacos , Relação Estrutura-Atividade , Células Tumorais Cultivadas
17.
Mol Cancer Res ; 19(3): 451-464, 2021 03.
Artigo em Inglês | MEDLINE | ID: mdl-33172974

RESUMO

DNA methyltransferase inhibitors (DNMTI) like 5-Azacytidine (5-Aza) are the only disease-modifying drugs approved for the treatment of higher-risk myelodysplastic syndromes (MDS), however less than 50% of patients respond, and there are no predictors of response with clinical utility. Somatic mutations in the DNA methylation regulating gene tet-methylcytosine dioxygenase 2 (TET2) are associated with response to DNMTIs, however the mechanisms responsible for this association remain unknown. Using bisulfite padlock probes, mRNA sequencing, and hydroxymethylcytosine pull-down sequencing at several time points throughout 5-Aza treatment, we show that TET2 loss particularly influences DNA methylation (5mC) and hydroxymethylation (5hmC) patterns at erythroid gene enhancers and is associated with downregulation of erythroid gene expression in the human erythroleukemia cell line TF-1. 5-Aza disproportionately induces expression of these down-regulated genes in TET2KO cells and this effect is related to dynamic 5mC changes at erythroid gene enhancers after 5-Aza exposure. We identified differences in remethylation kinetics after 5-Aza exposure for several types of genomic regulatory elements, with distal enhancers exhibiting longer-lasting 5mC changes than other regions. This work highlights the role of 5mC and 5hmC dynamics at distal enhancers in regulating the expression of differentiation-associated gene signatures, and sheds light on how 5-Aza may be more effective in patients harboring TET2 mutations. IMPLICATIONS: TET2 loss in erythroleukemia cells induces hypermethylation and impaired expression of erythroid differentiation genes which can be specifically counteracted by 5-Azacytidine, providing a potential mechanism for the increased efficacy of 5-Aza in TET2-mutant patients with MDS. VISUAL OVERVIEW: http://mcr.aacrjournals.org/content/molcanres/19/3/451/F1.large.jpg.


Assuntos
Azacitidina/farmacologia , Proteínas de Ligação a DNA/deficiência , Dioxigenases/deficiência , Leucemia Eritroblástica Aguda/tratamento farmacológico , Diferenciação Celular/efeitos dos fármacos , Proteínas de Ligação a DNA/metabolismo , Dioxigenases/metabolismo , Expressão Gênica , Humanos , Leucemia Eritroblástica Aguda/genética , Leucemia Eritroblástica Aguda/patologia
18.
Bull Exp Biol Med ; 170(2): 251-253, 2020 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-33263857

RESUMO

The study examined the effect of BSA on cultured cells of breast cancer, erythroleukemia, and ovarian carcinoma. BSA cytotoxic activity was examined with light microscopy and spectrophotometry in the concentration range of 6.25 to 50 mg/ml. The high concentrations of BSA disrupted the tumor cells in parallel with formation of vesicular debris. The destruction parameters were similar in diverse types of cells: BSA (50 mg/ml) disrupted 75.5±0.7% breast cancer cells, 75.8±0.2% erythroleukemia cells, and 78.8±0.1% cells of ovarian carcinoma (p≥0.05). The effect of BSA was dose-dependent in each type of cells. The data attest that BSA can disrupt various tumor cells in vitro.


Assuntos
Albuminas/química , Neoplasias da Mama/tratamento farmacológico , Carcinoma/tratamento farmacológico , Leucemia Eritroblástica Aguda/tratamento farmacológico , Neoplasias Ovarianas/tratamento farmacológico , Soroalbumina Bovina/metabolismo , Animais , Antineoplásicos/farmacologia , Bovinos , Linhagem Celular Tumoral , Ensaios de Seleção de Medicamentos Antitumorais , Feminino , Humanos , Técnicas In Vitro , Células K562 , Albumina Sérica Humana/metabolismo , Células Tumorais Cultivadas
19.
Chin J Nat Med ; 18(10): 749-759, 2020 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-33039054

RESUMO

This study aimed to investigate the effects of Sargassum fusiforme polysaccharide (SFPS I, II, and III) on the apoptosis and regulation of human erythroleukemia (HEL) cells. The effect of different doses of SFPS on HEL cell growth was detected using the Cell Counting Kit-8 method, and apoptosis was detected by Hoechst staining. Cell cycle distribution and apoptosis were detected using flow cytometry. Expression of the cell cycle gene, p53, antiapoptotic genes, Bcl-xL and Bcl-2, and pro-apoptotic genes, Bax, Bad, and Caspase-3, as well as the expression of the corresponding proteins, were detected using real-time quantitative polymerase chain reaction (qPCR) and Western blot. The results showed that SFPS II and III decreased HEL cell viability and induced HEL cell apoptosis. Different concentrations of SFPS (I, II, and III) were detected that induced much less toxic effect in normal human embryonic lung (MRC-5) cells, and SFPS I increased cell proliferation, indicating its favorable selectivity towards cancer cells. The mechanism by which SFPS induced apoptosis was also found to be related to the induction of cell cycle arrest in the G0/G1 phase and the increased expression of apoptosis-related genes and proteins. We concluded that SFPS induces HEL cell apoptosis, possibly via activation of the Caspase pathway, providing the theoretical basis for the development of SFPS-based anti-tumor drug products.


Assuntos
Antineoplásicos/farmacologia , Apoptose/efeitos dos fármacos , Leucemia Eritroblástica Aguda/patologia , Polissacarídeos/farmacologia , Sargassum/química , Ciclo Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Humanos , Leucemia Eritroblástica Aguda/tratamento farmacológico
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA