Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 50
Filtrar
1.
Cancer Sci ; 103(9): 1730-6, 2012 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-22676179

RESUMO

A proteasome degrades numerous regulatory proteins that are critical for tumor growth and is therefore recognized as a promising anticancer target. Determining proteasome activity in the tumors of mice bearing xenografts is essential for the development of novel proteasome inhibitors. We developed a system for in vivo imaging of proteasome inhibition in the tumors of living mice, using a proteasome-sensitive fluorescent reporter, ZsProSensor-1. This reporter consists of a green fluorescent protein, ZsGreen, fused to mouse ornithine decarboxylase, which is degraded by the proteasome without being ubiquitinated. In stably transfected cells expressing ZsProSensor-1, the fluorescent reporter was rapidly degraded under steady-state conditions, whereas it was stabilized in the presence of proteasome inhibitors. Subcutaneous inoculation of the transfected cells into nude mice resulted in tumor formation. When the proteasome inhibitor bortezomib was intravenously administered to mice bearing these tumors, the ZsProSensor-1 protein accumulated in the tumors and emitted a fluorescent signal in a dose-dependent manner. Robust fluorescence was sustained for 3 days and then gradually decreased to baseline levels within 15 days. Intravenous administration of bortezomib also showed potent antitumor activity. In contrast, oral administration of bortezomib did not result in fluorescent protein accumulation in tumors or exhibit any antitumor activity. These results indicate that in vivo imaging using the ZsProSensor-1 fluorescent protein can be used as an indicator of antitumor activity and will be a powerful tool for the development of novel proteasome inhibitors.


Assuntos
Antineoplásicos/farmacologia , Ácidos Borônicos/farmacologia , Genes Reporter , Proteínas de Fluorescência Verde/genética , Imagem Molecular , Complexo de Endopeptidases do Proteassoma/genética , Inibidores de Proteassoma , Pirazinas/farmacologia , Animais , Antineoplásicos/administração & dosagem , Ácidos Borônicos/administração & dosagem , Bortezomib , Linhagem Celular , Feminino , Proteínas de Fluorescência Verde/metabolismo , Células HEK293 , Humanos , Camundongos , Camundongos Nus , Neoplasias/diagnóstico , Neoplasias/genética , Neoplasias/metabolismo , Complexo de Endopeptidases do Proteassoma/metabolismo , Pirazinas/administração & dosagem , Ensaios Antitumorais Modelo de Xenoenxerto
2.
Int J Cancer ; 126(4): 810-8, 2010 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-19795463

RESUMO

Targeting stroma in tumor tissues is an attractive new strategy for cancer treatment. We developed in vitro coculture system, in which the growth of human prostate cancer DU-145 cells is stimulated by prostate stromal cells (PrSC) through insulin-like growth factor I (IGF-I). Using this system, we have been searching for small molecules that inhibit tumor growth through modulation of tumor-stromal cell interactions. As a result, we have found that leucinostatins and atpenins, natural antifungal antibiotics, inhibit the growth of DU-145 cells cocultured with PrSC more strongly than that of DU-145 cells alone. In this study we examined the antitumor effects of these small molecules in vitro and in vivo. When DU-145 cells were coinoculated with PrSC subcutaneously in nude mice, leucinostatin A was found to significantly suppress the tumor growth more than atpenin B. The antitumor effect of leucinostatin A in vivo was not obtained against the tumors of DU-145 cells alone. RT-PCR experiments revealed that leucinostatin A specifically inhibited IGF-I expression in PrSC without effect on expressions of other IGF axis molecules. Leucinostatins and atpenins are known to abrogate mitochondrial functions. However, when we used mitochondrial DNA-depleted, pseudo-rho(0) cells, we found that one of leucinostain A actions certainly depended on mitochondrial function, but it actually inhibited the growth of DU-145 cells more strongly in coculture with pseudo-rho(0) PrSC and reduced IGF-I expression in pseudo-rho(0) PrSC. Taken together, our results suggested that leucinostatin A inhibited prostate cancer cell growth through reduction of IGF-I expression in PrSC.


Assuntos
Antineoplásicos/uso terapêutico , Micotoxinas/uso terapêutico , Peptídeos/uso terapêutico , Neoplasias da Próstata/tratamento farmacológico , Células Estromais/efeitos dos fármacos , Animais , Peptídeos Catiônicos Antimicrobianos , Antineoplásicos/farmacologia , Divisão Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Técnicas de Cocultura , Primers do DNA , Humanos , Masculino , Camundongos , Camundongos Nus , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/patologia , Micotoxinas/farmacologia , Peptídeos/farmacologia , Neoplasias da Próstata/patologia , Piridonas/farmacologia , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Células Estromais/patologia
3.
Cancer Sci ; 101(3): 743-50, 2010 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-20028386

RESUMO

Although cytostatin analog protein phosphatase 2A (PP2A)-specific inhibitors are promising candidates of a new type of anticancer drug, their development has been hindered because of their liability. To find new classes of PP2A-specific inhibitors, we conducted a screening with microbial metabolites and found that rubratoxin A, a classical mycotoxin, is a highly specific and potent inhibitor of the enzyme. While rubratoxin A inhibits PP2A at Ki = 28.7 nm, it hardly inhibited any other phosphatases examined. Rubratoxin B, a close analog, also specifically but weakly inhibits PP2A at Ki = 3.1 microM. The inhibition of intracellular PP2A in cultured cells is obviously observed with 20 microM rubratoxin A treatment for 3 h, inducing the overphosphorylation in PP2A substrate proteins. Although rubratoxins and cytostatin differ in the apparent structures, these compounds share similarities in the structures in detail and PP2A-binding manners. Rubratoxin A showed higher suppression of tumor metastasis and reduction of the primary tumor volume than cytostatin in mouse experiments. As a successor of cytostatin analogs, rubratoxin A should be a good compound leading to the development of antitumor drugs targeting PP2A.


Assuntos
Antineoplásicos/farmacologia , Inibidores Enzimáticos/farmacologia , Micotoxinas/farmacologia , Metástase Neoplásica/prevenção & controle , Proteína Fosfatase 2/antagonistas & inibidores , Animais , Feminino , Camundongos , Camundongos Endogâmicos C57BL , Micotoxinas/metabolismo , Fosforilação
4.
Biochem Biophys Res Commun ; 392(3): 460-6, 2010 Feb 12.
Artigo em Inglês | MEDLINE | ID: mdl-20083087

RESUMO

Large areas of tumor are nutrient-starved and hypoxic due to a disorganized vascular system. Therefore, we screened small molecules to identify cytotoxic agents that function preferentially in nutrient-starved conditions. We found that efrapeptin F had preferential cytotoxicity to nutrient-deprived cells compared with nutrient-sufficient cells. Because efrapeptin F acts as a mitochondrial complex V inhibitor, we examined whether inhibitors of complex I, II, III, and V function as cytotoxic agents preferentially in nutrient-deprived cells. Interestingly, these inhibitors showed preferential cytotoxicity to nutrient-deprived cells and caused cell death under glucose-limiting conditions, irrespective of the presence or absence of amino acids and/or serum. In addition, these inhibitors were preferentially cytotoxic to nutrient-deprived cells even under hypoxic conditions. Further, efrapeptin F showed antitumor activity in vivo. These data indicate that mitochondrial inhibitors show preferential cytotoxicity to cancer cells under glucose-limiting conditions, and these inhibitors offer a promising strategy for anticancer therapeutic.


Assuntos
Antineoplásicos/farmacologia , Inibidores Enzimáticos/farmacologia , Glucose/deficiência , ATPases Mitocondriais Próton-Translocadoras/antagonistas & inibidores , Neoplasias Pancreáticas/enzimologia , Peptídeos/farmacologia , Linhagem Celular Tumoral , Humanos , Peptídeos e Proteínas de Sinalização Intercelular , Peptídeos/química
5.
Bioorg Med Chem Lett ; 20(19): 5839-42, 2010 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-20727746

RESUMO

The structure-activity relationship of the boronic acid derivatives of tyropeptin, a proteasome inhibitor, was studied. Based on the structure of a previously reported boronate analog of tyropeptin (2), 41 derivatives, which have varying substructure at the N-terminal acyl moiety and P2 position, were synthesized. Among them, 3-phenoxyphenylacetamide 6 and 3-fluoro picolinamide 22 displayed the most potent inhibitory activity toward chymotryptic activity of proteasome and cytotoxicity, respectively. The replacement of the isopropyl group in the P2 side chain to H or Me had negligible effects on the biological activities examined in this study.


Assuntos
Compostos de Boro/química , Ácidos Borônicos/química , Dipeptídeos/química , Inibidores Enzimáticos/química , Oligopeptídeos/química , Inibidores de Proteassoma , Compostos de Boro/síntese química , Compostos de Boro/toxicidade , Ácidos Borônicos/síntese química , Ácidos Borônicos/toxicidade , Linhagem Celular Tumoral , Inibidores Enzimáticos/síntese química , Inibidores Enzimáticos/toxicidade , Humanos , Oligopeptídeos/síntese química , Oligopeptídeos/toxicidade , Complexo de Endopeptidases do Proteassoma/metabolismo , Relação Estrutura-Atividade
6.
Biosci Biotechnol Biochem ; 74(9): 1913-9, 2010.
Artigo em Inglês | MEDLINE | ID: mdl-20834157

RESUMO

TP-110, a novel proteasome inhibitor, has been found to possess potent growth inhibition in human multiple myeloma cells. To enhance its therapeutic effects, we established TP-110-resistant RPMI-8226 (RPMI-8226/TP-110) cells and elucidated their resistance mechanisms. The IC50 value for TP-110 cytotoxicity in the RPMI-8226/TP-110 cells was about 10-fold higher than that of the parental sensitive cells. The RPMI-8226/TP-110 cells exhibited distinct drug resistance to other proteasome inhibitors. Furthermore, they showed high cross-resistance to the cytotoxic effects of doxorubicin, etoposide, taxol, and vincristine. P-glycoprotein (MDR1), encoded by ABCB1, was elevated in the RPMI-8226/TP-110 cells, and the MDR1 inhibitor verapamil overcame their resistance to TP-110. The results of DNA microarray and RT-PCR suggested that the expression of ABCB1 is significantly elevated in RPMI-8226/TP-110 cells. This indicates that resistance in RPMI-8226/TP-110 cells is involved in the expression of P-glycoprotein, a drug-efflux pump.


Assuntos
Membro 1 da Subfamília B de Cassetes de Ligação de ATP/biossíntese , Resistencia a Medicamentos Antineoplásicos , Mieloma Múltiplo/tratamento farmacológico , Oligopeptídeos/farmacologia , Subfamília B de Transportador de Cassetes de Ligação de ATP , Membro 1 da Subfamília B de Cassetes de Ligação de ATP/análise , Antineoplásicos/farmacologia , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Resistencia a Medicamentos Antineoplásicos/genética , Perfilação da Expressão Gênica , Regulação Neoplásica da Expressão Gênica , Humanos , Concentração Inibidora 50 , Mieloma Múltiplo/química , Mieloma Múltiplo/metabolismo , Inibidores de Proteassoma , Células Tumorais Cultivadas , Verapamil/farmacologia
7.
Cancer Sci ; 100(1): 150-7, 2009 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-19018764

RESUMO

Because stroma in tumor tissues can promote prostate cancer development, modulation of tumor-stromal cell interactions may represent an attractive new strategy for cancer treatment. Here, we report that phthoxazolin A and its analog inthomycin B inhibit the growth of human prostate cancer DU-145 cells by modulating tumor-stromal cell interactions. Using an in vitro coculture system, in which prostate cancer cell growth is upregulated by prostate stromal cells (PrSC), we found that phthoxazolin A and inthomycin B strongly inhibited the growth of DU-145 cells when in coculture with PrSC compared to DU-145 cells cultured alone. Although PrSC consist of both fibroblasts and myofibroblasts, phthoxazolin A and inthomycin B inhibited the expression of smooth muscle alpha-actin, a myofibroblast marker, without affecting vimentin and beta-actin expression. Because myofibroblasts secrete various factors that can promote tumor cell growth, we examined whether the inhibitory compounds affected the secretion of such factors from PrSC. Proteomic analysis and reverse transcription-polymerase chain reaction revealed that phthoxazolin A and inthomycin B inhibited the expression of several insulin-like growth factor binding proteins and insulin-like growth factor (IGF)-I by PrSC. Transforming growth factor-beta1 increased myofibroblast numbers and IGF-I levels in PrSC. Phthoxazolin A inhibited transforming growth factor-beta1 activity without altering phosphorylation of the downstream molecule smad2. Furthermore, conditioned medium from phthoxazolin A-treated PrSC failed to increase the phosphorylation of IGF-IR and Akt in DU-145 cells. Taken together, our results suggested that phthoxazolin A acts as a small-molecule modulator of tumor-stromal cell interactions that can indirectly suppress prostate cancer cell growth through inhibition of IGF-I production by PrSC.


Assuntos
Antineoplásicos/farmacologia , Comunicação Celular/efeitos dos fármacos , Álcoois Graxos/farmacologia , Oxazóis/farmacologia , Alcamidas Poli-Insaturadas/farmacologia , Neoplasias da Próstata/tratamento farmacológico , Linhagem Celular Tumoral , Humanos , Fator de Crescimento Insulin-Like I/antagonistas & inibidores , Fator de Crescimento Insulin-Like I/fisiologia , Masculino , Neoplasias da Próstata/patologia , Células Estromais/efeitos dos fármacos , Células Estromais/fisiologia , Fator de Crescimento Transformador beta1/fisiologia
8.
Biochem Biophys Res Commun ; 380(1): 171-6, 2009 Feb 27.
Artigo em Inglês | MEDLINE | ID: mdl-19166817

RESUMO

Chronic deprivation of nutrients is rare in normal tissues, however large areas of tumor are nutrient-starved and hypoxic due to a disorganized vascular system. Some cancers show an inherent ability to tolerate severe growth conditions. Therefore, we screened chemical compounds to identify cytotoxic agents that function preferentially in nutrient-deprived conditions. We found that AG1024, a specific inhibitor of insulin-like growth factor-1 receptor tyrosine kinase (IGF-1R), showed preferential cytotoxicity to human pancreatic cancer cells in nutrient-deprived conditions relative to cells in nutrient-sufficient conditions. The cytotoxicity of I-OMe-AG538 (another specific inhibitor of IGF-1R kinase) was also enhanced in nutrient-deprived cells. In addition, AG1024 and I-OMe-AG538 potently inhibited IGF-1R activation to nutrient-deprived cells. In contrast, conventional chemotherapeutic drugs, as well as inhibitors of PDGFR and EGFR kinases, elicited weak cytotoxicity. These data indicate that nutrient-deprived human pancreatic cancer cells have increased sensitivity to inhibition of IGF-1R activation. IGF-1R inhibitors offer a promising strategy for anticancer therapeutic approaches that are oriented toward tumor microenvironment.


Assuntos
Antineoplásicos/farmacologia , Neoplasias Pancreáticas/enzimologia , Inibidores de Proteínas Quinases/farmacologia , Receptor IGF Tipo 1/antagonistas & inibidores , Tirfostinas/farmacologia , Antineoplásicos/química , Antineoplásicos/isolamento & purificação , Sobrevivência Celular/efeitos dos fármacos , Ensaios de Seleção de Medicamentos Antitumorais , Humanos , Inibidores de Proteínas Quinases/química , Inibidores de Proteínas Quinases/isolamento & purificação , Tirfostinas/química
9.
Bioorg Med Chem Lett ; 19(8): 2343-5, 2009 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-19307118

RESUMO

Boronic acid derivatives of tyropeptin were synthesized with TP-110 as the lead compound. Due to the lability of the aminoboronic acid moiety, careful design of the deprotection and coupling sequence was required. Liquid-liquid partition chromatography was found to be a powerful tool for purification of compounds of this class. The obtained derivatives showed potent inhibitory activities against the human 20S proteasome in vitro.


Assuntos
Ácidos Borônicos/síntese química , Dipeptídeos/síntese química , Inibidores de Proteases/síntese química , Inibidores de Proteassoma , Ácidos Borônicos/farmacologia , Linhagem Celular Tumoral , Dipeptídeos/farmacologia , Humanos , Oligopeptídeos/química , Oligopeptídeos/farmacologia , Inibidores de Proteases/farmacologia , Complexo de Endopeptidases do Proteassoma/metabolismo
10.
Anticancer Res ; 29(4): 977-85, 2009 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-19414335

RESUMO

TP-110, a new proteasome inhibitor, has previously shown potent growth inhibition in various tumor cell lines. In this study, the mechanism of TP-110-induced apoptosis is investigated in a human multiple myeloma cell line. Treatment with TP-110 for 24 h in vitro induced apoptosis in multiple myeloma cell line RPMI8226. Although the expression of Bcl-2, Bcl-xL and Bax was not affected by the treatment of TP-110, cleavage of Bid and release of cytochrome c were enhanced. Interestingly, TP-110 reduced the intrinsic inhibitor of apoptosis proteins (IAPs), cIAP-1 and XIAP, that suppress executioner caspases. The reduction of IAPs was observed not only by TP-110, but also by another proteasome inhibitor, MG-132. These results indicate that proteasome inhibitors reduce the level of IAPs and that the apoptosis induced by TP-110 is correlated with the level of IAPs in leukemia cell lines. Additionally, a reduction of cIAP-1 and XIAP by TP-110 contributes to the sensitization of Fas-mediated apoptosis. Taken together, the alteration of the apoptosis regulatory proteins by a proteasome inhibitor induces apoptosis in tumor cells.


Assuntos
Apoptose/efeitos dos fármacos , Proteínas Inibidoras de Apoptose/metabolismo , Mieloma Múltiplo/tratamento farmacológico , Oligopeptídeos/farmacologia , Inibidores de Proteassoma , Caspases/metabolismo , Citocromos c/metabolismo , Regulação para Baixo , Humanos , Immunoblotting , Proteínas Inibidoras de Apoptose/antagonistas & inibidores , Mieloma Múltiplo/metabolismo , Mieloma Múltiplo/patologia , Poli(ADP-Ribose) Polimerases/metabolismo , Células Tumorais Cultivadas
11.
Anticancer Res ; 28(2A): 721-30, 2008.
Artigo em Inglês | MEDLINE | ID: mdl-18507013

