Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
1.
Front Immunol ; 14: 1162213, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37063901

RESUMO

Background: Selective cancer cell recognition is the most challenging objective in the targeted delivery of anti-cancer agents. Extruded specific cancer cell membrane coated nanoparticles, exploiting the potential of homotypic binding along with certain protein-receptor interactions, have recently proven to be the method of choice for targeted delivery of anti-cancer drugs. Prediction of the selective targeting efficiency of the cancer cell membrane encapsulated nanoparticles (CCMEN) is the most critical aspect in selecting this strategy as a method of delivery. Materials and methods: A probabilistic model based on binding scores and differential expression levels of Glioblastoma cancer cells (GCC) membrane proteins (factors and receptors) was implemented on python 3.9.1. Conditional binding efficiency (CBE) was derived for each combination of protein involved in the interactions. Selective propensities and Odds ratios in favour of cancer cells interactions were determined for all the possible combination of surface proteins for 'k' degree of interaction. The model was experimentally validated by two types of Test cultures. Results: Several Glioblastoma cell surface antigens were identified from literature and databases. Those were screened based on the relevance, availability of expression levels and crystal structure in public databases. High priority eleven surface antigens were selected for probabilistic modelling. A new term, Break-even point (BEP) was defined as a characteristic of the typical cancer cell membrane encapsulated delivery agents. The model predictions lie within ±7% of the experimentally observed values for both experimental test culture types. Conclusion: The implemented probabilistic model efficiently predicted the directional preference of the exposed nanoparticle coated with cancer cell membrane (in this case GCC membrane). This model, however, is developed and validated for glioblastoma, can be easily tailored for any type of cancer involving CCMEN as delivery agents for potential cancer immunotherapy. This probabilistic model would help in the development of future cancer immunotherapeutic with greater specificity.


Assuntos
Antineoplásicos , Glioblastoma , Nanopartículas , Humanos , Glioblastoma/tratamento farmacológico , Glioblastoma/metabolismo , Membrana Celular/metabolismo , Antineoplásicos/uso terapêutico , Membranas/metabolismo , Nanopartículas/química
2.
Front Oncol ; 11: 706586, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34322393

RESUMO

Advanced stage cancers are aggressive and difficult to treat with mono-therapeutics, substantially decreasing patient survival rates. Hence, there is an urgent need to develop unique therapeutic approaches to treat cancer with superior potency and efficacy. This study investigates a new approach to develop a potent combinational therapy to treat advanced stage leukemia. Biologically active α-amino amide analogs (RS)-N-(2-(cyclohexylamino)-2-oxo-1-phenylethyl)-N-phenylpropiolamide (α-AAA-A) and (RS)-N-(2-(cyclohexylamino)-2-oxo-1-phenylethyl)-N-phenylbut2-enamide (α-AAA-B) were synthesized using linear Ugi multicomponent reaction. Cytotoxicities and IC50 values of α-AAA-A and α-AAA-B against leukemia cancer cell lines (HL-60 and K562) were analyzed though MTT assay. Cytotoxic assay analyzed percent killing of leukemia cell lines due to the effect of γδ T cells alone or in combination with α-AAA-A or α-AAA-B. Synthesized biologically active molecule α-AAA-A exhibited increased cytotoxicity of HL-60 (54%) and K562 (44%) compared with α-AAA-B (44% and 36% respectively). Similarly, α-AAA-A showed low IC50 values for HL-60 (1.61 ± 0.11 µM) and K562 (3.01 ± 0.14 µM) compared to α-AAA-B (3.12 ± 0.15 µM and 6.21 ± 0.17 µM respectively). Additive effect of amide analogs and γδ T cells showed significantly high leukemia cancer cell killing as compared to γδ T cells alone. A unique combinational therapy with γδ T cells and biologically active anti-cancer molecules (α-AAA-A/B), concomitantly may be a promising cancer therapy.

3.
Bioinformation ; 17(3): 460-469, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34092966

RESUMO

Human Vg9/Vδ2 T cells (γδ T cells) are immune surveillance cells both in innate and adaptive immunity and are a possible target for anticancer therapies, which can induce immune responses in a variety of cancers. Small non-peptide antigens such as zoledronate can do activation and expansion of T cells in vitro. It is evident that for adoptive cancer therapies, large numbers of functional cells are needed into cancer patients. Hence, optimization of methods needs to be carried out for the efficient expansion of these T cells. Standardization of peripheral blood mononuclear cells (PBMCs) isolation was devised. Cytokines (interleukin 2 (IL-2) and interleukin 15 (IL-15)) and zoledronate were also standardized for different concentrations. It was found that an increased number of PBMCs were recovered when washing was done at 1100 revolution per minute (rpm). Significantly high expansion fold was (2524 ± 787 expansion fold) achieved when stimulation of PBMCs was done with 1 µM of zoledronate and both cytokines IL-2 and IL-15 supported the expansion and survival of cells at the concentrations of 100 IU/ml and 10 ng/ml respectively. 14-day cultures showed highly pure (91.6 ± 5.1%) and live (96.5 ± 2.5%) expanded γδ T cells. This study aimed to standardize an easy to manipulate technique for the expansion of γδ T cells, giving a higher yield.

4.
Molecules ; 26(8)2021 Apr 14.
Artigo em Inglês | MEDLINE | ID: mdl-33919694

RESUMO

Glioblastoma (GB) is an aggressive cancer with high microvascular proliferation, resulting in accelerated invasion and diffused infiltration into the surrounding brain tissues with very low survival rates. Treatment options are often multimodal, such as surgical resection with concurrent radiotherapy and chemotherapy. The development of resistance of tumor cells to radiation in the areas of hypoxia decreases the efficiency of such treatments. Additionally, the difficulty of ensuring drugs effectively cross the natural blood-brain barrier (BBB) substantially reduces treatment efficiency. These conditions concomitantly limit the efficacy of standard chemotherapeutic agents available for GB. Indeed, there is an urgent need of a multifunctional drug vehicle system that has potential to transport anticancer drugs efficiently to the target and can successfully cross the BBB. In this review, we summarize some nanoparticle (NP)-based therapeutics attached to GB cells with antigens and membrane receptors for site-directed drug targeting. Such multicore drug delivery systems are potentially biodegradable, site-directed, nontoxic to normal cells and offer long-lasting therapeutic effects against brain cancer. These models could have better therapeutic potential for GB as well as efficient drug delivery reaching the tumor milieu. The goal of this article is to provide key considerations and a better understanding of the development of nanotherapeutics with good targetability and better tolerability in the fight against GB.


Assuntos
Neoplasias Encefálicas/tratamento farmacológico , Sistemas de Liberação de Medicamentos , Glioblastoma/tratamento farmacológico , Nanopartículas Multifuncionais/uso terapêutico , Animais , Membrana Celular/química , Humanos , Nanotecnologia
5.
Curr Drug Targets ; 22(11): 1198-1206, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33588726

RESUMO

Over the past several years, remarkable progress towards the recognition of new therapeutic targets in tumor cells has led to the discovery and development of newer scaffolds of anti-tumor drugs. The exploration and exploitation of epigenetic regulation in tumor cells are of immense importance to both the pharmaceutical and academic biomedical literatures. Epigenetic mechanisms are indispensable for the normal development and maintenance of tissue-specific gene expression. Disruption of epigenetic processes to eradicate tumor cells is among the most promising intervention for cancer control. Polycomb repressive complex 2 (PRC2), a complex that methylates lysine 27 of histone H3 to promote transcriptional silencing, is involved in orchestrating significant pathways in a cell. Overexpression of PRC2 has been found in a number of cancerous malignancies, making it a major target for anti-cancer therapy. Despite its well-understood molecular mechanism, hyperactivation and drug resistance mutations in its subunits have become a matter of discussion. This review outlines the current understanding of the components of PRC2 in active complex formation and assesses their potential as a promising therapeutic target for cancer therapy. We also review the effects of mutations in the PRC2 components, in the purview of human cancers. Finally, we discuss some of the current challenges for therapeutic drug designs targeting the PRC2 complex.


Assuntos
Epigênese Genética , Neoplasias , Complexo Repressor Polycomb 2/antagonistas & inibidores , Desenho de Fármacos , Histonas/metabolismo , Humanos , Neoplasias/tratamento farmacológico , Neoplasias/genética , Complexo Repressor Polycomb 2/genética
6.
Semin Hematol ; 58(1): 45-55, 2021 01.
Artigo em Inglês | MEDLINE | ID: mdl-33509443

RESUMO

The DNA methyltransferase inhibitor azacytidine (aza) may reactivate pathways associated with plasma cell differentiation, cell cycle control, apoptosis, and immune recognition and thereby restore sensitivity to lenalidomide (len) and dexamethasone (dex) in relapsed and/or refractory multiple myeloma (RRMM). We aimed to develop an aza regimen that reaches epigenetically active levels 8 times in 28 days with less bone marrow toxicity than the myeloid malignancy standard of 7 consecutive doses to enable safe combination with len. Aza was escalated from 30 mg/m2 once a week up to a predefined maximum of 50 mg/m2 twice a week in combination with GFR-adjusted len (≥ 60 mL/min: 25 mg, 3059 mL/min: 10 mg) day 1 to 21 every 28 days and dex 40 mg once a week followed by a limited expansion study to a total N of 23 at the highest tolerated dose. Fifty-one patients (pts) with RRMM were screened, 42 were treated and 41 were evaluable for response based on at least 1 response assessment or progression after treatment start. The median number of prior lines of therapy was 5 (1-11) and 81% (34) were refractory to len and/or pomalidomide (pom). Two DLTs occurred in different cohorts, 1 neutropenic fever in 1/6 pts on the aza 40 mg/m2 twice a week GFR ≥ 60 mL/min cohort and 1 GGT elevation in 1/6 pts on the aza 50 mg/m2 GFR 30-59 mL/min cohort. An MTD was not reached and aza 50 mg/m2 SC twice a week was chosen for the expansion study. At least possibly related Grade 3/4 AEs occurred in 28 pts (67%) with the following in > 1 pt: neutropenia (N = 16, 38%), anemia (N = 6, 14%), lymphopenia (N = 5, 12%), thrombocytopenia (N = 4, 10%), leukopenia (N = 4, 10%), febrile neutropenia (N = 4, 10%), fatigue (N = 3, 7%), fever (N = 2, 5%), and infection (N = 2, 5%). At a median follow up time for alive pts of 60.2 months (range: 36.1-82.5 months), the overall response rate (≥ partial response) and clinical benefit response rate (≥ minor response) was 22 and 32%, respectively, with 4 very good partial responses (10%), 5 partial responses (12%), and 4 minor responses (10%). The median PFS was 3.1 months (95% confidence interval [CI]: 2.1-5.1 months), median TTP 2.7 months (95% CI: 2.1-7.5 months), and median OS 18.6 months (95% CI: 12.9-33.0 months). Achieving at least minor response and reaching TTP > 6 months was associated with approximately 35% lower median plasma levels of the enzyme that inactivates aza, plasma cytidine deaminase (CDA, P< .0001). Two of the len refractory pts achieved longer disease control than with any prior regimen and 1 responded immediately after progression on len, bortezomib, and prednisone. Analyses of the methylation state of over 480,000 CpG sites in purified myeloma cells at screening were possible in 11 pts and on day 28 in 8 of them. As in other studies, the majority of differentially methylated CpGs compared to normal plasma cells were hypomethylated in myeloma. Treatment decreased the number of CpGs that were differentially methylated in normal plasma cells by > 0.5% in 6 and by > 5% in 3 of the 8 pts, most pronounced in 2 pts with clinically convincing aza contribution who achieved a reduction in overall differentially methylated CpGs by 23 and 68%, respectively, associated with increased expression of immunoglobulin genes. The study demonstrated tolerability of twice a week SC aza at 50 mg/m2 with len and dex in RRMM and suggested aza may help overcome the len/pom refractory state, possibly by activating differentiation pathways. Relatively low response rates and association of clinical benefit with low plasma levels of the aza inactivating enzyme CDA suggest the aza regimen will need to be optimized further and pt selection may be required to maximize benefit.


Assuntos
Mieloma Múltiplo , Protocolos de Quimioterapia Combinada Antineoplásica/efeitos adversos , Azacitidina/efeitos adversos , Metilação de DNA , Dexametasona/efeitos adversos , Humanos , Lenalidomida/farmacologia , Lenalidomida/uso terapêutico , Mieloma Múltiplo/tratamento farmacológico , Mieloma Múltiplo/genética , Resultado do Tratamento
7.
Bioinformation ; 17(7): 673-679, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-35283582

RESUMO

Mono-therapeutics is rarely effective as a treatment option, which limits the survival of patients in advanced grade aggressive cancers. Combinational therapeutics (multiple drugs for multiple targets) to combat cancer is gaining momentum in recent years. Hence, it is of interest to document known data for combinational therapeutics in cancer treatment. An amalgamation of therapeutic agents enhances the efficacy and potency of the therapy. Combinational therapy can potentially target multiple pathways that are necessary for the cancer cells to proliferate, and/or target molecules, which may help cancer to become more aggressive and metastasize. In this review, we discuss combinational therapeutics, which include human γδ T cells in combinations with biologically active anti-cancer molecules, which synergistically may produce promising combinational therapeutics.

8.
Drug Discov Today ; 24(1): 179-188, 2019 01.
Artigo em Inglês | MEDLINE | ID: mdl-30031878

RESUMO

Polycomb repressive complex 2 (PRC2) plays a significant part in histone methylation - trimethylating K27 at H3, an epigenetic hallmark of gene silencing. Inhibition of PRC2 has been reported as a promising strategy for the treatment of various cancers. Significant efforts have been made toward the development of PRC2 inhibitors and some of them have progressed to clinical trials. The binding mode of these inhibitors is well understood. Here, we summarize the advances in drug discovery and development for PRC2 component inhibitors by focusing on their chemotypes, activity, selectivity and binding modes. We believe that such analysis will provide new avenues for the design and development of next-generation PRC2 inhibitors through establishment of a structure-based drug design platform.


Assuntos
Complexo Repressor Polycomb 2/antagonistas & inibidores , Animais , Epigênese Genética , Humanos , Neoplasias/tratamento farmacológico , Neoplasias/genética , Neoplasias/metabolismo , Complexo Repressor Polycomb 2/química , Complexo Repressor Polycomb 2/metabolismo
9.
PLoS One ; 12(2): e0171267, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28152102

RESUMO

Fasciolosis an economically important global disease of ruminants in the temperate and tropical regions, caused by Fasciola hepatica and F. gigantica, respectively, also poses a potential zoonotic threat. In India alone it causes huge losses to stakeholders. Anthelmintics including triclabendazole have been used to control this menace but the emerging resistance against the available compounds necessitates identification of novel and alternative therapeutic measures involving plant derived natural compounds for their anthelmintic potential. Thymoquinone (T) and curcumin (C), the active ingredients of Nigella sativa and Curcuma longa respectively have been used as antiparasitic agents but the information on their flukicidal effect is very limited. Adult flukes of F. gigantica were in vitro exposed to different concentrations of thymoquinone and curcumin separately for 3h at 37+ 1°C. A significant (p<0.05) reduction in the worm motility at 60 µM concentration of both T and C was observed though all the worms remained alive after 3h exposure, whereas the effect on egg shedding was statistically insignificant. Pronounced tegumental disruptions and erosion of spines in the posterior region and around the acetabulum was evident. A significant (p<0.05) decrease in glutathione-S-transferase and superoxide dismutase activity and reduced glutathione (GSH) level was observed, while protein carbonylation increased differentially. A significant inhibition of CathepsinL (CatL) gene expression in thymoquinone treated worms was also evident. Further, in silico molecular docking of T and C with CatL revealed a stronger interaction of curcumin with the involvement of higher number of amino acids as compared to thymoquinone that could be more effective in inhibiting the antioxidant enzymes of F. gigantica. It is concluded that both the compounds understudy will decrease the detoxification ability of F. gigantica, while inhibition of CatL will significantly affect their virulence potential. Thus, both thymoquinone and curcumin appeared to be promising anthelmintic compounds for further investigations.


Assuntos
Antiplatelmínticos/farmacologia , Benzoquinonas/farmacologia , Curcumina/farmacologia , Fasciola/efeitos dos fármacos , Animais , Relação Dose-Resposta a Droga , Glutationa/metabolismo , Glutationa Transferase/metabolismo , Testes de Sensibilidade Parasitária
10.
J Immunol ; 190(6): 2966-75, 2013 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-23408834

RESUMO

Epigenetic changes play important roles in carcinogenesis and influence initial steps in neoplastic transformation by altering genome stability and regulating gene expression. To characterize epigenomic changes during the transformation of normal plasma cells to myeloma, we modified the HpaII tiny fragment enrichment by ligation-mediated PCR assay to work with small numbers of purified primary marrow plasma cells. The nano-HpaII tiny fragment enrichment by ligation-mediated PCR assay was used to analyze the methylome of CD138(+) cells from 56 subjects representing premalignant (monoclonal gammopathy of uncertain significance), early, and advanced stages of myeloma, as well as healthy controls. Plasma cells from premalignant and early stages of myeloma were characterized by striking, widespread hypomethylation. Gene-specific hypermethylation was seen to occur in the advanced stages, and cell lines representative of relapsed cases were found to be sensitive to decitabine. Aberrant demethylation in monoclonal gammopathy of uncertain significance occurred primarily in CpG islands, whereas differentially methylated loci in cases of myeloma occurred predominantly outside of CpG islands and affected distinct sets of gene pathways, demonstrating qualitative epigenetic differences between premalignant and malignant stages. Examination of the methylation machinery revealed that the methyltransferase, DNMT3A, was aberrantly hypermethylated and underexpressed, but not mutated in myeloma. DNMT3A underexpression was also associated with adverse overall survival in a large cohort of patients, providing insights into genesis of hypomethylation in myeloma. These results demonstrate widespread, stage-specific epigenetic changes during myelomagenesis and suggest that early demethylation can be a potential contributor to genome instability seen in myeloma. We also identify DNMT3A expression as a novel prognostic biomarker and suggest that relapsed cases can be therapeutically targeted by hypomethylating agents.


Assuntos
Transformação Celular Neoplásica/imunologia , Metilação de DNA/genética , Metilação de DNA/imunologia , Mieloma Múltiplo/genética , Mieloma Múltiplo/imunologia , Transformação Celular Neoplásica/genética , Estudos de Coortes , Diagnóstico Precoce , Regulação Neoplásica da Expressão Gênica/imunologia , Humanos , Mieloma Múltiplo/patologia , Estadiamento de Neoplasias , Reação em Cadeia da Polimerase , Recidiva , Indução de Remissão , Reprodutibilidade dos Testes , Sindecana-1/biossíntese , Sindecana-1/genética , Células Tumorais Cultivadas
11.
PLoS One ; 7(12): e51850, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-23251640

RESUMO

Epirubicin (EPI), an anthracycline antitumour antibiotic, is a known intercalating and DNA damaging agent. Here, we study the molecular interaction of EPI with histones and other cellular targets. EPI binding with histone core protein was predicted with spectroscopic and computational techniques. The molecular distance r, between donor (histone H3) and acceptor (EPI) was estimated using Förster's theory of non-radiation energy transfer and the detailed binding phenomenon is expounded. Interestingly, the concentration dependent reduction in the acetylated states of histone H3 K9/K14 was observed suggesting more repressed chromatin state on EPI treatment. Its binding site near N-terminal lysines is further characterized by thermodynamic determinants and molecular docking studies. Specific DNA binding and inhibition of transcription factor (Tf)-DNA complex formation implicates EPI induced transcriptional inhibition. EPI also showed significant cell cycle arrest in drug treated cells. Chromatin fragmentation and loss of membrane integrity in EPI treated cells is suggestive of their commitment to cell death. This study provides an analysis of nucleosome dynamics during EPI treatment and provides a novel insight into its action.


Assuntos
Epirubicina/farmacologia , Homeostase/efeitos dos fármacos , Lisina/metabolismo , Fatores de Transcrição/metabolismo , Acetilação , Sítios de Ligação/efeitos dos fármacos , Sítios de Ligação/genética , Pontos de Checagem do Ciclo Celular/efeitos dos fármacos , Pontos de Checagem do Ciclo Celular/genética , Morte Celular/efeitos dos fármacos , Morte Celular/genética , Linhagem Celular , Permeabilidade da Membrana Celular/efeitos dos fármacos , Permeabilidade da Membrana Celular/genética , Cromatina/metabolismo , DNA/genética , Células HEK293 , Histonas/genética , Histonas/metabolismo , Homeostase/genética , Humanos , Lisina/genética , Nucleossomos/efeitos dos fármacos , Nucleossomos/genética , Nucleossomos/metabolismo , Saccharomyces cerevisiae , Termodinâmica , Fatores de Transcrição/genética , Transcrição Gênica/efeitos dos fármacos , Transcrição Gênica/genética
12.
PLoS One ; 7(9): e44690, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22984542

RESUMO

Current methods to study chromatin configuration are not well suited for high throughput drug screening since they require large cell numbers and multiple experimental steps that include centrifugation for isolation of nuclei or DNA. Here we show that site specific chromatin analysis can be achieved in one step by simply performing direct chromatin PCR (DC-PCR) on cells. The basic underlying observation was that standard hypotonic PCR buffers prevent global cellular chromatin solubilization during thermal cycling while more loosely organized chromatin can be amplified. Despite repeated heating to >90 °C, 41 of 61 tested 5' sequences of silenced genes (CDKN2A, PU.1, IRF4, FOSB, CD34) were not amplifiable while 47 could be amplified from expressing cells. Two gene regions (IRF4, FOSB) even required pre-heating of cells in isotonic media to allow this differentiation; otherwise none of 19 assayed sequences yielded PCR products. Cells with baseline expression or epigenetic reactivation gave similar DC-PCR results. Silencing during differentiation of CD34 positive cord blood cells closed respective chromatin while treatment of myeloma cells with an IRF4 transcriptional inhibitor opened a site to DC-PCR that was occupied by RNA polymerase II and NFκB as determined by ChIP. Translation into real-time PCR can not be achieved with commercial real-time PCR buffers which potently open chromatin, but even with simple ethidium bromide addition to standard PCR mastermix we were able to identify hits in small molecules screens that suppressed IRF4 expression or reactivated CDKN2A in myeloma cells using densitometry or visual inspection of PCR plates under UV light. While need in drug development inspired this work, application to genome-wide analysis appears feasible using phi29 for selective amplification of open cellular chromatin followed by library construction from supernatants since such supernatants yielded similar results as gene specific DC-PCR.


Assuntos
Cromatina/genética , Reação em Cadeia da Polimerase/métodos , Antígenos CD34/biossíntese , Antígenos CD34/química , Diferenciação Celular , Linhagem Celular Tumoral , Cromatina/química , Cromatina/metabolismo , Imunoprecipitação da Cromatina , Avaliação Pré-Clínica de Medicamentos/métodos , Sangue Fetal/metabolismo , Citometria de Fluxo/métodos , Perfilação da Expressão Gênica , Inativação Gênica , Temperatura Alta , Humanos , Fatores Reguladores de Interferon/metabolismo , NF-kappa B/metabolismo , Proteínas Proto-Oncogênicas c-myc/metabolismo , RNA Polimerase II/metabolismo , Reação em Cadeia da Polimerase em Tempo Real/métodos , Temperatura
13.
Cell Biochem Biophys ; 60(3): 209-18, 2011 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-21136304

RESUMO

Mitoxantrone (MTX), a choice of drug in cancer chemotherapeutic regime, is a potent and less toxic among anthracycline class of drugs. Here, we study the molecular interaction of MTX, with histone and its acetylation dynamics. Its binding with histone core protein was predicted with CD and UV-visible spectroscopic techniques. The MTX-protein complex resulted in the impediment of the histone acetyltransferase (HAT) activity in a dose dependent manner on MTX binding. Interestingly, the concentration dependent reduction in acetylated state of specific lysines K9/K14 was also observed on MTX treatment in vivo. The molecular distance r, between donor (histone H3) and acceptor (MTX) was estimated using Förster's theory of non-radiation energy transfer and the detailed binding phenomenon was expounded. MTX binding site near N-terminal lysines is characterized with an association constant of the order of 10(4). The positive thermodynamic values of both ∆H° and ∆S° were suggestive that the hydrophobic interactions dominate in MTX-protein binding. The binding site allocation predicted by computational modeling placed the drug molecule near N-terminal lysine K9 and K14 of histone H3, and corroborate with the thermodynamic interaction model. The study establishes that MTX-histone interaction affects protein acetylation state and also provided a mechanistic model for its binding. Hence, MTX interaction may affect chromatin structure and implicates its role in transcriptional regulation at epigenetic level.


Assuntos
Antineoplásicos/química , Histonas/química , Mitoxantrona/química , Acetilação , Antineoplásicos/farmacologia , Sítios de Ligação , Linhagem Celular , Cromatina/química , Cromatina/metabolismo , Dicroísmo Circular , Transferência Ressonante de Energia de Fluorescência , Histona Acetiltransferases/metabolismo , Histonas/metabolismo , Humanos , Interações Hidrofóbicas e Hidrofílicas , Mitoxantrona/farmacologia , Espectrofotometria Ultravioleta , Termodinâmica
14.
Biosci Rep ; 30(5): 331-40, 2010 Apr 21.
Artigo em Inglês | MEDLINE | ID: mdl-19691448

RESUMO

MTX (mitoxantrone) is perhaps the most promising drug used in the treatment of various malignancies. Comprehensive literature on the therapeutics has indicated it to be the least toxic in its class, although its mechanism of action is still not well defined. In the present study, we have evaluated the associated binding interactions of MTX with naked DNA. The mechanism of MTX binding with DNA was elucidated by steady-state fluorescence and a static-type quenching mechanism is suggested for this interaction. Thermodynamic parameters from van 't Hoff plots showed that the interaction of these drugs with DNA is an entropically driven phenomenon. The binding mode was expounded by attenuance measurements and competitive binding of a known intercalator. Sequence specificity of these drug-DNA complexes was analysed by FTIR (Fourier-transform infrared) spectroscopy and molecular modelling studies. CD spectroscopy and the plasmid nicking assay showed that the binding of this drug with DNA results in structural and conformational perturbations. EMSA (electrophoretic mobility-shift assay) results showed that these drug-DNA complexes prevent the binding of octamer TF (transcription factor) to DNA. In summary, the study implicates MTX-induced conformational instability and transcription inhibition on DNA binding.


Assuntos
Antineoplásicos , DNA , Mitoxantrona , Fatores de Transcrição , Animais , Antineoplásicos/química , Antineoplásicos/metabolismo , Bovinos , Linhagem Celular , Dicroísmo Circular , DNA/química , DNA/metabolismo , Humanos , Mitoxantrona/química , Mitoxantrona/metabolismo , Modelos Moleculares , Estrutura Molecular , Espectroscopia de Infravermelho com Transformada de Fourier , Termodinâmica , Fatores de Transcrição/química , Fatores de Transcrição/metabolismo
15.
Biosci Rep ; 30(6): 375-81, 2010 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-19951261

RESUMO

MTX (mitoxantrone), an anti-tumour antibiotic, is known to cause cell death by intercalating the DNA bases. But how it interferes with the cellular proliferation is not well known. Hence, in the present study, we have tried to evaluate the interaction of this drug using proliferation dynamics to gain a better understanding of MTX's antineoplastic action. Inhibition of proliferation by these drugs was detected by evaluating its effect on cell proliferation and growth curve of the cells. MTX was also found to affect the cell viability and, thereby, cell physiology. Typical apoptotic morphologies such as condensation of nuclei and membrane permeabilization were observed through CLSM (confocal laser scanning microscopy) and fluorescence spectroscopy, which implicates commitment to cell death. Cell-cycle distribution was measured by flow cytometric measurements. The analysis demonstrated significant cell-cycle arrest on MTX treatment. Inhibition of lacZ gene expression was also observed on drug treatment, which implicates its interaction with gene expression.


Assuntos
Antineoplásicos/farmacologia , Ciclo Celular/efeitos dos fármacos , Mitoxantrona/farmacologia , Saccharomyces cerevisiae/efeitos dos fármacos , Saccharomyces cerevisiae/crescimento & desenvolvimento , Proliferação de Células/efeitos dos fármacos , Contagem de Colônia Microbiana , Citometria de Fluxo , Regulação Fúngica da Expressão Gênica/efeitos dos fármacos , Óperon Lac/genética , Óperon Lac/fisiologia , Microscopia Confocal , Saccharomyces cerevisiae/citologia , Saccharomyces cerevisiae/ultraestrutura
16.
J Pharm Biomed Anal ; 48(4): 1096-104, 2008 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-18842373

RESUMO

Doxorubicin (DOX) binding to hemoglobin (Hb) was studied to investigate the drug induced protein dysfunction. The features of anti-tumor drug doxorubicin infused structural perturbation of human Hb were studied by circular dichroism (CD). The mechanism of DOX-Hb binding was elucidated by steady-state and synchronous fluorescence spectroscopy. The Stern-Volmer analysis indicated that the binding of Hb to DOX is characterized by more than one high affinity binding site with the association constants of the order of 10(5). Hydrophobic probe ANS was employed to elucidate the drug binding site. Binding mode expounded by thermodynamic parameters implied the role of hydrogen bonding, electrostatic and hydrophobic interaction in stabilizing the complex. The molecular distance between donor (Hb) and acceptor (DOX) was calculated according to Förster's theory of energy transfer. Fourier transform infrared (FT-IR) spectroscopy provides an insight to the changes occurring in protein on DOX binding. Treatment of Hb with DOX resulted in a dose dependent fragmentation of protein. The quantitative analysis revealed the release of acid soluble amino groups from the photoexcited Hb-DOX mixture. The free radical mediated degradation was suggested by its rescue on mannitol and superoxide dismutase (SOD) appliance. The loss of protein band further corroborates the concentration dependent Hb fragmentation. The molecular modeling complies with the thermodynamic data of forces involved in DOX binding and depicts its interaction in the proximity of oxygen binding pocket of Hb. Thus, this study enriches our understanding of the interaction dynamics of anticancer drugs to the physiologically important protein Hb.


Assuntos
Doxorrubicina/química , Oxiemoglobinas/metabolismo , Antibióticos Antineoplásicos/química , Sítios de Ligação , Soluções Tampão , Dicroísmo Circular/métodos , Doxorrubicina/isolamento & purificação , Humanos , Ligação de Hidrogênio , Concentração de Íons de Hidrogênio , Modelos Moleculares , Estrutura Molecular , Fosfatos/química , Ligação Proteica , Espectrometria de Fluorescência/métodos , Espectroscopia de Infravermelho com Transformada de Fourier , Eletricidade Estática , Termodinâmica
17.
Eur J Pharm Sci ; 35(5): 371-82, 2008 Dec 18.
Artigo em Inglês | MEDLINE | ID: mdl-18762252

RESUMO

Mitoxantrone (MTX) is a clinically used antitumor anthracycline, which is made available to the target tissues by transport protein human serum albumin (HSA). Being less toxic unlike other member of this family, its binding characteristics are therefore of immense interest. The circular dichroism (CD), fluorescence and Fourier transform infrared (FTIR) spectroscopies were employed to elucidate the mode and the mechanism for this interaction. MTX binding is characterized by one high affinity binding site with the association constants of the order of 10(5). Correlation between stability of N-MTX (drug bound N form of HSA) and B-MTX (drug bound B form of HSA) complexes with drug distribution has been discussed. The molecular distance, r, between donor (HSA) and acceptor (MTX) was estimated according to Forster's theory of non-radiation energy transfer. The features of MTX induced structural perturbation of human serum albumin (HSA) has been studied in detail by CD and FTIR analysis. Domain I was assigned to possess high affinity binding site for MTX. Molecular docking showed that the MTX binds HSA to a non-classical drug binding site. The binding dynamics was expounded by synchronous fluorescence, thermodynamic parameters and molecular modeling, which entails that hydrophobic interactions, hydrogen bonding and electrostatic forces, stabilizes the interaction.


Assuntos
Antineoplásicos/química , Mitoxantrona/química , Albumina Sérica/química , Algoritmos , Antineoplásicos/farmacocinética , Dicroísmo Circular , Transferência de Energia , Hemina/química , Humanos , Mitoxantrona/farmacocinética , Modelos Moleculares , Ligação Proteica , Conformação Proteica , Dobramento de Proteína , Albumina Sérica/isolamento & purificação , Espectrometria de Fluorescência , Espectroscopia de Infravermelho com Transformada de Fourier , Termodinâmica , Distribuição Tecidual , Triptofano/química
18.
Bioinformation ; 2(9): 401-4, 2008 Jul 14.
Artigo em Inglês | MEDLINE | ID: mdl-18795113

RESUMO

Binding modalities of doxorubicin (DOX), a widely used antineoplastic anthracyline antibiotic with hemoglobin (Hb) have been studied. The protein and the ligand were prepared using CORINA and protonated with insight II. The best conformation was sought by employing GOLDV. Molecular modeling calculations showed that DOX binds Hb to a non-classical drug binding site. The alpha subunit of Hb has been assigned to posses the binding site for DOX with a binding affinity (Ka) = 16.98 x10(3) mol(-1). The interaction was found to be thermodynamically favorable (DeltaG degrees = -66.23 KJmol(-1)). The analysis of DOX binding site to Hb suggested that the types of interactions that contribute in this binding are hydrophobic contacts, hydrogen bonding and electrostatic interactions.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA