Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 79
Filtrar
1.
Lab Invest ; 99(5): 612-624, 2019 05.
Artigo em Inglês | MEDLINE | ID: mdl-30664711

RESUMO

Nasal natural killer/T-cell lymphoma (NNKTL) is closely associated with Epstein-Barr virus (EBV) and is characterized by poor prognosis, resulting from rapid progression of lesions in the affected organs. Recent data have shown that NNKTL is associated with the aberrant expression of cyclin-dependent kinase 1 (CDK1) and its downstream target survivin, but little is known about the functional roles of CDK1 and survivin in NNKTL. In the current study, we show that knockdown of the EBV-encoded oncoprotein latent membrane protein 1 (LMP1) induces downregulation of CDK1 and survivin in NNKTL cells. Immunohistochemistry detected CDK1 and survivin expression in LMP1-positive cells of NNKTL biopsy specimens. Inhibition of CDK1 and survivin in NNKTL cells with several inhibitors led to a dose-dependent decrease in cell proliferation. In addition, the Sp1 inhibitor mithramycin, which can downregulate both CDK1 and survivin, significantly suppressed the growth of established NNKTL in a murine xenograft model. Our results suggest that LMP1 upregulation of CDK1 and survivin may be essential for NNKTL progression. Furthermore, targeting CDK1 and survivin with Sp1 inhibitors such as mithramycin may be an effective approach to treat NNKTL, which is considered to be a treatment-refractory lymphoma.


Assuntos
Proteína Quinase CDC2/metabolismo , Células Matadoras Naturais/metabolismo , Linfoma de Células T/metabolismo , Neoplasias Nasais/metabolismo , Survivina/metabolismo , Animais , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Proteína Quinase CDC2/antagonistas & inibidores , Proteína Quinase CDC2/genética , Linhagem Celular Tumoral , Feminino , Humanos , Células Matadoras Naturais/efeitos dos fármacos , Linfoma de Células T/tratamento farmacológico , Linfoma de Células T/genética , Masculino , Camundongos Endogâmicos NOD , Camundongos Knockout , Camundongos SCID , Pessoa de Meia-Idade , Neoplasias Nasais/tratamento farmacológico , Neoplasias Nasais/genética , Plicamicina/administração & dosagem , Interferência de RNA , Survivina/antagonistas & inibidores , Survivina/genética , Proteínas da Matriz Viral/genética , Proteínas da Matriz Viral/metabolismo , Ensaios Antitumorais Modelo de Xenoenxerto/métodos
2.
Int J Mol Sci ; 19(5)2018 May 09.
Artigo em Inglês | MEDLINE | ID: mdl-29747385

RESUMO

Osteoarthritis (OA) is the most common and increasing joint disease worldwide. Current treatment for OA is limited to control of symptoms. The purpose of this study was to determine the effect of specificity protein 1 (SP1) inhibitor Mithramycin A (MitA) on chondrocyte catabolism and OA pathogenesis and to explore the underlying molecular mechanisms involving SP1 and other key factors that are critical for OA. Here, we show that MitA markedly inhibited expressions of matrix-degrading enzymes induced by pro-inflammatory cytokine interleukin-1β (IL-1β) in mouse primary chondrocytes. Intra-articular injection of MitA into mouse knee joint alleviated OA cartilage destruction induced by surgical destabilization of the medial meniscus (DMM). However, modulation of SP1 level in chondrocyte and mouse cartilage did not alter catabolic gene expression or cartilage integrity, respectively. Instead, MitA significantly impaired the expression of HIF-2α known to be critical for OA pathogenesis. Such reduction in expression of HIF-2α by MitA was caused by inhibition of NF-κB activation, at least in part. These results suggest that MitA can alleviate OA pathogenesis by suppressing NF-κB-HIF-2α pathway, thus providing insight into therapeutic strategy for OA.


Assuntos
Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Cartilagem Articular/metabolismo , Cartilagem Articular/patologia , Osteoartrite/tratamento farmacológico , Plicamicina/análogos & derivados , Animais , Cartilagem Articular/efeitos dos fármacos , Células Cultivadas , Condrócitos/efeitos dos fármacos , Condrócitos/enzimologia , Condrócitos/metabolismo , Progressão da Doença , Indução Enzimática/efeitos dos fármacos , Interleucina-1beta/farmacologia , Articulações/patologia , Masculino , Metaloproteinases da Matriz/metabolismo , Camundongos Endogâmicos C57BL , NF-kappa B/metabolismo , Osteoartrite/enzimologia , Osteoartrite/patologia , Plicamicina/administração & dosagem , Plicamicina/farmacologia , Plicamicina/uso terapêutico , Fator de Transcrição Sp1/metabolismo
3.
Int J Nanomedicine ; 12: 5255-5269, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28769562

RESUMO

Previous studies have shown that mithramycin A (MIT) is a promising candidate for the treatment of pancreatic carcinoma through inhibiting transcription factor Sp1. However, systemic toxicities may limit its clinical application. Here, we report a rationally designed formulation of MIT-loaded nanoparticles (MIT-NPs) with a small size and sustained release for improved passive targeting and enhanced therapeutic efficacy. Nearly spherical MIT-NPs with a mean particle size of 25.0±4.6 nm were prepared by encapsulating MIT into methoxy poly(ethylene glycol)-block-poly(d,l-lactic-co-glycolic acid) (mPEG-PLGA) nanoparticles (NPs) with drug loading of 2.11%±0.51%. The in vitro release of the MIT-NPs lasted for >48 h with a sustained-release pattern. The cytotoxicity of MIT-NPs to human pancreatic cancer BxPC-3 and MIA Paca-2 cells was comparable to that of free MIT. Determined by flow cytometry and confocal microscopy, the NPs internalized into the cells quickly and efficiently, reaching the peak level at 1-2 h. In vivo fluorescence imaging showed that the prepared NPs were gradually accumulated in BxPC-3 and MIA Paca-2 xenografts and retained for 168 h. The fluorescence intensity in both BxPC-3 and MIA Paca-2 tumors was much stronger than that of various tested organs. Therapeutic efficacy was evaluated with the poorly permeable BxPC-3 pancreatic carcinoma xenograft model. At a well-tolerated dose of 2 mg/kg, MIT-NPs suppressed BxPC-3 tumor growth by 96%. Compared at an equivalent dose, MIT-NPs exerted significantly higher therapeutic effect than free MIT (86% versus 51%, P<0.01). Moreover, the treatment of MIT and MIT-NPs reduced the expression level of oncogene c-Myc regulated by Sp1, and notably, both of them decreased the protein level of CD47. In summary, the novel formulation of MIT-NPs shows highly therapeutic efficacy against pancreatic carcinoma xenograft. In addition, MIT-NPs can downregulate CD47 expression, implying that it might play a positive role in cancer immunotherapy.


Assuntos
Nanopartículas/administração & dosagem , Neoplasias Pancreáticas/tratamento farmacológico , Plicamicina/administração & dosagem , Poliésteres/química , Polietilenoglicóis/química , Animais , Antibióticos Antineoplásicos/administração & dosagem , Antibióticos Antineoplásicos/química , Antibióticos Antineoplásicos/farmacocinética , Linhagem Celular Tumoral , Portadores de Fármacos/administração & dosagem , Portadores de Fármacos/química , Liberação Controlada de Fármacos , Feminino , Humanos , Camundongos Endogâmicos BALB C , Microscopia Confocal , Nanopartículas/química , Tamanho da Partícula , Plicamicina/farmacocinética , Distribuição Tecidual , Ensaios Antitumorais Modelo de Xenoenxerto , Neoplasias Pancreáticas
4.
Arch Dermatol Res ; 309(8): 611-623, 2017 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-28695331

RESUMO

SAHA (vorinostat) is a histone deacetylase inhibitor approved by the USA Food and Drug Administration (FDA) for treating advanced refractory cutaneous T cell lymphomas. As SAHA alters the expression of many genes under control of the Sp1 transcription factor, we examined the effect of its association with the FDA-approved anticancer antibiotic Mithramycin A (MTR, plicamycin), a competitive inhibitor of Sp1 binding to DNA. Sézary syndrome (SS) cells, expanded ex vivo from peripheral blood mononuclear cells of 4 patients, were tested for their sensitivity to the drugs regarding cytotoxicity and differential responsive gene expression. Multivariate statistical methods were used to identify genes whose expression is altered by SAHA, MTR, and the synergist effect of the two drugs. MTR, like SAHA, induced the apoptosis of SS cells, while the two drugs in combination showed clear synergy or potentiation. Expression data stressed a likely important role of additive or synergistic epigenetic modifications in the combined effect of the two drugs, while direct inhibition of Sp1-dependent transcription seemed to have only limited impact. Ontological analysis of modified gene expression suggested that the two drugs, either independently or synergistically, counteracted many intertwined pro-survival pathways deregulated in SS cells, resistance of these tumors to intrinsic and extrinsic apoptosis, abnormal adhesion migration, and invasive properties, as well as immunosuppressive behavior. Our findings provide preliminary clues on the individual and combined effects of SAHA and MTR in SS cells and highlight a potential therapeutic interest of this novel pair of drugs for treatment of SS patients.


Assuntos
Ácidos Hidroxâmicos/uso terapêutico , Plicamicina/uso terapêutico , Síndrome de Sézary/tratamento farmacológico , Neoplasias Cutâneas/tratamento farmacológico , Antineoplásicos/administração & dosagem , Antineoplásicos/uso terapêutico , Linhagem Celular Tumoral , Quimioterapia Combinada , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Humanos , Ácidos Hidroxâmicos/administração & dosagem , Plicamicina/administração & dosagem , Transcriptoma , Vorinostat
5.
Tumour Biol ; 39(6): 1010428317708697, 2017 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-28631560

RESUMO

Mithramycin exhibits certain anticancer effects in glioma, metastatic cerebral carcinoma, malignant lymphoma, chorionic carcinoma and breast cancer. However, its effects on salivary adenoid cystic carcinoma remain unclear. Here, we report that mithramycin significantly inhibited epithelial-to-mesenchymal transition and invasion in human salivary adenoid cystic carcinoma cell lines. The underlying mechanism for this activity was further demonstrated to involve decreasing the expression of the transcription factors specificity protein 1 and SNAI1. Specificity protein 1 is a pro-tumourigenic transcription factor that is overexpressed in SACC-LM and SACC-83 cells, and its expression is inhibited by mithramycin. Moreover, chromatin immunoprecipitation assays showed that specificity protein 1 induced SNAI1 transcription through direct binding to the SNAI1 promoter. In summary, this study uncovered the mechanism through which mithramycin inhibits epithelial-to-mesenchymal transition and invasion in salivary adenoid cystic carcinoma cell lines, namely, via downregulating specificity protein 1 and SNAI1 expression, which suggests mithramycin may be a promising therapeutic option for salivary adenoid cystic carcinoma.


Assuntos
Carcinoma Adenoide Cístico/tratamento farmacológico , Plicamicina/administração & dosagem , Fatores de Transcrição da Família Snail/genética , Fator de Transcrição Sp1/genética , Carcinoma Adenoide Cístico/genética , Carcinoma Adenoide Cístico/patologia , Linhagem Celular Tumoral , Movimento Celular/efeitos dos fármacos , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Transição Epitelial-Mesenquimal/efeitos dos fármacos , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Humanos , Regiões Promotoras Genéticas/genética , Fatores de Transcrição da Família Snail/metabolismo , Fator de Transcrição Sp1/metabolismo , Ativação Transcricional/efeitos dos fármacos
6.
Sci Rep ; 6: 29082, 2016 07 07.
Artigo em Inglês | MEDLINE | ID: mdl-27384152

RESUMO

miRs (microRNAs, miRNAs) intricately regulate physiological and pathological processes. Although miR-7a/b protects against cardiomyocyte injury in ischemia/reperfusion injury, the function of miR-7a/b in myocardial infarction (MI)-induced cardiac remodeling remains unclear. Here, we sought to investigate the function of miR-7a/b in post-MI remodeling in a mouse model and to determine the underlying mechanisms involved. miR-7a/b overexpression improved cardiac function, attenuated cardiac remodeling and reduced fibrosis and apoptosis, whereas miR-7a/b silencing caused the opposite effects. Furthermore, miR-7a/b overexpression suppressed specific protein 1 (Sp1) and poly (ADP-ribose) polymerase (PARP-1) expression both in vivo and in vitro, and a luciferase reporter activity assay showed that miR-7a/b could directly bind to Sp1. Mithramycin, an inhibitor of the DNA binding activity of Sp1, effectively repressed PARP-1 and caspase-3, whereas knocking down miR-7a/b partially counteracted these beneficial effects. Additionally, an immunoprecipitation assay indicated that hypoxia triggered activation of the binding activity of Sp1 to the promoters of PARP-1 and caspase-3, which is abrogated by miR-7a/b. In summary, these findings identified miR-7a/b as protectors of cardiac remodeling and hypoxia-induced injury in H9c2 cardiomyoblasts involving Sp1 and PARP-1.


Assuntos
MicroRNAs/genética , Infarto do Miocárdio/genética , Poli(ADP-Ribose) Polimerase-1/genética , Traumatismo por Reperfusão/genética , Fator de Transcrição Sp1/genética , Animais , Apoptose/genética , Remodelamento Atrial/genética , Caspase 3/genética , Hipóxia Celular/genética , Proteínas de Ligação a DNA/genética , Modelos Animais de Doenças , Regulação da Expressão Gênica/efeitos dos fármacos , Humanos , Camundongos , Infarto do Miocárdio/fisiopatologia , Miócitos Cardíacos/metabolismo , Miócitos Cardíacos/patologia , Plicamicina/administração & dosagem , Traumatismo por Reperfusão/patologia
7.
Clin Cancer Res ; 22(5): 1197-210, 2016 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-26459178

RESUMO

PURPOSE: Specificity protein 1 (SP1) is an oncogenic transcription factor overexpressed in various human malignancies. This study sought to examine SP1 expression in malignant pleural mesotheliomas (MPM) and ascertain the potential efficacy of targeting SP1 in these neoplasms. EXPERIMENTAL DESIGN: qRT-PCR, immunoblotting, and immunohistochemical techniques were used to evaluate SP1 expression in cultured MPM cells and MPM specimens and normal mesothelial cells/pleura. MTS, chemotaxis, soft agar, ß-galactosidase, and Apo-BrdUrd techniques were used to assess proliferation, migration, clonogenicity, senescence, and apoptosis in MPM cells following SP1 knockdown, p53 overexpression, or mithramycin treatment. Murine subcutaneous and intraperitoneal xenograft models were used to examine effects of mithramycin on MPM growth in vivo. Microarray, qRT-PCR, immunoblotting, and chromatin immunoprecipitation techniques were used to examine gene expression profiles mediated by mithramycin and combined SP1 knockdown/p53 overexpression and correlate these changes with SP1 and p53 levels within target gene promoters. RESULTS: MPM cells and tumors exhibited higher SP1 mRNA and protein levels relative to control cells/tissues. SP1 knockdown significantly inhibited proliferation, migration, and clonogenicity of MPM cells. Mithramycin depleted SP1 and activated p53, dramatically inhibiting proliferation and clonogenicity of MPM cells. Intraperitoneal mithramycin significantly inhibited growth of subcutaneous MPM xenografts and completely eradicated mesothelioma carcinomatosis in 75% of mice. Mithramycin modulated genes mediating oncogene signaling, cell-cycle regulation, senescence, and apoptosis in vitro and in vivo. The growth-inhibitory effects of mithramycin in MPM cells were recapitulated by combined SP1 knockdown/p53 overexpression. CONCLUSIONS: These findings provide preclinical rationale for phase II evaluation of mithramycin in patients with mesothelioma.


Assuntos
Biomarcadores Tumorais/biossíntese , Neoplasias Pulmonares/tratamento farmacológico , Mesotelioma/tratamento farmacológico , Neoplasias Pleurais/tratamento farmacológico , Fator de Transcrição Sp1/biossíntese , Proteína Supressora de Tumor p53/biossíntese , Animais , Apoptose/efeitos dos fármacos , Biomarcadores Tumorais/genética , Proliferação de Células/efeitos dos fármacos , Senescência Celular/efeitos dos fármacos , Feminino , Regulação Neoplásica da Expressão Gênica , Humanos , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/patologia , Mesotelioma/genética , Mesotelioma/patologia , Mesotelioma Maligno , Camundongos , Pessoa de Meia-Idade , Neoplasias Pleurais/genética , Neoplasias Pleurais/patologia , Plicamicina/administração & dosagem , RNA Mensageiro/biossíntese , Fator de Transcrição Sp1/antagonistas & inibidores , Fator de Transcrição Sp1/genética , Proteína Supressora de Tumor p53/genética , Ensaios Antitumorais Modelo de Xenoenxerto
8.
PLoS One ; 9(12): e113624, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25479176

RESUMO

BACKGROUND AND PURPOSE: Increased expression of endothelin receptor type B (ETBR), a vasoactive receptor, has recently been implied in the reduced cerebral blood flow and exacerbated neuronal damage after ischemia-reperfusion (I/R). The study explores the regulatory mechanisms of ETBR to identify drug targets to restore normal cerebral artery contractile function as part of successful neuroprotective therapy. METHODS: We have employed in vitro methods on human and rat cerebral arteries to study the regulatory mechanisms and the efficacy of target selective inhibitor, Mithramycin A (MitA), to block the ETBR mediated contractile properties. Later, middle cerebral artery occluded (MCAO) rats were used to substantiate the observations. Quantative PCR, immunohistochemistry, western blot and wire myograph methods were employed to study the expression and contractile properties of cerebral arteries. RESULTS: Increased expression of specificity protein (Sp1) was observed in human and rat cerebral arteries after organ culture, strongly correlating with the ETBR upregulation. Similar observations were made in MCAO rats. Treatment with MitA, a Sp1 specific inhibitor, significantly downregulated the ETBR mRNA and protein levels. It also significantly reduced the ETBR mediated cerebrovascular contractility. Detailed analysis indicated that ERK1/2 mediated phosphorylation of Sp1 might be essential for ETBR transcription. CONCLUSION: Transcription factor Sp1 regulates the ETBR mediated vasoconstriction in focal cerebral ischemia via MEK-ERK signaling, which is also conserved in humans. The results show that MitA can effectively be used to block ETBR mediated vasoconstriction as a supplement to an existing ischemic stroke therapy.


Assuntos
Isquemia Encefálica/tratamento farmacológico , Circulação Cerebrovascular/efeitos dos fármacos , Receptores de Endotelina/biossíntese , Vasoconstrição/efeitos dos fármacos , Animais , Isquemia Encefálica/genética , Isquemia Encefálica/patologia , Artérias Cerebrais/efeitos dos fármacos , Artérias Cerebrais/patologia , Circulação Cerebrovascular/genética , Regulação da Expressão Gênica/efeitos dos fármacos , Humanos , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Técnicas de Cultura de Órgãos , Plicamicina/administração & dosagem , Plicamicina/análogos & derivados , Ratos , Receptor de Endotelina B , Receptores de Endotelina/efeitos dos fármacos , Receptores de Endotelina/genética , Fator de Transcrição Sp1/antagonistas & inibidores
9.
Int J Nanomedicine ; 7: 307-24, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22287841

RESUMO

This report shows that the DNA-binding drug, mithramycin, can be efficiently encapsulated in polymeric micelles (PM-MTH), based on Pluronic(®) block copolymers, by a new microfluidic approach. The effect of different production parameters has been investigated for their effect on PM-MTH characteristics. The compared analysis of PM-MTH produced by microfluidic and conventional bulk mixing procedures revealed that microfluidics provides a useful platform for the production of PM-MTH with improved controllability, reproducibility, smaller size, and polydispersity. Finally, an investigation of the effects of PM-MTH, produced by microfluidic and conventional bulk mixing procedures, on the erythroid differentiation of both human erythroleukemia and human erythroid precursor cells is reported. It is demonstrated that PM-MTH exhibited a slightly lower toxicity and more pronounced differentiative activity when compared to the free drug. In addition, PM-MTH were able to upregulate preferentially γ-globin messenger ribonucleic acid production and to increase fetal hemoglobin (HbF) accumulation, the percentage of HbF-containing cells, and their HbF content without stimulating α-globin gene expression, which is responsible for the clinical symptoms of ß-thalassemia. These results represent an important first step toward a potential clinical application, since an increase in HbF could alleviate the symptoms underlying ß-thalassemia and sickle cell anemia. In conclusion, this report suggests that PM-MTH produced by microfluidic approach warrants further evaluation as a potential therapeutic protocol for ß-thalassemia.


Assuntos
Química Farmacêutica/métodos , Micelas , Microfluídica , Plicamicina/análogos & derivados , Polímeros , Talassemia beta/tratamento farmacológico , Análise de Variância , Diferenciação Celular/efeitos dos fármacos , Células Cultivadas , Eritrócitos/efeitos dos fármacos , Eritrócitos/patologia , Células Precursoras Eritroides , Humanos , Células K562/efeitos dos fármacos , Dispositivos Lab-On-A-Chip , Plicamicina/administração & dosagem , Plicamicina/química , Reprodutibilidade dos Testes
10.
Cancer Res ; 71(15): 5182-93, 2011 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-21673052

RESUMO

Both betulinic acid (BA) and mithramycin A (MIT) exhibit potent antitumor activity through distinct mechanisms of Sp1 inhibition. However, it is unknown whether a combination of these two compounds results in a synergistic inhibitory effect on pancreatic cancer growth and/or has a therapeutic advantage over gemcitabine. In xenograft mouse models of human pancreatic cancer, treatment with either BA or MIT alone showed dose-dependent antitumor activity but led to systemic side effects as measured by overall weight loss. Treatment with a nontoxic dose of either compound alone had only marginal antitumor effects. Importantly, combination treatment with nontoxic doses of BA and MIT produced synergistic antitumor activity, including inhibitory effects on cell proliferation, invasion, and angiogenesis. The treatment combination also produced less discernible side effects than therapeutic doses of gemcitabine. Moreover, combined treatment of BA and MIT resulted in drastic inhibition of Sp1 recruitment onto Sp1 and VEGF promoters, leading to transcriptional inhibition of both Sp1 and VEGF and downregulation of Sp1 and VEGF protein expression. Ectopic overexpression of Sp1 rendered tumor cells resistant to BA, MIT, and the combination of the two. Overall, our findings argue that Sp1 is an important target of BA and MIT and that their combination can produce an enhanced therapeutic response in human pancreatic cancer.


Assuntos
Adenocarcinoma/tratamento farmacológico , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Neoplasias Pancreáticas/tratamento farmacológico , Plicamicina/farmacologia , Triterpenos/farmacologia , Adenocarcinoma/irrigação sanguínea , Adenocarcinoma/genética , Adenocarcinoma/patologia , Animais , Divisão Celular/efeitos dos fármacos , Linhagem Celular Tumoral/efeitos dos fármacos , Colágeno , Combinação de Medicamentos , Ensaios de Seleção de Medicamentos Antitumorais , Sinergismo Farmacológico , Feminino , Perfilação da Expressão Gênica , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Humanos , Laminina , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Invasividade Neoplásica , Neovascularização Patológica/tratamento farmacológico , Neoplasias Pancreáticas/irrigação sanguínea , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/patologia , Triterpenos Pentacíclicos , Plicamicina/administração & dosagem , Regiões Promotoras Genéticas/genética , Proteoglicanas , Proteínas Recombinantes de Fusão/biossíntese , Fator de Transcrição Sp1/genética , Triterpenos/administração & dosagem , Fator A de Crescimento do Endotélio Vascular/genética , Ensaios Antitumorais Modelo de Xenoenxerto , Ácido Betulínico
11.
Otolaryngol Head Neck Surg ; 145(3): 435-41, 2011 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-21521900

RESUMO

OBJECTIVE: To evaluate efficacy of a drug-eluting, dissolvable esophageal (DEDE) stent for the prevention of submucosal collagen deposition in a rat model of acute esophageal injury. SETTING: University laboratory. STUDY DESIGN: Interventional randomized controlled trial. SUBJECTS AND METHODS: Forty two adult, male, age-matched Sprague Dawley rats were randomized to undergo either sham esophageal surgery, esophageal burn injury, or esophageal burn injury and placement of a DEDE stent. All animals underwent open gastrotomy under anesthesia. In group 1, a cautery device was inserted through the gastrotomy into the distal esophagus and removed without creating an injury. In group 2, the cautery was placed in the distal esophagus and a circumferential thermal burn injury was created. In group 3, an identical burn injury was created and a DEDE stent was placed at the site of injury and secured. On postoperative day 28, animals were sacrificed, and the distal esophagi were harvested and processed for histology. Submucosal collagen area was quantified histologically and compared across the 3 experimental groups. RESULTS: After the investigators controlled for luminal circumference and multiple measurements, submucosal collagen area was increased in group 2 (burn) compared with group 1 (sham) (P = .012). Submucosal collagen area was decreased in group 3 (DEDE stent) compared with group 2 (P = .042). No statistically significant difference in submucosal collagen area was observed between animals in group 1 and group 3 (P = .800). CONCLUSIONS;Topical application of mithramycin-A via a DEDE stent modulates collagen deposition after acute thermal injury in the rat esophagus.


Assuntos
Queimaduras Químicas/tratamento farmacológico , Colágeno/efeitos dos fármacos , Stents Farmacológicos , Estenose Esofágica/prevenção & controle , Plicamicina/administração & dosagem , Administração Tópica , Animais , Queimaduras Químicas/patologia , Queimaduras Químicas/cirurgia , Colágeno/metabolismo , Modelos Animais de Doenças , Estenose Esofágica/induzido quimicamente , Masculino , Mucosa/efeitos dos fármacos , Mucosa/patologia , Distribuição Aleatória , Ratos , Ratos Sprague-Dawley , Valores de Referência , Resultado do Tratamento
13.
Cancer Res ; 70(3): 1111-9, 2010 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-20086170

RESUMO

Mithramycin (MIT) and tolfenamic acid (TA) inhibit the activity of the transcription factor Sp1. In the present study, we investigated whether pancreatic cancer treatment with a combination of these compounds has a synergistic effect on Sp1 activity, tumor growth, and their underlying response mechanisms. Treatment of pancreatic tumor xenografts with MIT and TA produced dose-dependent antitumor activity, and significant antitumor activity of either compound alone was directly associated with systemic side effects. Combination treatment with nontoxic doses of both compounds produced synergistic antitumor activity, whereas treatment with a nontoxic dose of either compound alone lacked a discernible antitumor effect. Synergistic therapeutic effects correlated directly with synergistic antiproliferation and antiangiogenesis in vitro. Moreover, combination treatment resulted in Sp1 protein degradation, drastically downregulating expression of Sp1 and vascular endothelial growth factor. Our findings established that Sp1 is a critical target of TA and MIT in human pancreatic cancer therapy, rationalizing clinical studies to determine the effect of existing pancreatic cancer therapy regimens on Sp1 signaling in tumors and normal pancreatic tissue, and the ability of Sp1-targeting strategies to modify cancer responses.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Neoplasias Pancreáticas/tratamento farmacológico , Fator de Transcrição Sp1/metabolismo , Animais , Western Blotting , Peso Corporal/efeitos dos fármacos , Linhagem Celular Tumoral , Imunoprecipitação da Cromatina , Relação Dose-Resposta a Droga , Sinergismo Farmacológico , Feminino , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Neoplasias Pancreáticas/metabolismo , Neoplasias Pancreáticas/patologia , Plicamicina/administração & dosagem , Plicamicina/análogos & derivados , Regiões Promotoras Genéticas/genética , Ligação Proteica , Fator de Transcrição Sp1/genética , Carga Tumoral/efeitos dos fármacos , Fator A de Crescimento do Endotélio Vascular/genética , Fator A de Crescimento do Endotélio Vascular/metabolismo , Ensaios Antitumorais Modelo de Xenoenxerto , ortoaminobenzoatos/administração & dosagem
14.
Brain Res ; 1301: 189-96, 2009 Dec 08.
Artigo em Inglês | MEDLINE | ID: mdl-19748494

RESUMO

The present study was undertaken to examine the effects of mithramycin, an inhibitor of transcription factor Specificity protein (Sp)-1, on the behavioral changes and dopaminergic neurotoxicity in the mouse striatum after administration of methamphetamine (METH). Pretreatment with mithramycin (75, 150 or 300 microg/kg) did not alter acute hyperlocomotion in mice after a single administration of METH (3 mg/kg). However, the development of behavioral sensitization in mice after repeated administration of METH (3 mg/kg/day, once daily for 5 days) was significantly blocked by pretreatment with mithramycin (300 microg/kg). Furthermore, pretreatment with mithramycin (300 microg/kg) significantly attenuated the hyperthermia in mice after repeated administration of METH (3 mg/kgx3, 3-h intervals). Moreover, the combination of pretreatment and subsequent administration of mithramycin (75, 150 or 300 microg/kg) significantly attenuated the reductions of dopamine (DA), its major metabolite 3,4-dihydroxyphenylacetic acid (DOPAC) and DA transporter (DAT) in the striatum after repeated administration of METH (3 mg/kgx3, 3-h intervals), and these attenuations were dose dependent. These findings suggest that mithramycin attenuates the development of behavioral sensitization and dopaminergic neurotoxicity in mice after repeated administration of METH. Therefore, mithramycin could have potential for the treatment of METH abusers, particularly since this drug has been approved by the Food and Drug Administration in the United States. In the future, however, another Sp1 inhibitors with fewer side effects might be more appropriate.


Assuntos
Corpo Estriado/efeitos dos fármacos , Dopamina/metabolismo , Metanfetamina/farmacologia , Plicamicina/administração & dosagem , Ácido 3,4-Di-Hidroxifenilacético/metabolismo , Análise de Variância , Animais , Comportamento Animal/efeitos dos fármacos , Cromatografia Líquida de Alta Pressão , Corpo Estriado/metabolismo , Proteínas da Membrana Plasmática de Transporte de Dopamina/metabolismo , Relação Dose-Resposta a Droga , Interações Medicamentosas , Febre/induzido quimicamente , Febre/prevenção & controle , Hipercinese/induzido quimicamente , Hipercinese/prevenção & controle , Imuno-Histoquímica , Masculino , Camundongos , Atividade Motora/efeitos dos fármacos , Fármacos Neuroprotetores/administração & dosagem , Inibidores da Síntese de Proteínas/administração & dosagem
15.
J Pharm Sci ; 98(4): 1452-62, 2009 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-18704956

RESUMO

Formulation of hydrophilic compounds in nanoparticles is problematic due to their escape to the external aqueous phase. The certain amphiphilic nature of mithramycin, utilized clinically in cancer, makes its incorporation into nanoparticles an interesting challenge, elucidating the formulation factors of amphiphilics in nanoparticles. We hypothesized that mithramycin nanoparticles could provide more effective therapy of restenosis due to its antiproliferating and potential monocyte inhibition properties. The nanoprecipitation technique (designed for lipophilic compounds) was found preferable, with better encapsulation efficiency, than the emulsification solvent diffusion (ESD) technique (79.3 +/- 3.1% and 40.8 +/- 1.1%, respectively). The double emulsion solvent diffusion (DESD) method, designed for hydrophilic compounds, yielded similar encapsulation efficiency (80%). Nanoparticles size was, 110 +/- 36, 130 +/- 30, and 160 +/- 31 nm, ESD, nanoprecipitation, and DESD techniques, respectively. Mithramycin solution and in nanoparticles significantly inhibited RAW264 macrophages and smooth muscle cells in a dose-dependent relationship, and reduced the number of circulating monocytes in rabbits. However, no inhibition of restenosis was obtained in the rat carotid model following i.v. administration of mithramycin nanoparticles. It can be concluded that PLGA-based polymeric nanoparticles of mithramycin can be formulated by techniques suitable for lipophilic/hydrophilic compounds. The ineffectiveness in the rat restenosis model is probably due to the short depletion period of circulating monocytes and lack of arterial targeting.


Assuntos
Portadores de Fármacos/química , Composição de Medicamentos/métodos , Ácido Láctico/química , Nanopartículas/química , Plicamicina/química , Ácido Poliglicólico/química , Tensoativos/química , Animais , Aorta Torácica/citologia , Estenose das Carótidas/tratamento farmacológico , Linhagem Celular , Proliferação de Células/efeitos dos fármacos , Emulsões , Contagem de Leucócitos , Macrófagos/efeitos dos fármacos , Masculino , Camundongos , Monócitos/citologia , Monócitos/efeitos dos fármacos , Músculo Liso Vascular/citologia , Tamanho da Partícula , Plicamicina/administração & dosagem , Plicamicina/farmacologia , Plicamicina/uso terapêutico , Copolímero de Ácido Poliláctico e Ácido Poliglicólico , Coelhos , Ratos , Solubilidade , Solventes/química
16.
Biochem Biophys Res Commun ; 342(4): 1168-73, 2006 Apr 21.
Artigo em Inglês | MEDLINE | ID: mdl-16516150

RESUMO

We show that a histone deacetylase (HDAC) inhibitor apicidin increases the transcriptional activity of cyclin E gene, which results in accumulation of cyclin E mRNA and protein in a time- and dose-dependent manner. Interestingly, apicidin induction of cyclin E gene is found to be mediated by Sp1- rather than E2F-binding sites in the cyclin E promoter, as evidenced by the fact that specific inhibition of Sp1 leads to a decrease in apicidin activation of cyclin E promoter activity and protein expression, but mutation of E2F-binding sites of cyclin E promoter region fails to inhibit the ability of apicidin to activate cyclin E transcription. In addition, this transcriptional activation of cyclin E by apicidin is associated with histone hyperacetylation of cyclin E promoter region containing Sp1-binding sites. Our results demonstrate that regulation of histone modification by an HDAC inhibitor apicidin contributes to induction of cyclin E expression and this effect is Sp1-dependent.


Assuntos
Ciclina E/metabolismo , Regulação da Expressão Gênica/efeitos dos fármacos , Inibidores de Histona Desacetilases , Histona Desacetilases/metabolismo , Peptídeos Cíclicos/administração & dosagem , Plicamicina/administração & dosagem , Transdução de Sinais/efeitos dos fármacos , Fator de Transcrição Sp1/metabolismo , Sítios de Ligação , Relação Dose-Resposta a Droga , Células HeLa , Humanos , Ligação Proteica , Ativação Transcricional/efeitos dos fármacos
17.
J Control Release ; 77(3): 167-81, 2001 Dec 13.
Artigo em Inglês | MEDLINE | ID: mdl-11733085

RESUMO

Arterial restenosis is responsible for the high failure rates of vascular reconstruction procedures. Local sustained drug delivery has shown promise in the prevention of restenosis. The drug release rate from mithramycin-loaded EVA matrices (0.1%) was evaluated, and their antirestenotic effect was studied in the rat carotid model and rabbit model of vascular grafts. The modulation of c-myc expression by mithramycin treatment was examined by immunohistochemistry in the rat carotid model. The proliferative response of injured rat arteries was studied by bromdeoxyuridine (BrdU) immunostaining. The impact of mithramycin treatment on vasomotor responses of the venous segments grafted into arterial circulation was studied ex vivo using vasoreactive compounds. Mithramycin was released exponentially from EVA matrices in PBS. Matrices co-formulated with PEG-4600 revealed enhanced release kinetics. The perivascular implantation of drug-loaded EVA-PEG matrices led to 50% reduction of neointimal formation, and reduced the c-myc expression and BrdU labeling in comparison to control implants. Decreased sensitivity of mithramycin-treated grafts to serotonin-induced vasoconstriction was observed. Local perivascular mithramycin treatment limits the functional alteration caused by the grafting of venous segments in high-pressure arterial environment, and potently inhibits stenosis secondary to grafting and angioplasty injury. The antirestenotic effect is associated with reduced c-myc expression and with subsequent decrease in SMC proliferation.


Assuntos
Artéria Carótida Primitiva/transplante , Sistemas de Liberação de Medicamentos/métodos , Oclusão de Enxerto Vascular/prevenção & controle , Inibidores do Crescimento/administração & dosagem , Veias Jugulares/transplante , Plicamicina/administração & dosagem , Túnica Íntima/efeitos dos fármacos , Animais , Artéria Carótida Primitiva/efeitos dos fármacos , Artéria Carótida Primitiva/fisiopatologia , Cateterismo/efeitos adversos , Células Cultivadas , Oclusão de Enxerto Vascular/patologia , Inibidores do Crescimento/farmacologia , Veias Jugulares/efeitos dos fármacos , Veias Jugulares/fisiopatologia , Masculino , Músculo Liso Vascular/patologia , Plicamicina/farmacologia , Coelhos , Ratos , Suínos , Túnica Íntima/patologia , Túnica Íntima/fisiopatologia
18.
Leuk Res ; 21(5): 375-80, 1997 May.
Artigo em Inglês | MEDLINE | ID: mdl-9225062

RESUMO

Thirteen patients with accelerated phase of chronic myeloid leukemia (CML-AC) were treated with intravenous plicamycin and subcutaneous alpha-interferon. Two patients stabilized, three patients had partial hematologic responses and one patient had a hematologic complete response with a major cytogenetic response. Two patients, progressing on hydroxyurea, did not respond, but demonstrated re-sensitization to hydroxyurea after completion of induction therapy and had prolonged return to chronic phase for 30 months and 25 months. Four non-responders subsequently received additional chemotherapy and responded. Median survival of all study patients from the development of accelerated phase of CML was 24 months: substantially longer than other reported series (median 6 months). Plicamycin appears to add efficacy to interferon in the stabilization of accelerated phase of CML.


Assuntos
Antibióticos Antineoplásicos/uso terapêutico , Antineoplásicos/uso terapêutico , Interferon-alfa/uso terapêutico , Leucemia Mielogênica Crônica BCR-ABL Positiva/terapia , Plicamicina/uso terapêutico , Adulto , Idoso , Antibióticos Antineoplásicos/administração & dosagem , Antineoplásicos/administração & dosagem , Biópsia , Medula Óssea/patologia , Esquema de Medicação , Feminino , Humanos , Hidroxiureia/uso terapêutico , Infusões Intravenosas , Injeções Subcutâneas , Interferon-alfa/administração & dosagem , Leucemia Mielogênica Crônica BCR-ABL Positiva/mortalidade , Leucemia Mielogênica Crônica BCR-ABL Positiva/patologia , Masculino , Pessoa de Meia-Idade , Projetos Piloto , Plicamicina/administração & dosagem , Taxa de Sobrevida
20.
Ann Pharmacother ; 26(11): 1369-73, 1992 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-1477439

RESUMO

OBJECTIVE: To report a case of possible severe thrombocytopenia associated with administration of a single dose of plicamycin. CASE SUMMARY: A 73-year-old man with prostate cancer was admitted to the hospital with hypercalcemia (total serum calcium concentration 4.02 mmol/L) and a low baseline platelet count (152 x 10(9)/L). Because of his symptomatic hypercalcemia, he was treated with NaCl 0.9%, furosemide, oral inorganic phosphate, and a single dose of plicamycin (15 micrograms/kg). Five days after plicamycin administration his platelet count decreased to 52 x 10(9)/L, and continued to decrease further even after the transfusion of four units of platelets to a nadir of 7 x 10(9)/L (hospital day 20). A second transfusion produced a small increase in his platelet count. The patient's clinical status continued to deteriorate, however, and he subsequently died. DISCUSSION: Plicamycin and other drugs that may induce thrombocytopenia are reviewed. The time course between plicamycin administration and the development of thrombocytopenia in our patient is assessed. Other contributing factors such as a low baseline platelet count and advanced age are also addressed. CONCLUSIONS: It is likely that the severe thrombocytopenia experienced by our patient was caused by a single dose of plicamycin. Adjusting the dosage for a patient's renal function as well as close monitoring of the platelet count are necessary when administering this drug. We report this case to remind clinicians of the potential for the development of severe thrombocytopenia following administration of a single dose of plicamycin.


Assuntos
Plicamicina/efeitos adversos , Trombocitopenia/induzido quimicamente , Idoso , Carcinoma/complicações , Esquema de Medicação , Humanos , Hipercalcemia/tratamento farmacológico , Hipercalcemia/etiologia , Masculino , Contagem de Plaquetas/efeitos dos fármacos , Plicamicina/administração & dosagem , Neoplasias da Próstata/complicações
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...