Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Pharmacol Rep ; 75(4): 876-890, 2023 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-37332080

RESUMO

Although Warburg's discovery of intensive glucose uptake by tumors, followed by lactate fermentation in oxygen presence of oxygen was made a century ago, it is still an area of intense research and development of new hypotheses that, layer by layer, unravel the complexities of neoplastic transformation. This seemingly simple metabolic reprogramming of cancer cells reveals an intriguing, multi-faceted nature that may link various phenomena including cell signaling, cell proliferation, ROS generation, energy supply, macromolecules synthesis/biosynthetic precursor supply, immunosuppression, or cooperation of cancerous cells with cancer-associated fibroblasts (CAFs), known as reversed Warburg effect. According to the current perception of the causes and consequences of the Warburg effect, PI3K/Akt/mTOR are the main signaling pathways that, in concert with the transcription factors HIF-1, p53, and c-Myc, modulate the activity/expression of key regulatory enzymes, including PKM2, and PDK1 to tune in the most optimal metabolic setting for the cancer cell. This in turn secures adequate levels of biosynthetic precursors, NADPH, NAD+, and rapid ATP production to meet the increased demands of intensively proliferating tumor cells. The end-product of "aerobic glycolysis", lactate, an oncometabolite, may provide fuel to neighboring cancer cells, and facilitate metastasis and immunosuppression together enabling cancer progression. The importance and possible applicability of the presented issue are best illustrated by numerous trials with various agents targeting the Warburg effect, constituting a promising strategy in future anti-cancer regimens. In this review, we present the key aspects of this multifactorial phenomenon, depicting the mechanisms and benefits behind the Warburg effect, and also pointing to selected aspects in the field of anticancer therapy.


Assuntos
Neoplasias , Fosfatidilinositol 3-Quinases , Humanos , Fosfatidilinositol 3-Quinases/metabolismo , Neoplasias/metabolismo , Transdução de Sinais , Oxigênio/metabolismo , Glicólise , Lactatos
2.
Int J Mol Sci ; 22(16)2021 Aug 17.
Artigo em Inglês | MEDLINE | ID: mdl-34445551

RESUMO

Cancer-specific isoenzyme of phosphofructokinase II (PFKFB4), as our previous research has shown, may be one of the most important enzymes contributing to the intensification of glycolysis in hypoxic malignant melanoma cells. Although the PFKFB4 gene seems to play a crucial role in the progression of melanoma, so far there are no complete data on the expression of PFKFB4 at the isoform level and the influence of hypoxia on alternative splicing. Using RT-qPCR and semi-quantitative RT-PCR, we presented the PFKFB4 gene expression profile at the level of six isoforms described in the OMIM NCBI database in normoxic and hypoxic melanoma cells. Additionally, using VMD software, we analyzed the structure of isoforms at the protein level, concluding about the catalytic activity of individual isoforms. Our research has shown that five isoforms of PFKFB4 are expressed in melanoma cells, of which the D and F isoforms are highly constitutive, while the canonical B isoform seems to be the main isoform induced in hypoxia. Our results also indicate that the expression profile at the level of the PFKFB4 gene does not reflect the expression at the level of individual isoforms. Our work clearly indicates that the PFKFB4 gene expression profile should be definitely analyzed at the level of individual isoforms. Moreover, the analysis at the protein level allowed the selection of those isoforms whose functional validation should be performed to fully understand the importance of PFKFB4 expression in the metabolic adaptation of malignant melanoma cells.


Assuntos
Processamento Alternativo , Biomarcadores Tumorais/metabolismo , Regulação Neoplásica da Expressão Gênica , Hipóxia/fisiopatologia , Melanoma/patologia , Oxigênio/metabolismo , Fosfofrutoquinase-2/genética , Biomarcadores Tumorais/genética , Glicólise , Humanos , Melanoma/genética , Melanoma/metabolismo , Células Tumorais Cultivadas
3.
Pharmacol Rep ; 72(5): 1407-1417, 2020 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-32715434

RESUMO

BACKGROUND: Multiple myeloma (MM) is defined as plasma cells malignancy, developing in the bone marrow. At the beginning of the disease, the malignant plasma cells are dependent on bone marrow microenvironment, providing growth and survival factors. Importantly, the recent studies pointed hypoxia as an important factor promoting progression of MM. In particular, hypoxia-triggered HIF-1 signaling was shown to promote chemoresistance, angiogenesis, invasiveness and induction of immature phenotype, suggesting that strategies targeting HIF-1 may contribute to improvement of anti-myeloma therapies. METHODS: The Western Blot and RT-PCR techniques were applied to analyze the influence of metformin on HIF-1 pathway in MM cells. To evaluate the effect of metformin on the growth of MM cell lines in normoxic and hypoxic conditions the MTT assay was used. The apoptosis induction in metformin treated hypoxic and normoxic cells was verified by Annexin V/PI staining followed by FACS analysis. RESULTS: Our results showed, for the first time, that metformin inhibits HIF-1 signaling in MM cells. Moreover, we demonstrated the effect of metformin to be mainly oxygen dependent, since the HIF-1 pathway was not significantly affected by metformin in anoxic conditions as well as after application of hypoxic mimicking compound, CoCl2. Our data also revealed that metformin triggers the growth arrest without inducing apoptosis in either normoxic or hypoxic conditions. CONCLUSIONS: Taken together, our study indicates metformin as a promising candidate for developing new treatment strategies exploiting HIF-1 signaling inhibition to enhance the overall anti-MM effect of currently used therapies, that may considerably benefit MM patients.


Assuntos
Fator 1 Induzível por Hipóxia/metabolismo , Metformina/farmacologia , Mieloma Múltiplo/tratamento farmacológico , Mieloma Múltiplo/metabolismo , Transdução de Sinais/efeitos dos fármacos , Apoptose/efeitos dos fármacos , Hipóxia Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Humanos , Microambiente Tumoral/efeitos dos fármacos
4.
Acta Biochim Pol ; 66(4): 619-625, 2019 Dec 11.
Artigo em Inglês | MEDLINE | ID: mdl-31826047

RESUMO

The CD146 (also known as MCAM, MUC-18, Mel-CAM) was initially reported in 1987, as a protein crucial for the invasiveness of malignant melanoma. Recently, it has been confirmed that CD146 has been involved in progression and poor overall survival of many cancers including breast cancer. Importantly, in independent studies, CD146 was reported to be a trigger of epithelial to mesenchymal transition in breast cancer cells. The goal of our current study was to verify the potential involvement of epigenetic mechanism behind the regulation of CD146 expression in breast cancer cells, as it has been previously reported in prostate cancer. First, we analysed the response of breast cancer cell lines, differing in the initial CD146 mRNA and protein content, to epigenetic modifier, 5-aza-2-deoxycytidine, and subsequently the methylation status of CD146 gene promoter was investigated, using direct bisulfite sequencing. We observed that treatment with demethylating agent led to induction of CD146 expression in all analysed breast cancer cell lines, both at mRNA and protein level, what was accompanied by increased expression of selected mesenchymal markers. Importantly, CD146 gene promoter analysis showed aberrant CpG island methylation in 2 out of 3 studied breast cancer cells lines, indicating epigenetic regulation of CD146 gene expression. In conclusion, our study revealed, for the first time, that aberrant methylation maybe involved in expression control of CD146, a very potent EMT inducer in breast cancer cells. Altogether, the data obtained may provide the basis for novel therapies as well as diagnostic approaches enabling sensitive and very accurate detection of breast cancer cells.


Assuntos
Neoplasias da Mama/genética , Metilação de DNA/efeitos dos fármacos , Decitabina/farmacologia , Neoplasias da Mama/diagnóstico , Neoplasias da Mama/tratamento farmacológico , Neoplasias da Mama/patologia , Antígeno CD146/genética , Ilhas de CpG/efeitos dos fármacos , Progressão da Doença , Epigênese Genética/efeitos dos fármacos , Transição Epitelial-Mesenquimal/efeitos dos fármacos , Feminino , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Humanos , Células MCF-7 , Regiões Promotoras Genéticas/efeitos dos fármacos , Análise de Sequência de DNA
5.
Cancer Cell Int ; 19: 292, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31754349

RESUMO

BACKGROUND/AIM: During cancer progression metabolic reprogramming is observed in parallel to the alternation in transcriptional profiles of malignant cells. Recent studies suggest that metabolic isoenzymes of phosphofructokinase II (PFK-II) - PFKFB3 and PFKFB4, often induced in hypoxic environment, significantly contribute to enhancement of glucose metabolism and in consequence cancer progression. MATERIALS AND METHODS: Using the publicly available data deposited in the R2 data base we performed a Kaplan-Meyer analysis for cancer patients divided into groups with high and low expression levels of PFKFB3/4, determined based on the median. RESULTS: Our data showed that high PFKFB3/4 expression significantly correlates with shorter overall survival in several cancers. Moreover, we found that neuroblastoma patients with poor overall survival and evidence free survival are characterized by high PFKFB3 and at the same time low PFKFB4 expression, whereas patients with high PFKFB4 expressions are characterized by significantly better overall survival/evidence free survival rates. CONCLUSION: Our analysis clearly indicates that expression of PFKFB3/4 isoenzymes may have a key prognostic value for several cancers. What's more, it seems that in neuroblastoma the prognostic value of PFK-II may be dependent on the relation between PFKFB3 and PFKFB4 isoenzyme expression, indicating that further studies analyzing the role of both cancer specific PFK-II isoenzymes are highly desired.

6.
Food Res Int ; 125: 108563, 2019 11.
Artigo em Inglês | MEDLINE | ID: mdl-31554073

RESUMO

The properties of durian fruit at five stages of ripeness were evaluated and compared. The physicochemical parameters such as titratable acidity (TA) and total soluble solids (TSS) increased, whereas the pH slightly decreased during the ripening process. The highest contents of polyphenols, flavonoids, flavanols, tannins, vitamin C and the antioxidant capacities, measured by radical scavenging assays, were found in ripe and overripe fruits. The structural properties of extracted polyphenols were evaluated by Fourier transform infrared spectroscopy (FTIR) and fluorescence spectroscopy. The interaction of polyphenols with the main drug carrier in blood human serum albumin (HSA) showed decrease in its fluorescence intensity. The binding properties of polyphenols were in direct correlation with the antioxidant capacities of the investigated fruits. HepG2 cells evaluated cytotoxic effect and the mechanism of cell death after treatment with durian. The metabolism of carbohydrates was examined on the expression of glycolysis-related genes (hexokinase 2 (HK2); 6-phosphofructo-2-kinase 4 (PFKFB4); facilitated glucose transporter member 1 (SLC2A1 (Glut1)) and lactate dehydrogenase A and utilization of glucose in the hepatocytes with durian treatment. Durian in immature stage had stronger cytotoxic effect and weak proapoptotic potential on HepG2 cells than the mature and overripe ones. The ripe and overripe fruits increased the expression of hepatic HK2 and PFKFB4 glycolytic genes and stimulated glucose utilization in HepG2 cells. The present results indicate that durians reveal different biological activity and may provide their broad and extensive use as medicinal or functional foods.


Assuntos
Antioxidantes/metabolismo , Bombacaceae/genética , Bombacaceae/metabolismo , Frutas/metabolismo , Expressão Gênica/genética , Glicólise/genética , Apoptose/genética , Frutas/genética , Células Hep G2 , Humanos , Espectrometria de Fluorescência , Espectroscopia de Infravermelho com Transformada de Fourier
7.
Anticancer Res ; 39(5): 2395-2403, 2019 May.
Artigo em Inglês | MEDLINE | ID: mdl-31092432

RESUMO

BACKGROUND/AIM: During cancer progression cells undergo epithelial-to-mesenchymal transition (EMT). Although EMT is a complex process, recently, it has been reported that CD146 overexpression in prostate cancer cells is sufficient to induce mesenchymal phenotype. The following study aimed to investigate whether the expression of CD146 is altered by an epigenetic modifier in prostate cancer cells, in vitro. MATERIALS AND METHODS: Three human prostate cancer cell lines were treated with 5-aza-2-deoxycytidine; the expression of CD146 and EMT-related factors was analyzed by RT-PCR and western Blot. The methylation status of the CD146 promoter area was assessed using bisulfite sequencing. RESULTS: Our data showed that, the expression of CD146 was evidently increased in all three studied cell lines in response to a demethylating agent, both at the mRNA and protein level, suggesting epigenetic regulation of the analyzed gene. However, there was no methylation in the studied CpG island in CD146 gene promoter. Moreover, the demethylating agent induced the expression of EMT-related transcription factors (SNAI1, SNAI2, TWIST1 and ZEB1), the pattern of which differed among the cell lines, as well as alterations in cell morphology; altogether accounting for the mesenchymal phenotype. CONCLUSION: The demethylating agent 5-aza-2-deoxycytidine triggers the expression of CD146 in prostate cancer cells independently on the methylation status of the analyzed CpG island fragment in CD146 gene promoter. Moreover, demethylation treatment induces a mesenchymal profile in prostate cancer cells.


Assuntos
Metilação de DNA/genética , Decitabina/farmacologia , Neoplasias da Próstata/genética , Antígeno CD146/genética , Linhagem Celular Tumoral , Metilação de DNA/efeitos dos fármacos , Epigênese Genética/efeitos dos fármacos , Transição Epitelial-Mesenquimal/efeitos dos fármacos , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Humanos , Masculino , Regiões Promotoras Genéticas/efeitos dos fármacos , Regiões Promotoras Genéticas/genética , Próstata/metabolismo , Próstata/patologia , Neoplasias da Próstata/tratamento farmacológico , Neoplasias da Próstata/patologia
8.
Anticancer Res ; 38(12): 6745-6752, 2018 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-30504385

RESUMO

BACKGROUND/AIM: Most melanomas develop in hypoxic conditions. Since hypoxia via HIF-1 induces glycolysis, a process essential for malignant melanoma growth/survival, the goal of this study was to analyze the influence of hypoxia on the expression of HIF-1 target genes involved in glucose metabolism. MATERIALS AND METHODS: The response of melanoma cell lines to hypoxic conditions was analyzed by RT-PCR and western blotting. A Kaplan-Meier survival analysis for patients with high and low expression level of PFKFB4 was performed. Further analysis of patients' data was performed using the R/Bioconductor environment. RESULTS: Induction of PFKFB4 gene expression can be considered a crucial mechanism behind glycolysis enhancement in hypoxic melanoma cells. Analysis of a publicly available database revealed that high PFKFB4 expression contributes to poor prognosis of melanoma patients. CONCLUSION: Currently available anti-melanoma therapeutic strategies may significantly benefit from agents targeting PFKFB4 activity.


Assuntos
Melanoma/genética , Melanoma/patologia , Oxigênio/farmacologia , Fosfofrutoquinase-2/genética , Neoplasias Cutâneas/genética , Neoplasias Cutâneas/patologia , Hipóxia Celular/fisiologia , Linhagem Celular Tumoral , Progressão da Doença , Relação Dose-Resposta a Droga , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Humanos , Melanoma/metabolismo , Melanoma/mortalidade , Análise em Microsséries , Metástase Neoplásica , Fosfofrutoquinase-2/fisiologia , Neoplasias Cutâneas/metabolismo , Neoplasias Cutâneas/mortalidade , Análise de Sobrevida , Regulação para Cima/efeitos dos fármacos , Regulação para Cima/genética , Melanoma Maligno Cutâneo
9.
Acta Biochim Pol ; 65(1): 101-109, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29543923

RESUMO

Multiple myeloma (MM) is characterized as a clonal expansion of malignant plasma cells in the bone marrow, which is often associated with pancytopenia and osteolytic bone disease. Interestingly, myeloma-infiltrated bone marrow is considered to be hypoxic, providing selection pressure for a developing tumour. Since HSP90 was shown to participate in stabilization of the subunit of the key transcription factor HIF-1, which controls the hypoxic response, the aim of this study was to investigate the influence of a HSP90 inhibitor 17-allylamino-17-demethoxygeldanamycin (17-AAG), on MM cells cultured under low oxygenation conditions. We confirmed that 17-AAG inhibits hypoxic induction of the HIF-1 target genes in malignant plasma cells and demonstrate the concentration range of severe hypoxia-specific cytotoxicity. Next, we selected the malignant plasma cells under severe hypoxia/re-oxygenation culture conditions in the presence or absence of 17-AAG and subsequently, the cells which survived were further expanded and analyzed. Interestingly, we have noticed significant changes in the survival and the response to anti-MM drugs between the parental cell lines and those selected in cyclic severe hypoxia in the presence and absence of 17-AAG. Importantly, we also observed that the lack of oxygen itself, irrespectively of HIF-1 inhibition, is the main/pivotal factor driving the selection process in the experiments presented here.


Assuntos
Benzoquinonas/uso terapêutico , Proteínas de Choque Térmico HSP90/antagonistas & inibidores , Lactamas Macrocíclicas/uso terapêutico , Mieloma Múltiplo/patologia , Plasmócitos/patologia , Hipóxia Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Sobrevivência Celular , Humanos , Subunidade alfa do Fator 1 Induzível por Hipóxia/genética , Mieloma Múltiplo/tratamento farmacológico , Plasmócitos/efeitos dos fármacos
10.
Chemistry ; 23(22): 5307-5316, 2017 Apr 19.
Artigo em Inglês | MEDLINE | ID: mdl-28230285

RESUMO

By reducing the surface tension of the air-water interface in alveoli, lung surfactant (LS) is crucial for proper functioning of the lungs. It also forms the first barrier against inhaled pathogens. In this study we inspect the interactions of LS models with a dangerous air pollutant, benzo[a]pyrene (BaP). Dipalmitoylphosphatidylcholine (DPPC), 1-palmitoyl-2-oleoylphosphatidylcholine, and their 1:1 mixture are used as LS models. Pressure-area isotherms are employed to study macroscopic properties of the monolayers. We find that addition of BaP has a condensing effect, manifested by lowering the values of surface pressure and shifting the isotherms to smaller areas. Atomistic details of this process are examined by means of molecular dynamics simulations. We show that initially BaP molecules are accumulated in the monolayers. Upon compression, they are forced to the headgroups region and eventually expelled to the subphase. BaP presence results in reduction of monolayer hydration in the hydrophilic region. In the hydrophobic region it induces increased chain ordering, reduction of monolayer fluidity, and advances transition to the liquid condensed phase in the DPPC system.


Assuntos
1,2-Dipalmitoilfosfatidilcolina/análogos & derivados , Benzo(a)pireno/química , Fosfatidilcolinas/química , Alvéolos Pulmonares/fisiologia , 1,2-Dipalmitoilfosfatidilcolina/química , 1,2-Dipalmitoilfosfatidilcolina/farmacologia , Simulação de Dinâmica Molecular , Alvéolos Pulmonares/química , Surfactantes Pulmonares , Tensão Superficial
11.
J Phys Chem B ; 119(22): 6668-79, 2015 Jun 04.
Artigo em Inglês | MEDLINE | ID: mdl-25959677

RESUMO

The interaction between five gemini amphiphilic pseudopeptides (GAPs) differing by the length of the central spacer and a model membrane lipid, 1,3-bis[1,2-dimyristoyl-sn-glycero-3-phospho]-sn-glycerol (cardiolipin) were studied with the aim to evaluate their possible antimicrobial properties. To this end, monomolecular films were formed at the air/water interface with pure cardiolipin or cardiolipin/GAPs mixtures; film properties were determined using surface pressure and surface potential measurements, as well as polarization-modulation infrared reflection-absorption spectroscopy. Moreover, to better understand the GAPs-phospholipid interaction at the molecular level, molecular dynamics simulations were performed. The results obtained indicate that the length of the central spacer has an effect on the interaction of GAPs with cardiolipin and on the properties of the lipid film. The GAPs with the longer linkers can be expected to be useful for biological membrane modification and for possible antimicrobial applications.


Assuntos
Cardiolipinas/química , Membrana Celular/química , Interações Hidrofóbicas e Hidrofílicas , Peptídeos/química , Cardiolipinas/metabolismo , Membrana Celular/metabolismo , Conformação Molecular , Simulação de Dinâmica Molecular , Peptídeos/metabolismo , Relação Estrutura-Atividade , Termodinâmica
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA