Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Int J Clin Oncol ; 25(9): 1563-1569, 2020 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-32656741

RESUMEN

DEK is a highly conserved nuclear factor that plays an important role in the regulation of multiple cellular processes. DEK was discovered to be an oncogene as a fusion with NUP214 gene, which results in producing DEK-NUP214 proteins, in a subset of patients with acute myeloid leukemia. Subsequently, DEK overexpression was reported in many cancers, thus DEK itself is considered to be an oncoprotein. DEK has been reported to play important roles in the progression of early and late stage squamous cell carcinoma (SCC) and is useful for early diagnosis of the disease. These findings have made DEK an attractive therapeutic target, especially for human papillomavirus (HPV)-associated SCC. However, the mechanism of DEK in SCC remains unclear. In this review, we discuss human DEK oncogene-related SCC.


Asunto(s)
Carcinoma de Células Escamosas/genética , Carcinoma de Células Escamosas/patología , Proteínas Cromosómicas no Histona/genética , Proteínas Oncogénicas/genética , Proteínas de Unión a Poli-ADP-Ribosa/genética , Carcinoma de Células Escamosas/diagnóstico , Regulación Neoplásica de la Expresión Génica , Humanos
2.
Oncol Rep ; 42(1): 176-188, 2019 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-31059083

RESUMEN

Tranilast [N­(3',4'­dimethoxycinnamoyl)­anthranilic acid], initially developed as an antiallergic drug, also exhibits a growth inhibitory effect on various types of cancer. Osteosarcoma is treated mainly with high­dose methotrexate, doxorubicin, cisplatin and ifosfamide; however, 20­30% of patients cannot be cured of metastatic disease. We investigated whether tranilast enhances the anticancer effects of chemotherapeutic drugs and analyzed its mechanism of action in osteosarcomas. Tranilast inhibited proliferation of HOS, 143B, U2OS and MG­63 osteosarcoma cells in a dose­dependent manner, as well as enhancing the effects of cisplatin and doxorubicin. The average combination index at effect levels for tranilast in combination with cisplatin was 0.57 in HOS, 0.4 in 143B, 0.39 in U2OS and 0.51 in MG­63 cells. Tranilast and cisplatin synergistically inhibited the viability of osteosarcoma cells. In flow cytometric analysis, although tranilast alone did not induce significant apoptosis, the combination of tranilast and cisplatin induced early and late apoptotic cell death. Expression of cleaved caspase­3, cleaved poly(ADP­ribose) polymerase and p­H2AX was enhanced by tranilast in combination with cisplatin. Tranilast alone increased expression of p21 and Bim protein in a dose­dependent manner. Cell cycle analysis using flow cytometry demonstrated that the combination of tranilast and cisplatin increased the number of cells in the G2/M phase. Compared with cisplatin alone, the combination increased levels of phospho­cyclin­dependent kinase 1 (Y15). In the 143B xenograft model, tumor growth was significantly inhibited by combined tranilast and cisplatin compared with the controls, whereas cisplatin alone did not significantly inhibit tumor growth. In conclusion, tranilast has a cytostatic effect on osteosarcoma cells and enhances the effect of anticancer drugs, especially cisplatin. Enhanced sensitivity to cisplatin was mediated by increased apoptosis through G2/M arrest. Since tranilast has been clinically approved and has few adverse effects, clinical trials of osteosarcoma chemotherapy in combination with tranilast are expected.


Asunto(s)
Antineoplásicos/administración & dosificación , Neoplasias Óseas/tratamiento farmacológico , Cisplatino/administración & dosificación , Osteosarcoma/tratamiento farmacológico , ortoaminobenzoatos/administración & dosificación , Animales , Antineoplásicos/farmacología , Caspasa 3/metabolismo , Ciclo Celular/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Cisplatino/farmacología , Relación Dosis-Respuesta a Droga , Sinergismo Farmacológico , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Ratones , Poli(ADP-Ribosa) Polimerasas/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto , ortoaminobenzoatos/farmacología
3.
Sci Rep ; 9(1): 7742, 2019 05 23.
Artículo en Inglés | MEDLINE | ID: mdl-31123329