RESUMO

Insulin-like growth factor-I (IGF-I) secreted from the prostate stroma mediates tumor-stromal cell interactions in prostate cancer development. We have recently reported that human prostate stromal cells (PrSC) stimulate human prostate cancer DU-145 cell growth via IGF-I. Transforming growth factor-beta1 (TGF-beta1) also plays a critical role in tumor-stromal cell interactions, but TGF-beta1 has pleiotropic effects as it can modulate growth of prostate cancer either positively or negatively. To elucidate the complex behavior of TGF-beta1, the effect of TGF-beta1 on the IGF axis in PrSC was studied. TGF-beta1 increased the expression of IGF-I and IGF binding protein-3 (IGFBP-3). RT-PCR experiments revealed that TGF-beta1 upregulated the mRNA expression of IGF-I and IGFBP-3. However, while TGF-beta1 significantly increased IGFBP-3 secretion by PrSC, it conversely reduced the amounts of biologically active IGF-I unbound to IGFBP-3. Immunohistochemical analyses of 49 human prostate cancer tissues showed that IGF-I expression in the stroma correlated positively with TGF-beta1 expression in the stroma (r = 0.551, p = 0.0001). Furthermore, TGF-beta1 actually suppressed the growth of DU-145 cells in coculture with PrSC. But, IGFBP-3 proteinases, such as prostate specific antigen (PSA) and matrix metalloproteinase-7 (MMP-7), restored the DU-145 cell growth, suggesting that degradation of IGFBP-3 potentiates cancer growth. Taken together, these results indicated that TGF-beta1 modulates the growth of prostate cancer, either positively or negatively, through the balance between the amounts of IGF-I and IGFBP-3. This complex behavior of TGF-beta1 on the IGF axis is one explanation for the pleiotropic activities of TGF-beta1 on the growth of prostate cancer.


Assuntos
Fator de Crescimento Insulin-Like I/metabolismo , Neoplasias da Próstata/metabolismo , Fator de Crescimento Transformador beta1/farmacologia , Diferenciação Celular , Linhagem Celular Tumoral , Técnicas de Cocultura , Humanos , Proteína 3 de Ligação a Fator de Crescimento Semelhante à Insulina , Proteínas de Ligação a Fator de Crescimento Semelhante a Insulina/metabolismo , Masculino , Células Estromais/metabolismo
12.
J Antibiot (Tokyo) ; 61(7): 442-8, 2008 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-18776656

RESUMO

Angiogenesis is a critical step for the tumor therapy. Many angiogenic factors are involved in the tumor angiogenesis. In the course of our screening for inhibitors of angiogenin secretion, one of angiogenic factors, we have isolated a new terrein glucoside (1) and terrein (2) from the fermentation broth of fungal strain Aspergillus sp. PF1381. The structure and absolute stereochemistry of 1 was determined to be (4S,5R)-5-[(alpha-D-glucopyranosyl)oxy]-4-hydroxy-3-(E-1-propenyl)-2-cyclopenten-1-one on the basis of spectral and enzymatic analyses. Compounds 1 and 2 equally inhibited angiogenin secretion from androgen-dependent prostate cancer cells, LNCaP-CR, with IC50 values of 13 microM. However, both compounds did not affect VEGF secretion, another angiogenic factor. Furthermore, both compounds inhibited tube formation of human umbilical vein endothelial cells (HUVEC). These results suggested that 1 and 2 act as angiogenesis inhibitors through the inhibition of angiogenin secretion.


Assuntos
Antineoplásicos/química , Antineoplásicos/farmacologia , Ciclopentanos/química , Ciclopentanos/farmacologia , Glucosídeos/química , Glucosídeos/farmacologia , Ribonuclease Pancreático/antagonistas & inibidores , Ribonuclease Pancreático/metabolismo , Antineoplásicos/isolamento & purificação , Aspergillus/crescimento & desenvolvimento , Aspergillus/metabolismo , Linhagem Celular Tumoral , Meios de Cultura/química , Ciclopentanos/isolamento & purificação , Células Endoteliais , Glucosídeos/isolamento & purificação , Humanos , Concentração Inibidora 50
13.
Cancer Res ; 66(8): 4419-25, 2006 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-16618768

RESUMO

Prostate cancer shows high expression of type I insulin-like growth factor (IGF-I) receptor (IGF-IR) and prostate stromal cells (PrSC) produce IGF-I. Although high plasma level of IGF-I is a risk factor of prostate cancer, the significance of the prostate stromal IGF-I in the regulation of prostate cancer remains elusive. Here we show that the stromal IGF-I certainly regulates the development of prostate cancer. Coinoculation of PrSC increased the growth of human prostate cancer LNCaP and DU-145 tumors in severe combined immunodeficient mice. The conditioned medium of PrSC, as well as IGF-I, induced phosphorylation of IGF-IR and increased the growth of LNCaP and DU-145 cells. PrSC, but not LNCaP and DU-145 cells, secreted significant amounts of IGF-I. Coculture with PrSC increased the growth of DU-145 cells in vitro but the pretreatment of PrSC with small interfering RNA of IGF-I did not enhance it. Furthermore, various chemical inhibitors consisting of 79 compounds with approximately 60 different targets led to the finding that only IGF-IR inhibitor suppressed the PrSC-induced growth enhancement of DU-145 cells. Thus, these results show that the prostate stromal IGF-I mediates tumor-stromal cell interactions of prostate cancer to accelerate tumor growth, supporting the idea that the IGF-I signaling is a valuable target for the treatment of prostate cancer.


Assuntos
Comunicação Celular/fisiologia , Fator de Crescimento Insulin-Like I/fisiologia , Neoplasias da Próstata/patologia , Processos de Crescimento Celular/fisiologia , Linhagem Celular Tumoral , Técnicas de Cocultura , Humanos , Fator de Crescimento Insulin-Like I/biossíntese , Fator de Crescimento Insulin-Like I/metabolismo , Masculino , Próstata/citologia , Próstata/metabolismo , Neoplasias da Próstata/metabolismo , Transdução de Sinais , Células Estromais/metabolismo , Células Estromais/patologia
14.
J Antibiot (Tokyo) ; 71(3): 363-371, 2018 03.
Artigo em Inglês | MEDLINE | ID: mdl-29348522

RESUMO

To overcome serious methicillin-resistant Staphylococcus aureus (MRSA) and Pseudomonas aeruginosa infections, we synthesized TS2037, 5,4″-diepi-arbekacin, a novel aminoglycoside antibiotic, and evaluated its biological properties. TS2037 showed broad-range, as well as robust antibacterial activities against Gram-positive and Gram-negative bacteria. The MIC50 and MIC90 of TS2037 against clinical isolates of MRSA (n = 54) were both 0.25 µg/mL, and no resistant strain was observed. The MIC50 and MIC90 of TS2037 against clinical isolates of P. aeruginosa (n = 54) were 1 and 4 µg/mL, respectively. TS2037 and arbekacin, anti-MRSA aminoglycoside, were more stable against AAC(6')-APH(2″), aminoglycoside-6'-N-acetyltransferase and 2″-O-phosphotransferase, produced by resistant S. aureus than gentamicin. Therapeutic efficacies of TS2037 in the mouse models of systemic infection with MRSA were superior to those of arbekacin, vancomycin, and linezolid. The efficacy of TS2037 against systemic infection caused by P. aeruginosa producing AAC(6')-II was superior to those of arbekacin and amikacin. In the nephrotoxicity risk screening, the release of free N-acetyl-ß-D-glucosaminidase from the kidney epithelial cell line after treatment with TS2037 at 2.5 and 5.0 µM were 2.0 and 2.1 (U/L), respectively, which were about two times higher than those of arbekacin. In conclusion, TS2037 exhibited the most potent antibacterial activity among aminoglycosides tested against both MRSA and P. aeruginosa in vitro and in vivo, although its nephrotoxicity risk remains to be improved.


Assuntos
Aminoglicosídeos/farmacologia , Antibacterianos/farmacologia , Aminoglicosídeos/química , Aminoglicosídeos/toxicidade , Animais , Antibacterianos/toxicidade , Linhagem Celular , Dibecacina/análogos & derivados , Dibecacina/farmacologia , Dibecacina/toxicidade , Farmacorresistência Bacteriana/efeitos dos fármacos , Células Epiteliais/efeitos dos fármacos , Canamicina Quinase/metabolismo , Nefropatias/induzido quimicamente , Staphylococcus aureus Resistente à Meticilina/efeitos dos fármacos , Camundongos , Infecções por Pseudomonas/tratamento farmacológico , Infecções por Pseudomonas/microbiologia , Pseudomonas aeruginosa/efeitos dos fármacos , Infecções Estafilocócicas/tratamento farmacológico , Infecções Estafilocócicas/microbiologia
15.
Biochim Biophys Acta ; 1722(3): 247-53, 2005 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-15777622

RESUMO

Megalin, a family of endocytic receptors related to the low-density lipoprotein (LDL) receptor, is a major pathway for proximal tubular aminoglycoside accumulation. We previously reported that aminoglycoside antibiotics reduce SGLT1-dependent glucose transport in pig proximal tubular epithelial LLC-PK1 cells in parallel with the order of their nephrotoxicity. In this study, using a model of gentamicin C (GMC)-induced reduction in SGLT1 activity, we examined whether ligands for megalin protect LLC-PK1 cells from the GMC-induced reduction in SGLT1 activity. We employed apolipoprotein E3 (apoE3) and lactoferrin as ligands for megalin. Then the cells were treated with various concentrations of apoE3, lactoferrin and bovine serum albumin with or without 100 microg/ml of GMC, and the SGLT1-dependent methyl alpha-D-glucopyranoside (AMG) uptake and levels of SGLT1 expression were determined. As a result, we demonstrated that the apoE3 significantly protects these cells from GMC-induced reduction in AMG uptake, but neither lactoferrin nor albumin does. In accord with a rise in AMG uptake activity, the mRNA and protein levels of SGLT1 were apparently up-regulated in the presence of apoE3. Furthermore, we found that the uptake of [3H] gentamicin is decreased by apoE3, and that apoE3 showed obvious protection against the GMC-dependent N-acetyl-beta-D-glucosamidase (NAG) release from LLC-PK1 cells. Thus, these results indicate that apoE3 could be a valuable tool for the prevention of aminoglycoside nephrotoxicity.


Assuntos
Apolipoproteínas E/fisiologia , Gentamicinas/farmacologia , Túbulos Renais Proximais/efeitos dos fármacos , Células LLC-PK1/fisiologia , Glicoproteínas de Membrana/metabolismo , Proteínas de Transporte de Monossacarídeos/metabolismo , Animais , Apolipoproteína E3 , Sequência de Bases , Primers do DNA , Túbulos Renais Proximais/metabolismo , Transportador 1 de Glucose-Sódio , Suínos
16.
Cancer Lett ; 242(1): 46-52, 2006 Oct 08.
Artigo em Inglês | MEDLINE | ID: mdl-16377078

RESUMO

Human androgen-dependent prostate cancer LNCaP cells are low tumorigenic even in immunodeficient mice and were killed by the synergistic effect of inflammatory cytokines, IL-beta and IL-6. To establish a highly tumorigenic LNCaP cell line, we isolated the cytokine-resistant LNCaP-CR cell line and examined the phenotypes. The parental LNCaP cells were induced to commit apoptosis by the addition of IL-1beta and IL-6, but LNCaP-CR cells showed strong resistance against the cytokine action. However, LNCaP-CR cells did not exhibit any resistance to various antitumor drugs investigated. While LNCaP cells formed only palpable tumors in SCID mice, LNCaP-CR cells readily made tumors and their growth was significantly higher than that of LNCaP cells. Moreover, LNCaP tumor-bearing mice gained the weight gradually, but LNCaP-CR tumor-bearing mice significantly lost their body weight. LNCaP-CR cells still responded to androgen action and expressed AR, erbB2, IL-1R, IL-6R, gp130, STAT3, p21, Bcl-2 and caspase-3 as well as LNCaP cells. These results indicate that LNCaP-CR cell line is a new type of tumorigenic LNCaP cell lines and should be useful for identifying responsible genes of tumorigenicity, cytokine resistance, and also cachexia.


Assuntos
Linhagem Celular Tumoral , Citocinas/metabolismo , Regulação Neoplásica da Expressão Gênica , Neoplasias da Próstata/tratamento farmacológico , Animais , Fragmentação do DNA , Resistencia a Medicamentos Antineoplásicos , Humanos , Inflamação , Interleucina-1beta/metabolismo , Interleucina-6/metabolismo , Masculino , Camundongos , Camundongos SCID , Fenótipo , Neoplasias da Próstata/patologia
17.
J Antibiot (Tokyo) ; 59(11): 693-7, 2006 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-17256467

RESUMO

Hypoxia-inducible factor-1 (HIF-1) is a central mediator of cellular responses to low oxygen and has recently become an important therapeutic target for solid tumor therapy. To identify small molecule inhibitors of the HIF-1 transcriptional activation, we have established a high through-put assay system using a stable transformant of mammalian cells that express a luciferase reporter gene construct containing a HIF-1 binding site. Using this system, we screened 5000 cultured broths of microorganisms, and we found that fermentation broth produced by Streptomyces strain 1759-27 showed significant inhibition of the reporter activity induced by hypoxic conditions. The active substance NBRI759-27 was purified and determined to be tartrolone C by several methods including X-ray crystallography. In the reporter gene assay, tartrolone C inhibited the HIF-1 transcriptional activity under hypoxic conditions with an IC50 value of 0.17 microg/ml.


Assuntos
Subunidade alfa do Fator 1 Induzível por Hipóxia/antagonistas & inibidores , Macrolídeos/farmacologia , Animais , Western Blotting , Células CHO , Hipóxia Celular , Linhagem Celular Tumoral , Cricetinae , Cricetulus , Cristalografia por Raios X , Humanos , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Concentração Inibidora 50 , Macrolídeos/química , Macrolídeos/isolamento & purificação , Modelos Moleculares , Ressonância Magnética Nuclear Biomolecular , Rotação Ocular , Espectrometria de Massas por Ionização por Electrospray , Espectrofotometria Ultravioleta , Streptomyces/química , Streptomyces/metabolismo
18.
J Antibiot (Tokyo) ; 59(4): 215-9, 2006 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-16830888

RESUMO

Kigamicin D did not show any immunosuppressive activity in mixed lymphocyte culture reaction and mitogen induced lymphocyte blastogenesis in vitro and graft versus host reaction in vivo. Natural killer cell activity in spleen cells was not affected by oral administration of kigamicin D. Instead, delayed-type hypersensitivity response to sheep red blood cells was stimulated at a broad dosage level. It is concluded that kigamicin D increases cellular immunity to specific antigen.


Assuntos
Doxorrubicina/análogos & derivados , Imunidade Celular/efeitos dos fármacos , Oxazóis/farmacologia , Animais , Células Cultivadas , Doxorrubicina/farmacologia , Reação Enxerto-Hospedeiro/efeitos dos fármacos , Hipersensibilidade Tardia , Células Matadoras Naturais/efeitos dos fármacos , Ativação Linfocitária/efeitos dos fármacos , Teste de Cultura Mista de Linfócitos , Camundongos , Camundongos Endogâmicos C3H , Modelos Animais , Baço/imunologia
19.
J Antibiot (Tokyo) ; 59(4): 209-14, 2006 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-16830887

RESUMO

Kigamicin D is a novel anticancer agent that was identified using a new screening strategy that targets the tolerance of cancer cells to nutrient starvation [1, 2]. Oral administration of kigamicin D was previously described to show a strong antitumor effect in human tumor xenograft models of pancreatic tumors [2]. In this paper we describe that kigamicin D shows the same selective cytotoxicity against normal human cells such as lung fibroblast and prostate stromal cells under nutrient starved condition as against cancer cells. Kigamicin D inhibited tumor cell-induced angiogenesis in a dorsal air sac assay. On the basis of these results we tested other human tumor xenograft models and transplantable syngeneic tumor models in order to determine the spectrum of activity of kigamicin D against various cancers. Kigamicin D showed a weak antitumor effect against LX-1 and DMS-273 lung cancers, but had no effect on DLD-1 colon cancers. When tested against syngeneic tumors, kigamicin D showed a weak antitumor effect against colon26, but showed augmentation of tumor growth on IMC carcinoma at a broad dosage level. Kigamicin D does not show good antitumor activity against human xenograft tumors except pancreatic tumors and murine syngeneic tumors. We found that kigamicin D has excellent antitumor effect specific to pancreatic cancers. Surprisingly, high dosage of kigamicin D increased tumor growth of IMC carcinoma by than 200%. The phenomenon suggests that kigamicin D may cause some immunological response to the tumor.


Assuntos
Antibióticos Antineoplásicos/uso terapêutico , Doxorrubicina/análogos & derivados , Neoplasias/tratamento farmacológico , Oxazóis/uso terapêutico , Animais , Antibióticos Antineoplásicos/farmacologia , Linhagem Celular Tumoral , Neoplasias do Colo/tratamento farmacológico , Modelos Animais de Doenças , Doxorrubicina/farmacologia , Doxorrubicina/uso terapêutico , Feminino , Humanos , Neoplasias Pulmonares/tratamento farmacológico , Camundongos , Camundongos Endogâmicos BALB C , Transplante de Neoplasias , Neoplasias/patologia , Neovascularização Patológica/prevenção & controle , Oxazóis/farmacologia , Neoplasias Pancreáticas/tratamento farmacológico , Ensaios Antitumorais Modelo de Xenoenxerto
20.
Int Immunopharmacol ; 5(2): 281-8, 2005 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-15652759

RESUMO

Deficiency of Fas expression is one of mechanisms involved in the immune evasion by tumors. Several antitumor drugs, such as doxorubicin (DOX) increase Fas expression in tumor cells and sensitize the cells to Fas-mediated apoptosis in vitro. However, the significance of the Fas expression in vivo is still unclear. Therefore, we examined a role of Fas expression on antitumor effect of DOX using a syngeneic tumor model of Lewis lung carcinoma (3LL) cells in C57BL/6-gld mice that lack functional Fas ligand (FasL). In vitro, anti-Fas agonistic antibody, Jo2, did not decrease a viable cell number of 3LL cells in the absence of DOX, whereas it significantly reduced the cell viability in the presence of DOX. The treatment with DOX alone at the same dose did not induce cell death. Flowcytometric analysis of Fas expression revealed that 3LL cells expressed only a marginal amount of Fas, but the treatment of the cells with DOX increased the expression of Fas in the cell surface. When splenic T cells were prepared from 3LL-bearing C57BL/6 mice, the splenic T cells significantly killed DOX-pretreated 3LL cells more than untreated 3LL cells. In the syngeneic models, DOX inhibited growth of 3LL solid tumor both in wild-type C57BL/6 mice and in Fas-deficient C57BL/6-lpr mice, but it failed in C57BL/6-gld mice, suggesting that the interaction between host FasL and tumor Fas is involved in the antitumor effect of DOX. Furthermore, Fas expression was increased in the solid tumor by the treatment of DOX. These results suggest that the antitumor effect of DOX is partly exerted by the Fas expression and host immune defense.


Assuntos
Antibióticos Antineoplásicos/farmacologia , Apoptose/efeitos dos fármacos , Carcinoma Pulmonar de Lewis/imunologia , Doxorrubicina/farmacologia , Receptor fas/biossíntese , Animais , Antibióticos Antineoplásicos/uso terapêutico , Apoptose/imunologia , Carcinoma Pulmonar de Lewis/tratamento farmacológico , Carcinoma Pulmonar de Lewis/metabolismo , Carcinoma Pulmonar de Lewis/patologia , Linhagem Celular Tumoral , Doxorrubicina/uso terapêutico , Proteína Ligante Fas , Feminino , Glicoproteínas de Membrana/biossíntese , Glicoproteínas de Membrana/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Transplante de Neoplasias , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Receptor fas/imunologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA