Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 118
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
Int Immunol ; 31(2): 69-79, 2019 02 15.
Artigo em Inglês | MEDLINE | ID: mdl-30299470

RESUMO

Kit/CD117 plays a crucial role in the cell-cell and cell-matrix adhesion of mammalian mast cells (MCs); however, it is unclear whether other adhesion molecule(s) perform important roles in the adhesion of MCs. In the present study, we show a novel Kit-independent adhesion mechanism of mouse cultured MCs mediated by Notch family members. On stromal cells transduced with each Notch ligand gene, Kit and its signaling become dispensable for the entire adhesion process of MCs from tethering to spreading. The Notch-mediated spreading of adherent MCs involves the activation of signaling via phosphatidylinositol 3-kinases and mitogen-activated protein kinases, similar to Kit-mediated spreading. Despite the activation of the same signaling pathways, while Kit supports the adhesion and survival of MCs, Notch only supports adhesion. Thus, Notch family members are specialized adhesion molecules for MCs that effectively replace the adhesion function of Kit in order to support the interaction of MCs with the surrounding cellular microenvironments.


Assuntos
Mastócitos/imunologia , Proteínas Proto-Oncogênicas c-kit/imunologia , Receptores Notch/imunologia , Animais , Adesão Celular/imunologia , Células Cultivadas , Mastócitos/citologia , Camundongos , Camundongos Endogâmicos C57BL
2.
Int J Clin Oncol ; 20(2): 262-7, 2015 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25652907

RESUMO

Whilst estrogen receptor (ER)-positive breast cancers are preferentially treated with hormone therapy, approximately one-third of them relapse. The mechanisms of refractoriness have been investigated by numerous studies but have not been fully clarified. Hormonal therapy resistance, particularly aromatase inhibitor (AI) resistance, may be related to the acquisition of alternative intracellular ER signaling. We have been investing the mechanisms using cancer specimens and cell lines by monitoring the transcription activity of ERs. AI refractory specimens showed diverse ER activity in the adenovirus estrogen receptor element-green fluorescent protein (ERE-GFP) assay and varied sensitivity to anti-estrogens, indicating the existence of multiple resistant mechanisms. We established six different types of cell lines mimicking AI resistance from ERE-GFP-introduced ER-positive cell lines. They revealed that multiple and alternative ER activating pathways were involved in the resistance, such as phosphorylation-dependent or androgen metabolite-dependent mechanisms. The response to fulvestrant and mammalian target of rapamycin inhibitor also varied among individual resistant cell lines. These results indicate that further subclassification of ER-positive breast cancer is extremely important to decide the therapeutic management of not only hormonal therapy but also new molecular target therapy.


Assuntos
Antineoplásicos Hormonais/uso terapêutico , Inibidores da Aromatase/uso terapêutico , Neoplasias da Mama/tratamento farmacológico , Resistencia a Medicamentos Antineoplásicos , Receptores de Estrogênio/metabolismo , Neoplasias da Mama/química , Linhagem Celular Tumoral , Feminino , Humanos , Ligantes , Fosforilação , Receptores de Estrogênio/análise , Receptores de Estrogênio/genética , Transdução de Sinais , Transcrição Gênica
3.
Breast Cancer Res Treat ; 143(1): 69-80, 2014 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-24292869

RESUMO

Aromatase inhibitors (AIs) exert antiproliferative effects by reducing local estrogen production from androgens in postmenopausal women with hormone-responsive breast cancer. Previous reports have shown that androgen metabolites generated by the aromatase-independent enzymes, 5α-androstane-3ß, 17ß-diol (3ß-diol), androst-5-ene-3ß, and 17ß-diol (A-diol), also activate estrogen receptor (ER) α. Estradiol (E2) can also reportedly be generated from estrone sulfate (E1S) pooled in the plasma. Estrogenic steroid-producing aromatase-independent pathways have thus been proposed as a mechanism of AI resistance. However, it is unclear whether these pathways are functional in clinical breast cancer. To investigate this issue, we assessed the transcriptional activities of ER in 45 ER-positive human breast cancers using the adenovirus estrogen-response element-green fluorescent protein assay and mRNA expression levels of the ER target gene, progesterone receptor, as indicators of ex vivo and in vivo ER activity, respectively. We also determined mRNA expression levels of 5α-reductase type 1 (SRD5A1) and 3ß-hydroxysteroid dehydrogenase type 1 (3ß-HSD type 1; HSD3B1), which produce 3ß-diol from androgens, and of steroid sulfatase (STS) and 17ß-hydroxysteroid dehydrogenase type 1 (17ß-HSD type 1; HSD17B1), which produce E2 or A-diol from E1S or dehydroepiandrosterone sulfate. SRD5A1 and HSD3B1 expression levels were positively correlated with ex vivo and in vivo ER activities. STS and HSD17B1 expression levels were positively correlated with in vivo ER activity alone. Elevated expression levels of these steroid-metabolizing enzymes in association with high in vivo ER activity were particularly notable in postmenopausal patients. Analysis of the expression levels of steroid-metabolizing enzymes revealed positive correlations between SRD5A1 and HSD3B1, and STS and HSD17B1. These findings suggest that the SRD5A1-HSD3B1 as well as the STS-HSD17B pathways, could contributes to ER activation, especially postmenopause. These pathways might function as an alternative estrogenic steroid-producing, aromatase-independent pathways.