RESUMEN

Phenotypic screening in drug discovery has been revived with the expectation of providing promising lead compounds and drug targets and improving the success rate of drug approval. However, target identification remains a major bottleneck in phenotype-based drug discovery. We identified the lead compounds K542 and K405 with a selective inhibition of cell viability against sphingosine-1-phosphate lyase 1 (SGPL1)-transduced ES-2 cells by phenotypic screening. We therefore performed an in vivo pharmacological examination and observed the antitumor activity of K542 in an HT-1080 tumor-bearing mouse xenograft model. SGPL1 was expected to be a therapeutic target in some cancers, suggesting that these lead molecules might be promising candidates; however, their mechanisms of action still remain unexplained. We therefore synthesized the affinity probe Ind-tag derived from K542 and identified the proteins binding to Ind-tag via a pull-down experiment. Proteomics and biochemical analyses revealed that the target molecule of these lead compounds was Nicotinamide phosphoribosyltransferase (NAMPT). We established K542-resistant DLD-1 and HT-1080 cells, and genetic analyses of these cells identified a missense mutation in the NAMPT-encoding gene. This enzymatic experiment clearly showed that K393 exerts enzymatic inhibition against NAMPT. These proteomics, genetics and biochemical analyses clarified that compounds K542 and K405 were NAMPT inhibitors.


Asunto(s)
Ensayos de Selección de Medicamentos Antitumorales/métodos , Nicotinamida Fosforribosiltransferasa/efectos de los fármacos , Nicotinamida Fosforribosiltransferasa/metabolismo , Aldehído-Liasas/efectos de los fármacos , Aldehído-Liasas/metabolismo , Animales , Antineoplásicos/farmacología , Línea Celular Tumoral , Citocinas/efectos de los fármacos , Citocinas/metabolismo , Descubrimiento de Drogas/métodos , Evaluación Preclínica de Medicamentos , Inhibidores Enzimáticos/farmacología , Humanos , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Modelos Moleculares , Neoplasias/tratamiento farmacológico , Fenotipo , Relación Estructura-Actividad
5.
Oral Oncol ; 73: 16-20, 2017 10.
Artículo en Inglés | MEDLINE | ID: mdl-28939069

RESUMEN

Oral squamous cell carcinoma (OSCC) patients have a low 5-year survival rate and poor prognosis. To improve survival and prognosis, the causes and processes involved in lesion development should be evaluated. For this purpose, the use of OSCC mouse models, such as chemically induced mouse models, genetically modified mouse models, and transplanted (xenograft) models, is crucial. These OSCC models exhibit both advantages and disadvantages when studying OSCC development and progression. Until a model resembling human OSCC is developed, both the advantages and disadvantages of each model should be carefully considered. In this review, we discuss OSCC mouse models and their use in cancer research worldwide.


Asunto(s)
Carcinoma de Células Escamosas/patología , Modelos Animales de Enfermedad , Neoplasias de la Boca/patología , Animales , Carcinógenos/toxicidad , Carcinoma de Células Escamosas/inducido químicamente , Carcinoma de Células Escamosas/genética , Progresión de la Enfermedad , Xenoinjertos , Humanos , Ratones , Ratones Transgénicos , Neoplasias de la Boca/inducido químicamente , Neoplasias de la Boca/genética
6.
Cancer Med ; 6(10): 2424-2439, 2017 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-28834425

RESUMEN

Oral squamous cell carcinoma (OSCC) develops through a multistep carcinogenic process involving field cancerization. The DEK gene is a proto-oncogene with functions in genetic and epigenetic modifications, and has oncogenic functions, including cellular proliferation, differentiation, and senescence. DEK overexpression is associated with malignancies; however, the functional roles of DEK overexpression are unclear. We demonstrated that DEK-expressing cells were significantly increased in human dysplasia/carcinoma in situ and OSCC. Furthermore, we generated ubiquitous and squamous cell-specific doxycycline (DOX)-inducible Dek mice (iDek and iDek-e mice respectively). Both DOX+ iDek and iDek-e mice did not show differences in the oral mucosa compared with DOX- mice. In the environment exposed to carcinogen, DOX-treated (DOX+) iDek mice showed field cancerization and OSCC development. Microarray analysis revealed that DEK overexpression was mediated by the upregulation of DNA replication- and cell cycle-related genes, particularly those related to the G1 /S transition. Tongue tumors overexpressing DEK showed increased proliferating cell nuclear antigen and elongator complex protein 3 expression. Our data suggest that DEK overexpression enhanced carcinogenesis, including field cancerization, in OSCC by stimulating the G1 /S phase transition and promoting DNA replication, providing important insights into the potential applications of DEK as a target in the treatment and prevention of OSCC.


Asunto(s)
Carcinógenos/administración & dosificación , Carcinoma de Células Escamosas/etiología , Transformación Celular Neoplásica/genética , Proteínas Cromosómicas no Histona/genética , Neoplasias de la Boca/etiología , Proteínas Oncogénicas/genética , Proteínas de Unión a Poli-ADP-Ribosa/genética , Animales , Carcinoma de Células Escamosas/metabolismo , Carcinoma de Células Escamosas/patología , Línea Celular Tumoral , Proliferación Celular , Transformación Celular Neoplásica/metabolismo , Modelos Animales de Enfermedad , Células Madre Embrionarias/metabolismo , Expresión Génica , Humanos , Ratones , Modelos Biológicos , Neoplasias de la Boca/metabolismo , Neoplasias de la Boca/patología , Antígeno Nuclear de Célula en Proliferación/genética , Antígeno Nuclear de Célula en Proliferación/metabolismo , Proto-Oncogenes Mas
7.
Oncol Rep ; 38(2): 775-784, 2017 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-28627675

RESUMEN

Serrated lesions in the colorectum are currently predominantly classified as hyperplastic polyps (HPs), sessile serrated adenomas/polyps (SSA/Ps), and traditional serrated adenomas (TSAs) according to their morphology. However, the histological morphology and the molecular changes in the serrated lesions are still unclear. We performed immunohistochemistry for Ki67, p16INK4a, and WNT5A in human HPs (n=22), SSA/Ps (n=41), and TSAs (n=19). The distribution of Ki67 and p16INK4a positive cells in TSAs was different from that in HPs and SSA/Ps. Co-expression of Ki67 and P16INK4a was infrequent in HPs and SSA/Ps; p16INK4a-positive cells were found in the crypt cleft and stromal WNT5A-positive stromal cells were localized near the cleft in SSA/Ps, while intraepithelial lymphocytes (IELs) in SSA/Ps were more abundant than HPs. In conclusion, our study provides evidence that HPs branch because of the increase in and patchy distribution of senescent and proliferative cells, with increased and misdistributed stromal and inflammatory cells, which might contribute to creation of L- and/or T-shaped crypts, which are of distinctive shapes in SSA/Ps. Our findings may facilitate better understanding and therapy in the serrated lesions.


Asunto(s)
Adenoma/genética , Neoplasias Colorrectales/genética , Inhibidor p16 de la Quinasa Dependiente de Ciclina/genética , Antígeno Ki-67/genética , Proteína Wnt-5a/genética , Adenoma/patología , Anciano , Animales , Biomarcadores de Tumor/genética , Pólipos del Colon/genética , Pólipos del Colon/patología , Neoplasias Colorrectales/patología , Femenino , Regulación Neoplásica de la Expresión Génica/genética , Humanos , Inmunohistoquímica , Linfocitos Intraepiteliales/patología , Masculino , Persona de Mediana Edad
8.
Oncotarget ; 8(25): 41078-41090, 2017 Jun 20.
Artículo en Inglés | MEDLINE | ID: mdl-28467793

RESUMEN

Non-ampullary duodenal adenoma with activation of Wnt/ß-catenin signalling is common in familial adenomatous polyposis (FAP) patients, whereas sporadic non-ampullary adenoma is uncommon. The adenoma-carcinoma sequence similar to colon cancer is associated with duodenal tumors in FAP, but not always in sporadic tumors. We obtained 37 non-ampullary duodenal tumors, including 25 adenomas and 12 adenocarcinomas, were obtained from biopsies and endoscopic resections. We performed immunohistochemistry for ß-catenin, the hallmark of Wnt activation, and aldehyde dehydrogenase 1 (ALDH1), a putative cancer stem cell marker. In non-ampullary lesions, abnormal nuclear localization of ß-catenin was observed in 21 (84.0%) of 25 adenomas and 4 (33.3%) of 12 adenocarcinomas. In the proximal duodenum, nuclear ß-catenin was less frequent in both adenomas and adenocarcinomas. Gastric duodenal metaplasia (GDM) was observed only in the proximal duodenum. All adenomas with GDM were the gastric foveolar and pyloric gland types, and showed only membranous ß-catenin. The intestinal-type adenomas had nuclear ß-catenin in the proximal and distal duodenum. ALDH1-positive cells were more frequent in adenocarcinomas than adenomas. Nuclear ß-catenin accumulation frequently occurred in ALDH1-positive cells in adenoma, but not in adenocarcinoma. In the non-ampullary proximal duodenum, Wnt/ß-catenin pathway activation was more closely associated with adenomas than adenocarcinomas, and while it might cooperate with ALDH1 in adenoma, it does not in adenocarcinoma. The pathogenesis thus may differ between sporadic adenoma and adenocarcinoma of non-ampullary duodenal lesions, especially in the proximal and distal duodenum.


Asunto(s)
Adenocarcinoma/patología , Adenoma/patología , Neoplasias Duodenales/patología , Duodeno/patología , Adenocarcinoma/metabolismo , Adenoma/metabolismo , Adulto , Anciano , Familia de Aldehído Deshidrogenasa 1 , Neoplasias Duodenales/metabolismo , Duodeno/metabolismo , Femenino , Humanos , Inmunohistoquímica , Isoenzimas/biosíntesis , Masculino , Metaplasia , Persona de Mediana Edad , Retinal-Deshidrogenasa/biosíntesis , Vía de Señalización Wnt , beta Catenina/biosíntesis
9.
J Orthop Surg (Hong Kong) ; 25(1): 2309499016684096, 2017 01.
Artículo en Inglés | MEDLINE | ID: mdl-28176598

RESUMEN

Intraosseous epidermoid cysts are exceedingly rare. Known as pseudotumors, not true neoplasms, intraosseous epidermoid cysts usually involve the phalanges, the skull, and the toes. Intraosseous epidermoid cysts typically present as destructive osteolytic lesions on X-ray, mimicking malignant bone tumors. Here, we present two cases of an intraosseous epidermoid cyst in the distal phalanx treated with curettage and synthetic bone graft, followed by a review of the relevant literature. In both cases, the patient presented with a painful enlargement of the fingertip following a minor trauma. Magnetic resonance imaging demonstrated lesions involving the distal phalanx that had a low signal on T1-weighted imaging (WI) and a high intensity on T2-WI. In both cases, the lesions were not enhanced by gadolinium. Good remodeling and functional recoveries were obtained. For physically active patients with substantial bone defects, synthetic bone graft may be recommended.


Asunto(s)
Sustitutos de Huesos/uso terapéutico , Durapatita/uso terapéutico , Quiste Epidérmico/diagnóstico , Quiste Epidérmico/cirugía , Falanges de los Dedos de la Mano , Osteólisis/cirugía , Adulto , Quiste Epidérmico/complicaciones , Femenino , Humanos , Imagen por Resonancia Magnética , Masculino , Osteólisis/diagnóstico por imagen , Osteólisis/etiología , Adulto Joven
10.
Molecules ; 21(9)2016 Sep 03.
Artículo en Inglés | MEDLINE | ID: mdl-27598119

RESUMEN

Aromatic groups were incorporated into 9H-9-silafluorene units at the 9-position (mono-9H-silafluorenes) and 9,9'-positions (di-9H-9-silafluorenes). The aryl substituents showed weak conjugation to the 9H-9-silafluorene for 9-aryl substituted ones 1-7 and a 9,9'-phenylene substituted one (compound 8) and they exhibited similar absorption and emission spectra. The 9H-9-silafluorene 10 containing a 5,5'-(2,2'-bithiophenyl) group showed a significantly red-shifted absorption and fluorescence maxima in the solid-state. Single-crystal X-ray diffraction studies found J-type aggregated structures formed by intermolecular CH-π interactions (ca. 2.6-2.7 Å). Density functional theory (DFT), time-dependent DFT (TD-DFT), and configuration interaction single (CIS) calculations were conducted to explain the observed optical properties.


Asunto(s)
Fluorescencia , Colorantes Fluorescentes , Modelos Químicos , Compuestos de Organosilicio , Colorantes Fluorescentes/síntesis química , Colorantes Fluorescentes/química , Compuestos de Organosilicio/síntesis química , Compuestos de Organosilicio/química
11.
Oncotarget ; 6(28): 24722-32, 2015 Sep 22.
Artículo en Inglés | MEDLINE | ID: mdl-26160842

RESUMEN

Aldehyde dehydrogenase 1A1 (ALDH1A1) is considered to be a cancer stem cell marker in several human malignancies. However, the role of ALDH1A1 in hepatocellular carcinoma (HCC) has not been well elucidated. In this study, we investigated the relationship between ALDH1A1 and clinicopathological findings and examined whether ALDH1A1 deserves to be a cancer stem cell marker in HCC. Sixty HCC samples obtained from surgical resection were collected for immunohistochemical (IHC) staining. Of these 60 samples, 47 samples of HCC tumorous and non-tumorous tissues were evaluated with qRT-PCR. There was no significant difference in the ALDH1A1-mRNA level between tumorous and non-tumorous tissues. Tumorous ALDH1A1-mRNA level had no relationship with the clinicopathological features. Immunoreactivity of ALDH1A1 was classified into two groups based on the percentage of ALDH1A1-overexpressing cells. The ALDH1A1-high group was significantly associated with low serum levels of α-fetoprotein, small tumor diameter, very little lymphovascular invasion, more differentiated pathology and good stage. The ALDH1A1-high group showed more favorable prognosis for recurrence-free survival. In double-staining IHC, ALDH1A1 was not co-expressed with BMI1, EpCAM, CD13, CD24, CD90 and CD133, which reported as cancer stem cell markers in HCC. In conclusion, ALDH1A1-overexpressing cells could appear to be differentiated cells rather than cancer stem cells in HCC.


Asunto(s)
Aldehído Deshidrogenasa/análisis , Biomarcadores de Tumor/análisis , Carcinoma Hepatocelular/enzimología , Diferenciación Celular , Neoplasias Hepáticas/enzimología , Células Madre Neoplásicas/enzimología , Adulto , Anciano , Anciano de 80 o más Años , Aldehído Deshidrogenasa/genética , Familia de Aldehído Deshidrogenasa 1 , Biomarcadores de Tumor/genética , Carcinoma Hepatocelular/genética , Carcinoma Hepatocelular/mortalidad , Carcinoma Hepatocelular/patología , Carcinoma Hepatocelular/terapia , Supervivencia sin Enfermedad , Femenino , Regulación Enzimológica de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Humanos , Inmunohistoquímica , Estimación de Kaplan-Meier , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/mortalidad , Neoplasias Hepáticas/patología , Neoplasias Hepáticas/terapia , Masculino , Persona de Mediana Edad , Análisis Multivariante , Células Madre Neoplásicas/patología , Fenotipo , Modelos de Riesgos Proporcionales , ARN Mensajero/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Retinal-Deshidrogenasa , Factores de Riesgo , Regulación hacia Arriba
12.
Int J Oncol ; 46(3): 963-72, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25530098

RESUMEN

Shikonin (SHK), a natural small agent (MW 288.3), reportedly induces cell death in various tumor cells. We have found that SHK also exerts potent cytocidal effects on human multiple myeloma (MM) cells, but its anticancer mechanism in MM cells remains to be elucidated. SHK at 2.5-5 µM induced apoptosis in seven MM cell lines, including the bortezomib-resistant cell line KMS11/BTZ. The IC50 value of SHK against KMS11/BTZ was comparable to that of a parental cell line KMS11 (1.1 and 1.56 µM, respectively). SHK induces accumulation of ubiquitinated proteins and activates XBP-1 in MM cells, suggesting that SHK functions as a proteasome inhibitor, eventually inducing ER stress-associated apoptosis. SHK increases levels of HSP70/72, which protects cells from apoptosis, and exerts greater cytocidal effects in combination with the HSP70/72 inhibitor VER-155008. At higher concentrations (10-20 µM), SHK induced cell death, which was completely inhibited by a necroptosis inhibitor, necrostatin-1 (Nec-1), while the cytocidal activity was unaffected by Z-VAD-FMK, strongly suggesting that cell death is induced by SHK at high concentrations through necroptosis. The present data show for the first time that SHK induces cell death in MM cells. SHK efficiently induces apoptosis and combination of heat shock protein inhibitor with low dose SHK enhances apoptosis, while high dose SHK induces necroptosis in MM cells. These findings together support the use of SHK as a potential therapeutic agent for MM.


Asunto(s)
Antineoplásicos/farmacología , Mieloma Múltiple/tratamiento farmacológico , Mieloma Múltiple/patología , Naftoquinonas/farmacología , Inhibidores de Proteasoma/farmacología , Antineoplásicos/administración & dosificación , Apoptosis/efectos de los fármacos , Ácidos Borónicos/farmacología , Bortezomib , Muerte Celular/efectos de los fármacos , Línea Celular Tumoral/efectos de los fármacos , Proteínas de Unión al ADN/genética , Relación Dosis-Respuesta a Droga , Resistencia a Antineoplásicos/efectos de los fármacos , Estrés del Retículo Endoplásmico/efectos de los fármacos , Humanos , Imidazoles/farmacología , Indoles/farmacología , Naftoquinonas/administración & dosificación , Necrosis/patología , Nucleósidos de Purina/farmacología , Pirazinas/farmacología , Factores de Transcripción del Factor Regulador X , Factores de Transcripción/genética , Proteína 1 de Unión a la X-Box
13.
Immunopharmacol Immunotoxicol ; 36(5): 316-28, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-25077632

RESUMEN

CONTEXT: Serum IgG, IgE and IgM have been shown to enhance the primary antibody responses upon exposure to the soluble antigens recognized by those antibodies. However, how IgA affects these responses remains unknown. OBJECTIVE: We investigated the effects of intravenously administered monoclonal IgA on the immune responses in mice. MATERIALS AND METHODS: DBA/1J mice were immunized with ovalbumin in the presence or absence of anti-ovalbumin monoclonal IgA. The Th1 and Th2 immune responses to ovalbumin and the anaphylaxis induced by re-exposure to ovalbumin were measured. RESULTS: IgA complexed with antigen attenuated the primary antibody responses to the antigen in mice, in contrast to IgG2b and IgE. The primary antibody responses, i.e. the de novo synthesis of anti-ovalbumin IgG2a, IgG1 and IgE in the serum, and the subsequent anaphylaxis induced with re-exposure to ovalbumin were reduced by the co-injection of anti-ovalbumin monoclonal IgA at ovalbumin immunization. The Th1, Th2 and Tr1 cytokines interferon-γ, interleukin-4 and interleukin-10, respectively, released from ovalbumin-restimulated cultured splenocytes collected from allergic mice were also reduced by the treatment. The induction of interferon-γ and interleukin-4 secretion by splenocytes from ovalbumin-immunized mice stimulated in vitro with ovalbumin was also significantly reduced by the antigen complexed with anti-ovalbumin IgA. CONCLUSION: These data suggest that the direct inhibition of Th1 and Th2 activation by anti-ovalbumin monoclonal IgA participates in the inhibition of the primary antibody responses. IgA plays important immunosuppressive roles under physiological and pathological conditions and is a promising candidate drug for the treatment of immune disorders.


Asunto(s)
Anafilaxia/prevención & control , Formación de Anticuerpos/efectos de los fármacos , Complejo Antígeno-Anticuerpo/farmacología , Inmunoglobulina A/farmacología , Inmunoglobulinas Intravenosas/farmacología , Células TH1/inmunología , Células Th2/inmunología , Animales , Complejo Antígeno-Anticuerpo/administración & dosificación , Células Cultivadas , Epítopos/inmunología , Femenino , Inmunoglobulina A/administración & dosificación , Inmunoglobulinas Intravenosas/administración & dosificación , Activación de Linfocitos/efectos de los fármacos , Ratones , Ratones Endogámicos DBA , Ovalbúmina/inmunología , Células TH1/efectos de los fármacos , Células Th2/efectos de los fármacos
14.
Immunol Res ; 58(1): 106-17, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-24366664

RESUMEN

Administration of the influenza vaccination to patients with an egg allergy is major health concern. Contaminating egg antigens occasionally induce severe anaphylactic shock in these patients following administration of the vaccination; therefore, the development of a safer vaccination is needed. In the present study, we investigated whether a mixture of four newly and previously generated anti-ovalbumin (OVA) IgA monoclonal antibodies (mAbs) could inhibit both anaphylactic shock upon a subcutaneous OVA challenge and subsequent further sensitization against OVA in passively anti-OVA IgE-sensitized mice and actively sensitized mice with an injection of OVA. The prevention of anaphylaxis by anti-OVA IgA mAbs was suggested to be mediated through the inhibition of OVA binding to allergenic antibodies such as anti-OVA IgE on mast cells and deceleration of the rate of OVA penetration from the injected site into the systemic circulation. Anti-OVA IgA mAbs inhibited further sensitization against OVA in mice actively sensitized with OVA, but did not affect sensitization against the unrelated antigen, phosphorylcholine-keyhole limpet hemocyanin co-injected with OVA. Our findings indicate that adding the anti-egg antigen IgA to the influenza vaccine should reduce not only the risk of inducing anaphylactic shock, but also undesired further sensitization against egg antigens following the vaccination without affecting the intended beneficial effect of the vaccine, namely the upregulation of immune responses to influenza viruses.


Asunto(s)
Anafilaxia/inmunología , Anafilaxia/terapia , Anticuerpos Monoclonales/inmunología , Anticuerpos Monoclonales/farmacología , Vacunas contra la Influenza/farmacología , Ovalbúmina/inmunología , Anafilaxia/prevención & control , Animales , Anticuerpos Monoclonales/efectos adversos , Modelos Animales de Enfermedad , Humanos , Vacunas contra la Influenza/efectos adversos , Vacunas contra la Influenza/inmunología , Masculino , Ratones , Ratones Endogámicos DBA , Ovalbúmina/antagonistas & inhibidores , Estadísticas no Paramétricas
15.
Science ; 332(6032): 966-70, 2011 May 20.
Artículo en Inglés | MEDLINE | ID: mdl-21512002

RESUMEN

Protein synthesis and autophagic degradation are regulated in an opposite manner by mammalian target of rapamycin (mTOR), whereas under certain conditions it would be beneficial if they occurred in unison to handle rapid protein turnover. We observed a distinct cellular compartment at the trans side of the Golgi apparatus, the TOR-autophagy spatial coupling compartment (TASCC), where (auto)lysosomes and mTOR accumulated during Ras-induced senescence. mTOR recruitment to the TASCC was amino acid- and Rag guanosine triphosphatase-dependent, and disruption of mTOR localization to the TASCC suppressed interleukin-6/8 synthesis. TASCC formation was observed during macrophage differentiation and in glomerular podocytes; both displayed increased protein secretion. The spatial coupling of cells' catabolic and anabolic machinery could augment their respective functions and facilitate the mass synthesis of secretory proteins.


Asunto(s)
Autofagia , Senescencia Celular , Vesículas Citoplasmáticas/metabolismo , Proteínas/metabolismo , Serina-Treonina Quinasas TOR/metabolismo , Aminoácidos/metabolismo , Animales , Línea Celular , Citoplasma/metabolismo , Vesículas Citoplasmáticas/ultraestructura , Retículo Endoplásmico Rugoso/ultraestructura , Genes ras , Aparato de Golgi/ultraestructura , Células HL-60 , Humanos , Interleucina-6/metabolismo , Interleucina-8/metabolismo , Lisosomas/metabolismo , Lisosomas/ultraestructura , Ratones , Proteínas de Unión al GTP Monoméricas/genética , Proteínas de Unión al GTP Monoméricas/metabolismo , Nocodazol/farmacología , Fagosomas/metabolismo , Fagosomas/ultraestructura , Fenotipo , Podocitos/metabolismo , Podocitos/ultraestructura , Biosíntesis de Proteínas , Vacuolas/ultraestructura , Red trans-Golgi/metabolismo , Red trans-Golgi/ultraestructura
16.
Clin Cancer Res ; 16(10): 2792-802, 2010 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-20406843

RESUMEN

PURPOSE: The heat shock protein 90 (Hsp90) plays an important role in chaperoning oncogenic client proteins in multiple myeloma (MM) cells, and several Hsp90 inhibitors have shown antitumor activities both in vitro and in vivo. However the precise mechanism of action of Hsp90 inhibitor in MM has not been fully elucidated. EXPERIMENTAL DESIGN: We evaluated the antitumor activities of KW-2478, a nonansamycin Hsp90 inhibitor, in MM cells with various chromosomal translocations of immunoglobulin heavy chain (IgH) loci both in vitro and in vivo. RESULTS: Our studies revealed that exposure of KW-2478 to MM cells resulted in growth inhibition and apoptosis, which were associated with degradation of well-known client proteins as well as a decrease in IgH translocation products (FGFR3, c-Maf, and cyclin D1), and FGFR3 was shown to be a new client protein of Hsp90 chaperon complex. In addition, KW-2478 depleted the Hsp90 client Cdk9, a transcriptional kinase, and the phosphorylated 4E-BP1, a translational inhibitor. Both inhibitory effects of KW-2478 on such transcriptional and translational pathways were shown to reduce c-Maf and cyclin D1 expression. In NCI-H929 s.c. inoculated model, KW-2478 showed a significant suppression of tumor growth and induced the degradation of client proteins in tumors. Furthermore, in a novel orthotopic MM model of i.v. inoculated OPM-2/green fluorescent protein, KW-2478 showed a significant reduction of both serum M protein and MM tumor burden in the bone marrow. CONCLUSIONS: These results suggest that targeting such diverse pathways by KW-2478 could be a promising strategy for the treatment of MM with various cytogenetic abnormalities.


Asunto(s)
Antineoplásicos/farmacología , Proteínas HSP90 de Choque Térmico/antagonistas & inhibidores , Morfolinas/farmacología , Mieloma Múltiple/tratamiento farmacológico , Transducción de Señal/efectos de los fármacos , Animales , Western Blotting , Proliferación Celular/efectos de los fármacos , Humanos , Ratones , Ratones Desnudos , Mieloma Múltiple/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Ensayos Antitumor por Modelo de Xenoinjerto
17.
Biochem Biophys Res Commun ; 377(4): 1085-90, 2008 Dec 26.
Artículo en Inglés | MEDLINE | ID: mdl-18977202

RESUMEN

Bcl-2 is an intracellular membrane protein that prevents cells from undergoing apoptosis in response to various cell-death signals. It negatively regulates mitochondrial outer membrane permeabilization, which is responsible for the release of apoptogenic factors and the subsequent activation of caspases. A microbial metabolite, aranorosin, was identified as an inhibitor of the anti-apoptotic function of Bcl-2. Based on its structure, a more potent derivative, K050, was synthesized. Apoptosis could be induced in a cell line that overexpressed Bcl-2 when cells were treated with an anti-Fas antibody in addition to K050, at sub-micromolar concentrations. Furthermore, K050 inhibited anti-apoptotic functions regulated by Bcl-2, resulting in a Fas-triggered mitochondrial transmembrane potential loss, the activation of caspase-9, and a morphological change to apoptosis. Inhibition of cell-based function of Bcl-2 and its anti-apoptotic effects could serve as useful pharmacological effects. Thus, a novel aranorosin derivative, K050, could be a potent therapeutic agent against Bcl-2-overexpressing human malignancies.


Asunto(s)
Apoptosis/efectos de los fármacos , Proteínas Proto-Oncogénicas c-bcl-2/antagonistas & inhibidores , Núcleo Celular/efectos de los fármacos , Núcleo Celular/ultraestructura , Cicloheximida/farmacología , Furanos/química , Furanos/farmacología , Células HeLa , Humanos , Compuestos de Espiro/síntesis química , Compuestos de Espiro/química , Compuestos de Espiro/farmacología , Receptor fas/antagonistas & inhibidores
18.
Bioorg Med Chem Lett ; 18(5): 1588-91, 2008 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-18243703

RESUMEN

Hsp90 is an attractive chemotherapeutic target because it is essential to maturation of multiple oncogenes. We describe the conformational significance of EH21A1-A4, phenolic derivatives of geldanamycin isolated from Streptomyces sp. Their native free structures are similar to the active form of geldanamycin bound to Hsp90 protein. Their conformational character is a probable reason for their high-affinity binding. Lack of toxic benzoquinone in EH21A1-A4 also adds to their potential as lead compounds for anti-tumor drugs.


Asunto(s)
Proteínas HSP90 de Choque Térmico/antagonistas & inhibidores , Lactamas Macrocíclicas/química , Lactamas Macrocíclicas/farmacología , Antineoplásicos/química , Antineoplásicos/farmacología , Benzoquinonas , Línea Celular Tumoral , Humanos , Modelos Moleculares , Estructura Molecular , Streptomyces/química
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...