Assuntos
Aromatase/metabolismo , Neoplasias da Mama/metabolismo , Redes e Vias Metabólicas , Esteroides/metabolismo , Adulto , Antineoplásicos Hormonais/farmacologia , Antineoplásicos Hormonais/uso terapêutico , Inibidores da Aromatase/farmacologia , Inibidores da Aromatase/uso terapêutico , Neoplasias da Mama/tratamento farmacológico , Neoplasias da Mama/patologia , Estradiol/sangue , Estradiol/metabolismo , Feminino , Humanos , Pessoa de Meia-Idade , Modelos Biológicos , Gradação de Tumores , Estadiamento de Neoplasias , Pós-Menopausa , Receptores de Estrogênio/metabolismo , Fatores de Risco , Transdução de Sinais
4.
Eur J Immunol ; 42(6): 1459-67, 2012 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-22622847

RESUMO

Under homeostatic conditions, skin DCs migrate to regional LNs transporting self-antigens (self-Ags). The transport of self-Ags is considered to be critical for maintaining peripheral tolerance. Although the chemokine receptor CCR7 potently induces the migration of skin DCs to regional LNs, Ccr7(-/-) (Ccr7-KO) mice do not show skin auto-immune diseases. To resolve this inconsistency, we examined Ccr7-KO epidermis- or dermis-hyperpigmented transgenic (Tg) mice, in which the transport of skin self-Ags is traceable by melanin granules (MGs). Under CCR7-deficient conditions, the transport of epidermal MGs to regional LNs was impaired at 7 weeks of age. However, epidermal MGs could be transported when they had accumulated in the dermis. Ccr7-KO-dermis-pigmented Tg mice confirmed the presence of CCR7-independent transport from the dermis. Compared with WT-dermis-pigmented Tg mice, the amount of transported melanin and number of MG-laden CD11c(+) cells were both approximately 40% of the WT levels, while the number of MG-laden CD205(+) or CD207(+) cells decreased to about 10% in skin regional LNs of Ccr7-KO-dermis-pigmented Tg mice. Cell sorting highlighted the involvement of CD11c(+) cells in the CCR7-independent transport. Here, we show that CCR7-independent transport of skin self-Ags occurs in the dermis. This system might contribute to the continuous transport of self-Ags, and maintain peripheral tolerance.


Assuntos
Autoantígenos/metabolismo , Derme/imunologia , Receptores CCR7/fisiologia , Animais , Transporte Biológico , Antígeno CD11c/análise , Antígeno CD11c/fisiologia , Tolerância Imunológica , Linfonodos/imunologia , Melaninas/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fator de Células-Tronco/fisiologia
5.
Breast Cancer Res Treat ; 139(3): 731-40, 2013 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-23780684

RESUMO

Aromatase inhibitors (AIs) have been reported to exert their antiproliferative effects in postmenopausal women with hormone receptor-positive breast cancer not only by reducing estrogen production but also by unmasking the inhibitory effects of androgens such as testosterone (TS) and dihydrotestosterone (DHT). However, the role of androgens in AI-resistance mechanisms is not sufficiently understood. 5α-Androstane-3ß,17ß-diol (3ß-diol) generated from DHT by 3ß-hydroxysteroid dehydrogenase type 1 (HSD3B1) shows androgenic and substantial estrogenic activities, representing a potential mechanism of AI resistance. Estrogen response element (ERE)-green fluorescent protein (GFP)-transfected MCF-7 breast cancer cells (E10 cells) were cultured for 3 months under steroid-depleted, TS-supplemented conditions. Among the surviving cells, two stable variants showing androgen metabolite-dependent ER activity were selected by monitoring GFP expression. We investigated the process of adaptation to androgen-abundant conditions and the role of androgens in AI-resistance mechanisms in these variant cell lines. The variant cell lines showed increased growth and induction of estrogen-responsive genes rather than androgen-responsive genes after stimulation with androgens or 3ß-diol. Further analysis suggested that increased expression of HSD3B1 and reduced expression of androgen receptor (AR) promoted adaptation to androgen-abundant conditions, as indicated by the increased conversion of DHT into 3ß-diol by HSD3B1 and AR signal reduction. Furthermore, in parental E10 cells, ectopic expression of HSD3B1 or inhibition of AR resulted in adaptation to androgen-abundant conditions. Coculture with stromal cells to mimic local estrogen production from androgens reduced cell sensitivity to AIs compared with parental E10 cells. These results suggest that increased expression of HSD3B1 and reduced expression of AR might reduce the sensitivity to AIs as demonstrated by enhanced androgen metabolite-induced ER activation and growth mechanisms. Androgen metabolite-dependent growth of breast cancer cells may therefore play a role in AI-resistance.


Assuntos
Androgênios/metabolismo , Inibidores da Aromatase/farmacologia , Neoplasias da Mama/tratamento farmacológico , Neoplasias da Mama/patologia , Resistencia a Medicamentos Antineoplásicos , Receptores de Estrogênio/metabolismo , Androstano-3,17-diol/metabolismo , Androstano-3,17-diol/farmacologia , Neoplasias da Mama/metabolismo , Di-Hidrotestosterona/metabolismo , Di-Hidrotestosterona/farmacologia , Feminino , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Humanos , Letrozol , Células MCF-7/efeitos dos fármacos , Complexos Multienzimáticos/genética , Complexos Multienzimáticos/metabolismo , Nitrilas/farmacologia , Progesterona Redutase/genética , Progesterona Redutase/metabolismo , Receptores Androgênicos/metabolismo , Elementos de Resposta/genética , Transdução de Sinais , Esteroide Isomerases/genética , Esteroide Isomerases/metabolismo , Testosterona/metabolismo , Testosterona/farmacologia , Triazóis/farmacologia
6.
J Inherit Metab Dis ; 36(3): 565-73, 2013 May.
Artigo em Inglês | MEDLINE | ID: mdl-23160874

RESUMO

BACKGROUND AND AIMS: In two Japanese infants with neonatal cholestasis, 3-oxo-Δ(4)-steroid 5ß-reductase deficiency was diagnosed based on mutations of the SRD5B1 gene. Unusual bile acids such as elevated 3-oxo-Δ(4) bile acids were detected in their serum and urine by gas chromatography-mass spectrometry. We studied effects of oral chenodeoxycholic acid treatment. PATIENTS AND METHODS: SRD5B1 gene analysis used peripheral lymphocyte genomic DNA. Diagnosis and treatment of these two patients were investigated retrospectively and prospectively investigated. RESULTS: With respect to SRD5B1, one patient was heterozygous (R266Q, a novel mutation) while the other was a compound heterozygote (G223E/R261C). Chenodeoxycholic acid treatment was effective in improving liver function and decreasing unusual bile acids such as 7α-hydroxy- and 7α,12α-dihydroxy-3-oxo-4-cholen-24-oic acids in serum and urine. CONCLUSION: Primary bile acid treatment using chenodeoxycholic acid was effective for these patients treated in early infancy before the late stage of chronic cholestatic liver dysfunction.


Assuntos
Ácidos e Sais Biliares/metabolismo , Ácido Quenodesoxicólico/uso terapêutico , Colestase/diagnóstico , Colestase/tratamento farmacológico , Colestase/genética , Oxirredutases/genética , Povo Asiático , Colestase/metabolismo , Feminino , Fármacos Gastrointestinais/uso terapêutico , Humanos , Recém-Nascido , Doenças do Recém-Nascido/diagnóstico , Doenças do Recém-Nascido/tratamento farmacológico , Doenças do Recém-Nascido/genética , Doenças do Recém-Nascido/metabolismo , Fígado/efeitos dos fármacos , Fígado/metabolismo , Fígado/patologia , Masculino , Mutação/fisiologia
7.
Breast Cancer ; 30(5): 727-738, 2023 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-37166625

RESUMO

BACKGROUND: Triple-negative breast cancer (TNBC) has an aggressive phenotype and poor outcome, however no specific targeted therapy has been established for TNBC lacking germline BRCA1/2 pathogenic variants. To develop a novel therapeutic strategy, we explored the potential of resveratrol (RSV) for TNBC treatment. METHODS: We investigated the effects of RSV on malignant phenotypes of TNBC cells as well as on apoptosis induced by ABT263, a specific inhibitor of BCL-2 and BCL-xL, using morphological observation, migration assay, ß-galactosidase staining, and Hoechst staining. To elucidate the underlying mechanisms of RSV-mediated effects, expression levels and histone acetylation levels of cadherin 1 (CDH1, E-cadherin) and cyclin dependent kinase inhibitor 1A (CDKN1A, p21) were determined by RT-qPCR, western blotting, and chromatin immunoprecipitation. Furthermore, knockdown analysis was conducted to evaluate the involvement of E-cadherin and/or p21 in RSV potentiation on cytotoxic activity of ABT263. RESULTS: RSV treatment induced epithelial-like cellular morphology and suppressed the migration capacity in MDA-MB-231 and BT-549-Luc TNBC cells. ß-galactosidase-positive cells were increased after RSV treatment, indicating the induction of cellular senescence, in MDA-MB-231 cells but not in BT-549-Luc cells. RSV increased the expression and histone acetylation of CDH1 and CDKN1A in both cells. Interestingly, pre-treatment with RSV enhanced the induction of apoptosis in the ABT263-treated MDA-MB-231 and BT-549-Luc cells, and knockdown of CDKN1A decreased ABT263-induced apoptosis in RSV-treated MDA-MB-231 cells. CONCLUSIONS: RSV represses the metastatic capacity and enhances the cytotoxic activity of ABT263 in TNBC cells. Our results suggested that RSV can potentially be used as a repressor of metastasis or a sensitizer to ABT263 for TNBC treatment via up-regulation of CDH1 and CDKN1A through epigenetic mechanisms.


Assuntos
Antineoplásicos , Neoplasias de Mama Triplo Negativas , Humanos , Resveratrol/farmacologia , Resveratrol/uso terapêutico , Neoplasias de Mama Triplo Negativas/tratamento farmacológico , Neoplasias de Mama Triplo Negativas/genética , Neoplasias de Mama Triplo Negativas/patologia , Proteína BRCA1/genética , Proteína BRCA1/metabolismo , Histonas/genética , Histonas/metabolismo , Histonas/farmacologia , Proliferação de Células , Epigênese Genética , Linhagem Celular Tumoral , Proteína BRCA2/genética , Antineoplásicos/uso terapêutico , Apoptose , Caderinas/genética , Caderinas/metabolismo
8.
Biochem Biophys Res Commun ; 417(1): 141-6, 2012 Jan 06.
Artigo em Inglês | MEDLINE | ID: mdl-22138652

RESUMO

While murine B- and T-lymphopoiesis require overlapping molecules, they occur in separate organs: the bone marrow (BM) and the thymus, respectively. The BM microenvironment is incapable of supporting T-lymphopoiesis because of insufficient interactions of Notch1 with delta-like ligand (Dll). Notch1/Dll interactions also play a role in the suppression of B-lymphopoiesis in the thymus. However, it is still unclear whether the Notch1/Dll interaction alone explains why the thymus does not support B-lymphopoiesis. In this study, we compared the precursor population colonizing the thymus with that in the BM by culturing them on stromal cells expressing abundant Dll1. We demonstrated that Flt3(+) Il7r(+) B220(+) Cd19(+) BM cells gave rise to B cells under this condition. We defined them as resistant to Dll1. In the thymus, Dll1-resistant cells were undetectable. This suggested that the absence of Dll1-resistant cells might explain the absence of B-lymphopoiesis in the thymus.


Assuntos
Linfócitos B/imunologia , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Linfopoese , Proteínas de Membrana/metabolismo , Receptor Notch1/metabolismo , Timo/imunologia , Animais , Antígenos CD19/metabolismo , Células Cultivadas , Antígenos Comuns de Leucócito/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Receptores de Interleucina-7/metabolismo , Tirosina Quinase 3 Semelhante a fms/metabolismo
9.
Biochem Biophys Res Commun ; 428(2): 303-8, 2012 Nov 16.
Artigo em Inglês | MEDLINE | ID: mdl-23085228

RESUMO

Differentiation into osteoclasts is induced by a macrophage colony-stimulating factor and receptor activator of nuclear-factor κB ligand. The macrophage-like cell line, C7 has the potential to differentiate into osteoclasts when it is cultured with both factors for 6 days. Although C7 is an established cell line, the frequency of differentiation into this lineage was less than 10%, and the ratio was maintained at a constant level, even after repeated cloning. In this study, to increase the differentiation of C7 cells to osteoclasts, C7 derivative treatments with several activators and/or inhibitors were performed for 3 days prior to setting osteoclast induction analysis; however, a reagent to significantly up-regulate the frequency of differentiation was not found. Only extended cultures for osteoclastogenesis exponentially increased the frequency of osteoclast precursors. It is likely that C7 cell differentiation into committed osteoclast precursors is on 'autopilot' rather than requiring specific signals to drive this process.


Assuntos
Diferenciação Celular , Macrófagos/citologia , Osteoclastos/citologia , Animais , Linhagem Celular , Linhagem da Célula , Humanos , Fator Estimulador de Colônias de Macrófagos/farmacologia , Macrófagos/efeitos dos fármacos , Camundongos , Processos Estocásticos
10.
Biochem Biophys Res Commun ; 428(2): 259-63, 2012 Nov 16.
Artigo em Inglês | MEDLINE | ID: mdl-23068099

RESUMO

Carcinoma-associated fibroblasts (CAFs) are associated with tumor progression and metastasis, and are able to activate estrogen receptor (ER) in breast cancer. We established a stable transformant of a human breast cancer cell line to detect CAF-specific ER-activating ability, and found that this CAF ability varied among tumors. Some studies have reported a high frequency of alterations among tumor suppressor genes in stromal cells, but do not generally agree as to the frequency. Moreover, the activation mechanism of CAF-induced estrogen signals, including the effects of these gene aberrations, is not fully understood. We investigated the relevance of tumor suppressor gene aberrations and ER-activating ability in CAFs derived from 20 breast cancer patients. Although CAF-specific ER-activating abilities varied among individual cases, all CAFs maintained wild-type alleles for TP53 and PTEN. Also, copy number aberrations in these genes were not observed in any CAFs. Our results suggest that the ER-activating ability of the CAFs is regulated independently of aberrations in these genes; and that other mechanisms of tumor-stromal interaction may affect activation of estrogen signals in breast cancer.


Assuntos
Neoplasias da Mama/genética , Carcinoma/genética , Fibroblastos/patologia , Regulação Neoplásica da Expressão Gênica , PTEN Fosfo-Hidrolase/genética , Receptores de Estrogênio/metabolismo , Proteína Supressora de Tumor p53/genética , Neoplasias da Mama/patologia , Carcinoma/patologia , Linhagem Celular Tumoral , Feminino , Fibroblastos/metabolismo , Dosagem de Genes , Humanos , Células Estromais/metabolismo , Células Estromais/patologia
11.
Breast Cancer Res Treat ; 133(2): 427-36, 2012 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-21909982

RESUMO

Neoadjuvant chemotherapy (NAC) has become the standard treatment for advanced breast cancer. Several prognostic markers, including estrogen receptor-α (ERα), are used to predict the response to NAC. However, the molecular significance of ERα expression in the efficacy of chemotherapy is not yet fully understood. To examine this issue, we first evaluated ERα transcriptional activity in breast cancer cells derived from pre-NAC specimens using estrogen response element-green fluorescent protein (ERE-GFP) as a reporter gene, and found that, in the cases for which ERα activities determined by GFP expression were not detected or low, pCR (pathological complete response) could be achieved even though ERα protein was expressed. Next, we examined the effects of alterations in ERα expression levels on sensitivity to paclitaxel, a key drug in NAC, by stable expression of ERα in ER-negative SKBR3 cells and by siRNA-mediated down-regulation of ERα in ER-positive MCF-7 cells, and showed that ERα expression and sensitivity to paclitaxel showed an inverse correlation. We also established paclitaxel-resistant MCF-7 cell clones and found that they have higher estrogen-induced ER activity than parent cells. Paclitaxel is a microtubule-stabilizing agent, while HDAC6 (histone deacetylase 6), which we previously identified as an estrogen-regulated gene, enhances cell motility by destabilizing microtubules via deacetylation of α-tubulin. Finally, we demonstrate herein that ERα knockdown in MCF-7 cells prevents deacetylation of α-tubulin, thereby increasing sensitivity to paclitaxel. Taken together, these results suggest that ERα expression directly regulates sensitivity to paclitaxel in NAC for breast cancer via the effect on microtubule stability.


Assuntos
Antineoplásicos Fitogênicos/uso terapêutico , Neoplasias da Mama/tratamento farmacológico , Neoplasias da Mama/genética , Receptor alfa de Estrogênio/genética , Paclitaxel/uso terapêutico , Acetilação , Neoplasias da Mama/metabolismo , Linhagem Celular Tumoral , Resistencia a Medicamentos Antineoplásicos/genética , Receptor alfa de Estrogênio/metabolismo , Feminino , Regulação Neoplásica da Expressão Gênica , Inativação Gênica , Desacetilase 6 de Histona , Histona Desacetilases/metabolismo , Humanos , Terapia Neoadjuvante , Transcrição Gênica , Resultado do Tratamento , Tubulina (Proteína)/metabolismo
12.
J Immunol ; 185(7): 3905-12, 2010 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-20810995

RESUMO

Mast cells (MCs) accumulate in chronic inflammatory sites; however, it is not clear which adhesion molecules are involved in this process. Recently, the expression of Notch ligands was reported to be upregulated in inflammatory sites. Although Notch receptors are known as signaling molecules that can activate integrins, their contributions to the adhesion of MCs have not been studied. In this study, we demonstrated that mouse MCs efficiently adhered to stromal cells forced to express a Notch ligand, Delta-like 1 (Dll1). Surprisingly, the adhesion was a consequence of direct cell-cell interaction between MCs and Dll1-expressing stromal cells rather than activation of downstream effectors of Notch receptor(s)-Dll1. The adhesion of MCs to Dll1-expressing stromal cells remained even when the cell metabolism was arrested. The recognition was blocked only by inhibition of Notch receptor(s)-Dll1 interaction by addition of soluble DLL1, or mAbs against Dll1 or Notch2. Taken together, these results indicate that Notch receptor(s) and Dll1 directly promote the adhesion of MCs to stromal cells by acting as adhesion molecules. This appreciation that Notch receptor-ligand interactions have an adhesion function will provide an important clue to molecular basis of accumulation of MCs to inflammatory sites.


Assuntos
Comunicação Celular/imunologia , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Mastócitos/metabolismo , Células Estromais/metabolismo , Animais , Proteínas de Ligação ao Cálcio , Moléculas de Adesão Celular/imunologia , Moléculas de Adesão Celular/metabolismo , Separação Celular , Citometria de Fluxo , Expressão Gênica , Peptídeos e Proteínas de Sinalização Intercelular/imunologia , Mastócitos/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Receptores Notch/imunologia , Receptores Notch/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Células Estromais/imunologia
13.
Surg Endosc ; 26(2): 523-7, 2012 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-21938576

RESUMO

BACKGROUND: Even for pediatric patients, the use of laparoscopic appendectomy has been widely accepted, and three trocars usually are necessary to perform a laparoscopic appendectomy. However, single-port appendectomy for children represents an attractive alternative. To reduce the number of incisions and trocars, the authors have adopted a transumbilical laparoscopically assisted single-port appendectomy (TULAA) approach. This study aimed to evaluate the results of their single-channel, single-port appendectomy. METHODS: A retrospective study of TULAA was performed during 12 years with 500 children ages 2-16 years (median, 10.2 years). The TULAA approach is a single-channel surgery using a 12 mm conventional single-port. The vertical incision through the umbilicus is used for laparoscopic access. Two laparoscopic instruments, a 5 mm telescope and a 5 mm grasper, are inserted simultaneously into the single-channel. The grasper holds the base of the appendix, and the appendix is exteriorized through the umbilical incision. Thereafter, a conventional appendectomy is performed extracorporeally. RESULTS: The TULAA procedure was successful for 416 patients (83.2%). These successful TULAA procedures required a mean surgery time of 44.5 min. The pathologic diagnosis of the appendix was acute for 59 patients, phlegmonous for 203 patients, gangrenous for 152 patients, and not detected for two patients. Complications occurred for 47 of these patients (11.3%). Most of the complications were associated with severe intraabdominal inflammation. Two patients needed reoperation under general anesthesia. Conversion to multitrocar surgery or open appendectomy was performed for 84 of the patients (16.8%). CONCLUSIONS: The TULAA procedure is a preferable operation for acute appendicitis in children because it is simple and provides good cosmetic results.


Assuntos
Apendicectomia/métodos , Apendicite/cirurgia , Laparoscopia/métodos , Doença Aguda , Adolescente , Criança , Pré-Escolar , Feminino , Humanos , Complicações Intraoperatórias/etiologia , Tempo de Internação , Masculino , Complicações Pós-Operatórias/etiologia , Estudos Retrospectivos , Resultado do Tratamento , Umbigo
14.
Breast Cancer ; 29(1): 77-91, 2022 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-34346034

RESUMO

BACKGROUND: There are various treatments for estrogen-positive breast cancer, mainly hormone therapy and molecular-targeted drugs. Acquiring resistance to these drugs is a major clinical problem. Additionally, little is known about the effect of drug resistance on the DNA repair mechanism. Poly ADP ribose polymerase (PARP) inhibitors currently used for treating HER2-negative metastatic breast cancer with BRCA mutations have been shown to be effective in BRCA-deficient cells with impaired homologous recombination repair. Here, we investigated the effect of drug resistance acquisition on the DNA repair mechanism and the effect of PARP inhibitors on ER (estrogen receptor) -positive breast cancer. METHODS: We investigated changes in the expression of DNA repair mechanism-related factors and repair ability of double-strand breaks (DSB) in various drug-resistant cell lines established in our laboratory. Additionally, PARP inhibitor susceptibility was investigated using olaparib. RESULTS: DSB repairs in MCF-7 and hormone therapy-resistant model cells were normal, and these cells demonstrated low sensitivity to olaparib. The resistant cell lines against CDK4/6 inhibitors, fulvestrant and mTOR/PI3K inhibitors showed decreased DSB repair ability and high olaparib sensitivity. They showed low sensitivity to CDK4/6 inhibitors, a close link between acquiring resistance to CDK4/6 inhibitors and hypersensitivity to olaparib. CONCLUSIONS: Our study suggests some cases of acquiring drug resistance impairs DSB repair ability and sensitizes ER-positive breast cancer to PARP inhibitors.


Assuntos
Neoplasias da Mama/tratamento farmacológico , Resistencia a Medicamentos Antineoplásicos , Terapia de Alvo Molecular , Inibidores de Poli(ADP-Ribose) Polimerases/farmacologia , Linhagem Celular Tumoral , Quinase 4 Dependente de Ciclina/antagonistas & inibidores , Quinase 6 Dependente de Ciclina/antagonistas & inibidores , Quebras de DNA de Cadeia Dupla , Reparo do DNA , Feminino , Humanos , Ftalazinas/farmacologia , Piperazinas/farmacologia , Receptores de Estrogênio
15.
Anticancer Res ; 42(2): 759-766, 2022 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-35093874

RESUMO

BACKGROUND/AIM: Carboxyl terminus of Hsc70-interacting protein (CHIP) is a ubiquitin ligase that induces ubiquitination and degradation of its target proteins including oncoproteins. We reported that its down-regulation is associated with tumor progression and metastasis of breast cancer. However, the mechanism through which CHIP gene affects cancer cells is unclear. MATERIALS AND METHODS: We extracted RNA from 45 primary breast cancer samples and compared CHIP mRNA expression profiles, promoter DNA methylation status, and clinicopathological information. RESULTS: CHIP mRNA expression was significantly correlated with the tumor progression status. In several samples, a pinpoint CpG methylation in the CHIP gene promoter region was significantly correlated with CHIP mRNA expression. When this specific CpG was methylated in estrogen receptor (ER)-positive cases, a significant difference in 5-year recurrence was not found compared with ER-negative cases. CONCLUSION: CpG methylation contributes to the long-term prognosis of ER-positive breast cancer.


Assuntos
Neoplasias da Mama/genética , Ubiquitina-Proteína Ligases/genética , Neoplasias da Mama/metabolismo , Neoplasias da Mama/patologia , Ilhas de CpG , Metilação de DNA , Progressão da Doença , Feminino , Regulação Neoplásica da Expressão Gênica , Humanos , Prognóstico , Regiões Promotoras Genéticas , RNA Mensageiro/genética , Receptores de Estrogênio/metabolismo , Recidiva , Células Tumorais Cultivadas
16.
Cancer Sci ; 102(10): 1848-54, 2011 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-21707867

RESUMO

Estrogens play an important role in the pathobiology of breast cancer. In postmenopausal women, peripheral synthesis of estrogens from adrenal/ovarian androgens, dehydroepiandrosterone (DHEA) or androstenedione (Adione), by estrogen-metabolizing enzymes is important. Besides estrone (E1) and estradiol (E2), androgen metabolites, such as androstene-3ß, 17ß-diol (Aenediol) or 5α-androstane-3ß, 17ß-diol (Aanediol), are known to have estrogenic functions, although they have been studied much less in breast cancer. To precisely elucidate steroid metabolism in breast cancer patients and to identify the pathobiological role of estrogenic androgen metabolites, concentrations of DHEA, Adione, Aenediol, Aanediol, E1, and E2 in pairs of serum and tumor tissue from patients with primary breast cancer were measured by liquid chromatography-tandem mass spectrometry. Cell proliferation assays using Aenediol were performed for four breast cancer cell lines. Serous E2 concentration was extremely low in postmenopausal women; however, a marked increase in tumor tissue was observed in hormone receptor-positive cases. E1 concentration, in contrast, was sustained at a higher level, even in postmenopausal serum, and did not increase in tumor tissue irrespective of the hormone receptor status. Dehydroepiandrosterone was most abundant in all samples, and exhibited a similar pattern as Adione and Aenediol. 5α-Androstane-3ß, 17ß-diol was undetectable in most samples. Androstene-3ß, 17ß-diol proliferated estrogen receptor-apositive breast cancer cells in the absence of E2. The intratumoral increase of E2, but not E1, in hormone receptor-positive postmenopausal breast cancer tissue, as well as the proliferative role of Aenediol, was elucidated.


Assuntos
Androstenodiol/metabolismo , Neoplasias da Mama/metabolismo , Hormônios Esteroides Gonadais/sangue , Adulto , Androstenodiona/biossíntese , Androstenodiona/sangue , Neoplasias da Mama/sangue , Neoplasias da Mama/patologia , Linhagem Celular Tumoral , Proliferação de Células , Desidroepiandrosterona/biossíntese , Desidroepiandrosterona/sangue , Estradiol/biossíntese , Estradiol/sangue , Receptor alfa de Estrogênio/metabolismo , Estrona/biossíntese , Estrona/sangue , Feminino , Humanos , Pessoa de Meia-Idade , Pós-Menopausa , Pré-Menopausa
17.
Cancer Sci ; 102(5): 918-25, 2011 May.
Artigo em Inglês | MEDLINE | ID: mdl-21281401

RESUMO

Glypican-3 (GPC3) is an onco-fetal antigen that is overexpressed in human hepatocellular carcinoma (HCC), and is only expressed in the placenta and embryonic liver among normal tissues. Previously, we identified an HLA-A2-restricted GPC3(144-152) (FVGEFFTDV) peptide that can induce GPC3-reactive CTLs without inducing autoimmunity in HLA-A2 transgenic mice. In this study, we carried out a phase I clinical trial of HLA-A2-restricted GPC3(144-152) peptide vaccine in 14 patients with advanced HCC. Immunological responses were analyzed by ex vivo γ-interferon enzyme-linked immunospot assay. The frequency of GPC3(144-152) peptide-specific CTLs after vaccination (mean, 96; range, 5-441) was significantly larger than that before vaccination (mean, 6.5; range, 0-43) (P < 0.01). An increase in the GPC3(144-152) peptide-specific CTL frequency was observed in 12 (86%) of 14 patients after vaccination. Additionally, there was a significant correlation between the maximum value of GPC3(144-152) peptide-specific CTLs after vaccination and the dose of the peptide injected (P = 0.0166, r = 0.665). Moreover, we established several GPC3(144-152) peptide-specific CTL clones from PBMCs of patients vaccinated with GPC3(144-152) peptide by single cell sorting using Dextramer and CD107a antibody. These CTL clones had high avidity (the recognition efficiency showing 50% cytotoxicity was 10(-10) or 10(-11) M) and could recognize HCC cell lines expressing GPC3 in an HLA-class I-restricted manner. These results suggest that GPC3(144-152) peptide vaccine can induce high avidity CTLs capable of killing HCC cells expressing GPC3. This trial was registered with University Hospital Medical Information Network number 000001395.


Assuntos
Vacinas Anticâncer/imunologia , Carcinoma Hepatocelular/imunologia , Glipicanas/imunologia , Antígeno HLA-A2/imunologia , Neoplasias Hepáticas/imunologia , Linfócitos T Citotóxicos/imunologia , Idoso , Afinidade de Anticorpos , Vacinas Anticâncer/uso terapêutico , Carcinoma Hepatocelular/patologia , Carcinoma Hepatocelular/terapia , Separação Celular , Células Clonais , Citotoxicidade Imunológica/imunologia , Feminino , Citometria de Fluxo , Humanos , Neoplasias Hepáticas/patologia , Neoplasias Hepáticas/terapia , Masculino , Pessoa de Meia-Idade , Estadiamento de Neoplasias , Vacinas de Subunidades Antigênicas/imunologia , Vacinas de Subunidades Antigênicas/uso terapêutico
18.
Breast Cancer Res Treat ; 122(2): 395-407, 2010 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-19847644

RESUMO

Multiple coactivator and corepressor complexes play an important role in endocrine processes and breast cancer; in particular, estrogen and estrogen receptor-alpha (ERalpha) promote the proliferation of breast cancer cells. Menin is a tumor suppressor encoded by Men1 that is mutated in the human-inherited tumor syndrome multiple endocrine neoplasia type 1 (MEN1); it also serves as a critical link in the recruitment of nuclear receptor-mediated transcription. Here, we show that menin expressed in breast cancer cell line MCF-7 is colocalized with ERalpha and functions as a direct coactivator of ER-mediated transcription in breast cancer cells. In MCF-7 cells, coexpression of menin and estrogen-response element-luciferase induced the activity of the latter in a hormone-dependent manner. Cells knocked down for ERalpha exhibited impaired ERE-luciferase activity induced by menin. Mammalian two-hybrid assay and GST pull-down assays indicated that menin could interact with the AF-2 domain of ERalpha. These results indicate that menin is a direct activator of ERalpha function. Tamoxifen inhibited the binding of menin to AF-2 in mammalian two-hybrid assay, but in menin-overexpressing clones, tamoxifen suppressed ERE-luciferase activity only to the levels of nontreated wild-type MCF-7. In a clinical study with 65 ER-positive breast cancer samples-all of which had been treated with tamoxifen for 2-5 years as adjuvant therapies--menin-positive tumors had a worse outcome than menin-negative ones. These indicated that menin can function as a transcriptional regulator of ERalpha and is a possible predictive factor for tamoxifen resistance.


Assuntos
Neoplasias da Mama/metabolismo , Resistencia a Medicamentos Antineoplásicos , Estradiol/metabolismo , Antagonistas de Estrogênios/uso terapêutico , Receptor alfa de Estrogênio/metabolismo , Proteínas Proto-Oncogênicas/metabolismo , Tamoxifeno/uso terapêutico , Animais , Sítios de Ligação , Neoplasias da Mama/tratamento farmacológico , Neoplasias da Mama/genética , Neoplasias da Mama/mortalidade , Neoplasias da Mama/patologia , Células COS , Linhagem Celular Tumoral , Quimioterapia Adjuvante , Chlorocebus aethiops , Intervalo Livre de Doença , Receptor alfa de Estrogênio/antagonistas & inibidores , Receptor alfa de Estrogênio/genética , Feminino , Regulação Neoplásica da Expressão Gênica , Humanos , Estimativa de Kaplan-Meier , Pessoa de Meia-Idade , Estadiamento de Neoplasias , Proteínas Nucleares/metabolismo , Prognóstico , Regiões Promotoras Genéticas , Mapeamento de Interação de Proteínas , Proteínas Proto-Oncogênicas/genética , Interferência de RNA , Proteínas Recombinantes de Fusão/metabolismo , Fatores de Tempo , Ativação Transcricional , Transfecção , Técnicas do Sistema de Duplo-Híbrido
19.
Front Immunol ; 11: 775, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32508808

RESUMO

In allergic contact dermatitis (ACD) and contact hypersensitivity (CHS), the healed skin shows greater swelling than the naïve skin in the same individual upon re-exposure to the same hapten. This "local skin memory" (LSM) in healed skin was maintained for a prolonged period of time and mediated by skin CD8+-resident memory T (TRM) cells in C57BL/6 mice. However, the number of CD4+ T cells is elevated in ACD-healed human skin, and the contribution of CD4+ TRM cells to the formation of LSM currently remains unclear. We herein demonstrated that immediately after CHS subsided, the healed skin in BALB/c mice showed an accumulation of hapten-specific CD4+ and CD8+ TRM cells, with a predominance of CD4+ TRM cells. The presence of CD4+ or CD8+ TRM cells in the healed skin was sufficient for the induction of a flare-up reaction upon a re-challenge. The CD4+ and CD8+ TRM cells both produced interferon-γ and tumor necrosis factor early after the re-challenge. Moreover, while CD8+ TRM cells gradually decreased over time and were eventually lost from the healed skin at 40-51 weeks after the resolution of CHS, the CD4+ TRM cell numbers remained elevated during this period. The present results indicate that the long-term maintenance of LSM is mediated by CD4+ TRM cells, and thus CD4+ TRM cells are an important target for the treatment of recurrent human ACD.


Assuntos
Linfócitos T CD4-Positivos/imunologia , Dermatite de Contato/imunologia , Memória Imunológica/imunologia , Pele/imunologia , Transferência Adotiva , Animais , Linfócitos T CD8-Positivos/imunologia , Dermatite Alérgica de Contato/imunologia , Haptenos/imunologia , Haptenos/farmacologia , Interferon gama/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Camundongos SCID , Camundongos Transgênicos , Fator de Necrose Tumoral alfa/metabolismo
20.
J Steroid Biochem Mol Biol ; 201: 105698, 2020 07.
Artigo em Inglês | MEDLINE | ID: mdl-32404282

RESUMO

Estrogen receptor (ER)α and the human epidermal growth factor receptor (HER) family are inversely expressed in ERα-positive cancer in association with resistance to hormonal therapy, but the mechanism underlying their relationship remains unknown. We analyzed the effect of HER family ligands on the expression of ER and the HER family in ERα-positive MCF-7 and T47D breast cancer cell lines in 3D spheroid culture. Here, we demonstrated for the first time that heregulin-1ß (HRG), a HER3 and HER4 ligand, most effectively regulated ER/HER family expression by decreasing ERα mRNA expression and increasing HER family mRNA expression. HRG treatment attenuated fulvestrant-mediated growth inhibition, and promoted the migration of MCF-7 cells. Moreover, HRG increased the CD44+/CD24- cell fraction and side population cells, both of which are recognized as prospective breast cancer stem cell markers. HRG activated both phosphatidylinositol 3-kinase/AKT/mammalian target of rapamycin (PI3K/AKT/mTOR) and mitogen-activated protein kinase (MAPK) pathways. Inhibitors of these pathways reduced the growth of MCF-7 cells, but the addition of HRG has different effects on these pathways. HRG blocked the inhibitory effect of mTOR inhibitors, such as rapamycin and everolimus, on cell growth but not that of a PI3K inhibitor. Furthermore, HRG slightly decreased the inhibitory effect of an AKT inhibitor on cell growth. In contrast, HRG enhanced the MEK inhibitor-induced inhibition of cell growth. These findings suggest that HRG-stimulated signaling pathways allow ERα-positive breast cancer cells to escape from growth inhibition caused by everolimus, via MAPK signaling and/or other signaling pathways. Everolimus improves progression-free survival in combination with exemestane as second-line therapy for metastatic hormone receptor-positive breast cancer. Our study suggests that HRG is a novel target for ERα-positive breast cancer therapy.


Assuntos
Neoplasias da Mama/genética , Receptor alfa de Estrogênio/genética , Neuregulina-1 , Receptor ErbB-2/genética , Antineoplásicos Hormonais/farmacologia , Neoplasias da Mama/metabolismo , Neoplasias da Mama/patologia , Linhagem Celular Tumoral , Movimento Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Antagonistas do Receptor de Estrogênio/farmacologia , Receptor alfa de Estrogênio/metabolismo , Everolimo/farmacologia , Feminino , Fulvestranto/farmacologia , Humanos , Terapia de Alvo Molecular , Células-Tronco Neoplásicas/efeitos dos fármacos , Sirolimo/farmacologia , Serina-Treonina Quinases TOR/antagonistas & inibidores , Resultado do Tratamento
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA