Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 33
Filtrar
Mais filtros

Base de dados
Tipo de documento
País de afiliação
Intervalo de ano de publicação
1.
Biol Pharm Bull ; 40(12): 2191-2198, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-29199242

RESUMO

Cortisol levels in bodily fluids represent a useful index for pituitary-adrenal function, and thus practical anti-cortisol antibodies are required. We have studied "antibody-breeding" approaches, which involve in vitro evolution of antibodies to improve their antigen-binding performances. Here, we produced an antibody fragment to measure serum cortisol levels with over 30-fold enhanced affinity after single mutagenesis and selection steps. A mouse anti-cortisol antibody, Ab-CS#3, with insufficient affinity for practical use, was chosen as the prototype antibody. A "wild-type" single-chain Fv fragment (wt-scFv; Ka, 3.4×108 M-1) was prepared by bacterial expression of a fusion gene combining the VH and VL genes for this antibody. Then, random point mutations were generated separately in VH or VL by error-prone PCR, and the resulting products were used to assemble scFv genes, which were displayed on filamentous phages. Repeated panning of the phage library identified a mutant scFv (scFv#m1-L10) with an over 30-fold enhanced affinity (Ka 1.2×1010 M-1). Three amino acid substitutions (Cys49Ser, Leu54Pro, and Ser63Gly) were observed in its VL sequence. In a competitive enzyme-linked immunosorbent assay (ELISA), the mutant scFv generated dose-response curves with measuring range ca. 0.03-0.6 ng/assay cortisol, midpoint of which (0.15 ng/assay) was 7.3-fold lower than that of wt-scFv. Although cortisone, 11-deoxycortisol, and prednisolone showed considerable cross-reactivity, the mutant scFv should enable sensitive routine cortisol assays, except for measurement after metyrapone or high-dose of prednisolone administrations. Actually, cortisol levels of control sera obtained with the scFv-based ELISA were in the reference range.


Assuntos
Afinidade de Anticorpos , Evolução Molecular Direcionada/métodos , Hidrocortisona/análise , Região Variável de Imunoglobulina/imunologia , Sequência de Aminoácidos/genética , Animais , Anticorpos Monoclonais Murinos/química , Anticorpos Monoclonais Murinos/genética , Anticorpos Monoclonais Murinos/imunologia , Reações Cruzadas , Ensaio de Imunoadsorção Enzimática , Hibridomas , Hidrocortisona/antagonistas & inibidores , Hidrocortisona/imunologia , Região Variável de Imunoglobulina/química , Região Variável de Imunoglobulina/genética , Camundongos , Mutagênese , Mutação Puntual
2.
Anal Chem ; 87(24): 12387-95, 2015 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-26625180

RESUMO

In this study, we show the utility of Gaussia luciferase (GLuc), which is much smaller than previously found luciferases, as the fusion partner with artificial antibody species for developing sensitive immunoassay systems. As an example, we constructed a bioluminescent enzyme-linked immunosorbent assay (BL-ELISA) system determining the major glucocorticoid cortisol. A monoclonal antibody was newly elicited against a cortisol-albumin conjugate, and the genes encoding its variable domains (VH and VL) were cloned and combined to encode a single-chain Fv fragment (scFv). scFv was then linked to the wild-type GLuc gene or that encoding GLuc mutants reported to show improved emission kinetics and expressed in the periplasmic space of several Escherichia coli strains. Notably, the wild-type GLuc fusion protein (scFv-wtGLuc) showed the most suitable luminescent properties for BL-ELISAs. In our system, scFv-wtGLuc was reacted competitively with the analyte and immobilized cortisol moieties, and the bound GLuc activity was monitored with coelenterazine as the substrate. Successful batch-type luminescence detection was achieved using a plate reader without built-in injectors. The midpoint and limit of detection in a typical dose-response curve were 4.1 and 0.26 pg/assay, respectively, thus exhibiting much more sensitivity than conventional cortisol immunoassays. Serum cortisol levels (as the sum with cortisone) for healthy subjects, determined without any pretreatment, were compatible with reported reference ranges. The scFv-wtGLuc probe was stable over a year under storage as periplasmic extracts at -30 °C or with repeated freeze-thawing. These results suggest that GLuc fusions with antibody fragments might serve as useful and highly sensitive immunoassay probes in various clinical settings.


Assuntos
Anticorpos Monoclonais/genética , Ensaio de Imunoadsorção Enzimática/métodos , Hidrocortisona/análise , Luciferases/genética , Proteínas Recombinantes de Fusão/genética , Animais , Anticorpos Monoclonais/imunologia , Biomarcadores/análise , Feminino , Região Variável de Imunoglobulina/genética , Região Variável de Imunoglobulina/imunologia , Luciferases/análise , Luciferases/metabolismo , Medições Luminescentes , Camundongos , Camundongos Endogâmicos BALB C , Estrutura Molecular , Proteínas Recombinantes de Fusão/imunologia , Proteínas Recombinantes de Fusão/metabolismo
4.
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
5.
Anal Chem ; 85(10): 4930-7, 2013 May 21.
Artigo em Inglês | MEDLINE | ID: mdl-23631394

RESUMO

Conventional immunization-based antibodies are not always applicable to monitor trace amounts of clinical biomarkers due to insufficient antigen-binding affinity. The development of in vitro affinity maturation for native antibodies has paved the way to solve this problem, but there has been minimal success in obtaining improved antibody mutants for small molecule biomarkers (haptens). We here describe a "molecular breeding" approach, in which a practical anti-hapten antibody mutant has successfully been generated from a prototype antibody with poor binding properties. Random point mutations were introduced by error-prone polymerase chain reaction in the V(H) and V(L) genes of a second-generation mutant of a single-chain Fv fragment (scFv) that binds estradiol-17ß (E2), whose parent mouse Fab fragment showed poor affinity for E2 (Ka = 5.2 × 10(7) M(-1)). The resulting scFv gene library was expressed on filamentous phage particles. Phage clones with strong E2 binding were isolated with dissociation-independent methods using newly developed reagents. The binding characteristics and clinical applicability of the soluble scFvs prepared from the selected clones were examined in a competitive enzyme-linked immunosorbent assay (ELISA). We finally obtained a third-generation scFv mutant, scFv#m3-a18 (11 amino acid substitutions), which showed ~250-fold greater Ka (1.3 × 10(10) M(-1)) than the parent Fab, and yielded sensitive ELISA dose-response curves for E2 (limit of detection < 0.5 pg/assay) with practical specificity. The average human serum E2 levels, determined after acetonitrile extraction, were compatible with reported reference ranges. The present results will prompt a new era for preparing diagnostic reagents.


Assuntos
Afinidade de Anticorpos , Evolução Molecular Direcionada , Ensaio de Imunoadsorção Enzimática/métodos , Mutação , Anticorpos de Cadeia Única/genética , Anticorpos de Cadeia Única/imunologia , Animais , Biomarcadores/sangue , Estradiol/sangue , Estradiol/imunologia , Humanos , Modelos Moleculares , Conformação Proteica , Anticorpos de Cadeia Única/química
6.
Anal Chem ; 85(23): 11553-9, 2013 Dec 03.
Artigo em Inglês | MEDLINE | ID: mdl-24256209

RESUMO

Most immunoassays use probes that convert concentrations of analytes into signal intensity. To prepare the probes, analytes are usually linked to a reporter protein (e.g., enzymes) with the aid of chemical reagents. However, these conventional methods yield a mixture of heterogeneous products and consequently reduce assay performance. "Clonable" homogeneous probes, i.e., recombinant molecules in which a target protein is genetically fused to a reporter with a defined coupling ratio, are now available for analyzing protein biomarker concentrations. Here, we have expanded this strategy to measure small biomarkers (haptens) by genetically fusing proteinaceous molecules that mimic target haptens with enzymes. 11-Deoxycortisol (11-DC) was chosen as the model hapten, and the ß-type anti-idiotype antibodies (ßId-Abs) that recognize the paratope of anti-hapten antibodies were used as the target hapten mimic. The V(H) and V(L) genes of a ßId-Ab, targeting a mouse anti-11-DC antibody (CET-M8), were assembled to encode a single-chain Fv fragment (ßId-scFv), which was then fused with a gene encoding a variant of alkaline phosphatase. The product, ßId-scFv-ALP' protein, had satisfactory enzyme activity and bound to CET-M8 in a competitive manner with 11-DC. A colorimetric enzyme-linked immunosorbent assay (ELISA) for 11-DC, based on the competitive reaction between the analyte and ßId-scFv-ALP' against immobilized CET-M8, was found to be sensitive (limit of detection = 22 pg/assay) and specific (cross-reactivity with cortisol, 0.24%) for clinical use and could be used to determine serum 11-DC levels after a simple solvent extraction. The anti-idiotype scFv-enzyme fusion proteins proposed here can be prepared reproducibly as homogeneous products with a 1:1 coupling ratio and would facilitate standardization of immunoassays for small biomarkers.


Assuntos
Anticorpos Anti-Idiotípicos/química , Clonagem Molecular/métodos , Anticorpos de Cadeia Única/química , Adolescente , Adulto , Animais , Anticorpos Anti-Idiotípicos/genética , Biomarcadores/química , Feminino , Humanos , Imunoensaio/métodos , Masculino , Camundongos , Pessoa de Meia-Idade , Estrutura Secundária de Proteína , Anticorpos de Cadeia Única/genética , Adulto Jovem
7.
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
8.
Anal Chim Acta ; 1161: 238180, 2021 May 29.
Artigo em Inglês | MEDLINE | ID: mdl-33896564

RESUMO

Enzyme-linked immunosorbent assays (ELISAs) are essential for monitoring various biomarkers. Competitive and noncompetitive (sandwich) assay formats are used to determine hapten and macromolecule levels, respectively. Both formats require more sensitive detection of reporter enzymes for greater assay sensitivities. We previously reported the utility of wild-type Gaussia luciferase (wtGLuc) as a fusion partner with antibody single-chain Fv fragments (scFvs) for developing sensitive luminescent ELISAs. Here, we evaluated utility of NanoLuc luciferase (NLuc), a recently developed luciferase, as fusion partner with scFvs from the view of comparison with wtGLuc and a mutant of alkaline phosphatase (ALP'). Thyroxine (T4) and T4-labeled albumin were chosen as model haptenic and macromolecular antigens, respectively. An in-house-prepared anti-T4 scFv was fused with NLuc, wtGLuc, or ALP'. The scFv-NLuc fusion protein showed 47-fold and 29-fold lower limit of detection [LOD; 59 zmol (per assay)] than the wtGLuc- and ALP'-fusions, respectively. In a competitive T4 ELISA, the NLuc-fusion showed 9.3- and 6.3-fold lower LOD, (0.67 pg) than the wtGLuc- and ALP'-fusions, respectively, with a higher specificity in clinical applications. A typical colorimetric ELISA using a peroxidase-labeled second antibody showed 70-fold higher LOD than NLuc-based ELISA. Another advantage of the NLuc-fusion was shown in the sandwich assays; the LOD of T4-labeled albumin (5.0 fmol) was >6-fold lower than that of the other luminescent ELISAs. In an additional sandwich assay developed to count bacteriophage particles, NLuc enabled more sensitive determination than wtGLuc, whereas ALP' showed nearly equivalent performance. Its slowest alteration rate for light intensity after starting the enzyme reaction should enable robust batch-by-batch assay operations. Thus, we concluded that scFv-NLuc fusions serve as suitable probes in various types of immunoassays and may facilitate higher sensitivities with practical specificities.


Assuntos
Haptenos , Fragmentos de Imunoglobulinas , Ensaio de Imunoadsorção Enzimática , Imunoensaio , Luciferases/genética , Proteínas Recombinantes de Fusão/genética
9.
J Steroid Biochem Mol Biol ; 201: 105671, 2020 07.
Artigo em Inglês | MEDLINE | ID: mdl-32289430

RESUMO

The estrogen receptor (ER) plays a role in the progression of hormone-dependent breast cancer and is a hormone therapy target. Estrogen acts as a transcription factor (genomic action) and also produces a quick non-genomic reaction through intracellular signaling pathways. The membrane associated ER (mER) may regulate both these signals and hormone therapy resistance. However, the details remain unclear because a reliable method to distinguish the signals induced by the estradiol (E2)-mER and E2-nuclear ER complex has not been established. In the present study, we prepared the novel ligand Qdot-6-E2, selective for mER, by coupling E2 with insoluble quantum dot nano-beads. We investigated the characteristics of mER signaling pathways and its contribution to hormone therapy resistance using different cell lines including estrogen depletion resistant (EDR) cells with different mechanisms. Qdot-6-E2 stimulated proliferation of nuclear ER-positive cells, but nuclear ER-negative cells showed no response. In addition, Qdot-6-E2 indirectly activated nuclear ER and increased mRNA expression of target genes. We confirmed that E2 was not dissociated from Qdot-6-E2 using a mammalian one-hybrid assay. We visually demonstrated that Qdot-6-E2 acts from the outside of cells. The gene expression profile induced by Qdot-6-E2-mER was different from that induced by E2-nuclear ER. The effect of anti-ER antibody, the GFP-ER fusion protein localization, and the effect of palmitoyl acyltransferase inhibitor also indicated the existence of mER. Regarding intracellular phosphorylation signaling pathways, the MAPK (Erk 1/2) and the PI3K/Akt pathways were both activated by Qdot-6-E2. In EDR cells, only nuclear ER-positive cells showed increased cell proliferation with increased localization of ERα to the membrane fraction. These findings suggested that Qdot-6-E2 reacts with ERα surrounding the cell membrane and that mER signals help the cells to survive under estrogen-depleted conditions by re-localizing the ER to use trace amounts of E2 more effectively. We expect that Qdot-6-E2 is a useful tool for studying the mER.


Assuntos
Neoplasias da Mama/metabolismo , Estradiol/metabolismo , Estrogênios/metabolismo , Pontos Quânticos/metabolismo , Receptores de Estrogênio/metabolismo , Antineoplásicos Hormonais/uso terapêutico , Neoplasias da Mama/tratamento farmacológico , Linhagem Celular Tumoral , Membrana Celular/metabolismo , Proliferação de Células , Resistencia a Medicamentos Antineoplásicos , Feminino , Regulação Neoplásica da Expressão Gênica , Humanos , Ligantes , Transcriptoma
10.
Breast Cancer ; 27(5): 963-972, 2020 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-32297248

RESUMO

BACKGROUND: Cyclin-dependent kinase (CDK) 4/6 inhibitors represent a significant advancement in the treatment of estrogen receptor (ER)-positive human epidermal growth factor receptor 2-negative advanced breast cancer. However, mechanisms of alterations after acquired resistance to CDK4/6 inhibitors and the optimal treatment options are still not established. METHODS: Abemaciclib-resistant cell lines were established from the models of estrogen deprivation-resistant cell lines which retained ER expression and activated ER function derived from MCF-7 breast cancer cell lines. Ribocilib-resistant cell lines were established in the same method as previously reported. RESULTS: Both abemaciclib- and ribociclib-resistant cell lines showed decreased ER expression. ER transcriptional activity was maintained in these cell lines; however, the sensitivity to 4-hydroxytamoxifen and fulvestrant was almost completely lost. These cell lines did not exhibit any ERα gene mutation. Abemaciclib-resistant cell lines demonstrated low sensitivity to other CDK4/6 inhibitors; sensitivities to PI3K inhibitor, mTOR inhibitor, and chemotherapeutic drugs were maintained. CONCLUSIONS: Dependence on ER signaling appears to decrease after the development of acquired resistance to CDK4/6 inhibitors. Further, CDK4/6 inhibitor-resistant cells acquired cross-resistance to other CDK4/6 inhibitors, PI3K/Akt/mTOR inhibitor therapy and chemotherapeutic drugs might serve as optimal treatment options for such breast cancers.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/farmacologia , Neoplasias da Mama/tratamento farmacológico , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Inibidores de Proteínas Quinases/farmacologia , Receptores de Estrogênio/metabolismo , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Neoplasias da Mama/patologia , Quinase 4 Dependente de Ciclina/antagonistas & inibidores , Quinase 6 Dependente de Ciclina/antagonistas & inibidores , Antagonistas de Estrogênios/farmacologia , Antagonistas de Estrogênios/uso terapêutico , Feminino , Humanos , Células MCF-7 , Fosfatidilinositol 3-Quinases/metabolismo , Inibidores de Fosfoinositídeo-3 Quinase , Inibidores de Proteínas Quinases/uso terapêutico , Proteínas Proto-Oncogênicas c-akt/antagonistas & inibidores , Receptores de Estrogênio/antagonistas & inibidores , Serina-Treonina Quinases TOR/antagonistas & inibidores
11.
Cancer Sci ; 100(10): 1773-8, 2009 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-19575750

RESUMO

Recent remarkable progress in hormonal therapy has provided great benefit to breast cancer patients, but it also evokes novel issues: how accurately can the efficacy of each hormonal therapy be predicted and how can hormonal therapy-resistant patients be treated? These clinically important issues must be closely related to the biological events in each cancer, such as the alteration of intracellular multiple estrogen signaling pathways and the estrogen-related cancer microenvironment, which has recently revealed by molecular biological studies on estrogen and its receptors. However, the estrogen signaling status in individual breast cancers has not been clarified yet. Here we present the context of these issues and introduce our study of new tools which enable the visualization of estrogen signals in individual cancers. The assessment of estrogen receptor (ER)-alpha activity in individual cancers or ER-activating ability of the cancer microenvironment in each breast cancer patient revealed several new findings and interesting observations. We hope that these approaches provide new clues about the estrogen-dependent mechanisms of breast cancer development, and will be useful to advance the diagnosis and treatment of breast cancer patients.


Assuntos
Neoplasias da Mama/metabolismo , Estrogênios/metabolismo , Biologia Molecular/métodos , Proteômica/métodos , Transdução de Sinais/fisiologia , Neoplasias da Mama/genética , Feminino , Humanos , Receptores de Estrogênio
12.
Breast Cancer ; 26(3): 272-281, 2019 May.
Artigo em Inglês | MEDLINE | ID: mdl-30328006

RESUMO

BACKGROUND: Hormone therapy targeting the estrogen receptor (ER) pathway is the most common treatment used for ER-positive breast cancer. However, some patients experience de novo or acquired resistance, which becomes a critical problem. Activation of the insulin-like growth factor (IGF) pathway allows breast cancer cells to proliferate and is associated with the ER pathway. Little is known about the role of the IGF pathway in hormone therapy and resistance; therefore, we investigated whether the inhibition of this pathway may represent a novel therapeutic target for overcoming hormone therapy resistance in ER-positive breast cancers. METHODS: Crosstalk between the ER and IGF pathways was analyzed in breast cancer cell lines by inhibiting or stimulating either one or both pathways. We studied the effect of insulin-like growth factor one receptor (IGF1R) inhibition in aromatase inhibitor-resistant breast cancer cell lines and fulvestrant-resistant cell lines which were uniquely established in our laboratory. RESULTS: Under normal conditions, IGF signaling is controlled by ER signaling to promote cell growth. Temporary disruption of the estrogen supply results in attenuated ER signaling, and IGF-1 dramatically increased relative growth compared with normal conditions. In addition, IGF1R inhibitor strongly suppressd cell growth in hormone-resistant breast cancer cells where ER remains than cells where ER decreased or was almost lost. CONCLUSIONS: Our study suggests that inhibition of the IGF pathway may be an effective strategy for ER-positive breast cancer therapy, even in hormone therapy-resistant cases.


Assuntos
Antineoplásicos Hormonais/farmacologia , Neoplasias da Mama/metabolismo , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Receptores de Estrogênio/metabolismo , Receptores de Somatomedina/metabolismo , Transdução de Sinais , Inibidores da Aromatase/farmacologia , Neoplasias da Mama/tratamento farmacológico , Neoplasias da Mama/patologia , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Antagonistas do Receptor de Estrogênio/farmacologia , Estrogênios/deficiência , Estrogênios/farmacologia , Feminino , Humanos , Fator de Crescimento Insulin-Like I/antagonistas & inibidores , Fator de Crescimento Insulin-Like I/metabolismo , Células MCF-7 , Receptor Cross-Talk/efeitos dos fármacos , Receptor IGF Tipo 1 , Receptores de Estrogênio/deficiência , Receptores de Estrogênio/genética , Receptores de Somatomedina/antagonistas & inibidores , Receptores de Somatomedina/genética
13.
Breast Cancer ; 26(6): 748-757, 2019 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-31119683

RESUMO

BACKGROUND: Hormonal therapy is an effective treatment for luminal-like breast cancer. Aromatase inhibitor (AI) is widely used for estrogen receptor-positive, postmenopausal breast cancers. However, resistance is occurred and becomes a serious clinical concern. In general, progression of cancer strongly depends on tumor microenvironment, which may, therefore, also contribute to the development of AI resistance. METHODS: We evaluated tumor microenvironment-derived factors with respect to AI resistance using typical estrogen receptor-positive breast cancer cell lines. We established tumor microenvironment-dependent AI-resistant models and elucidated the underlying mechanisms. RESULTS: T-47D cells had a higher dependence on microenvironment-derived factors, such as estrogen or growth factors, for survival than MCF-7 cells. We, therefore, evaluated tumor microenvironment growth factors with respect to AI resistance using T-47D cells. We established three resistant cell lines (V1, V2, and V3) that survived estrogen deprivation and growth factor-supplemented conditions. These cell lines were deficient in estrogen receptor α expression and estrogen-dependent growth. Among six representative growth factors, epidermal growth factor was the most influential. In these models, HER2 protein was overexpressed without gene amplification and intracellular phosphorylation pathways were activated compared to parental cell lines. Molecular targeting inhibitors revealed that V1 and V2 primarily rely on the PI3 K pathway for survival, whereas V3 relies on the MAPK pathway. CONCLUSIONS: This study demonstrates the importance of tumor microenvironment-derived factors for the development of AI resistance. These resistant models did not utilize the same resistance mechanism, suggesting that flexible strategies are essential in conquering resistance.


Assuntos
Neoplasias da Mama/tratamento farmacológico , Neoplasias da Mama/metabolismo , Resistencia a Medicamentos Antineoplásicos , Fator de Crescimento Epidérmico/metabolismo , Receptor alfa de Estrogênio/metabolismo , Microambiente Tumoral , Inibidores da Aromatase/efeitos adversos , Inibidores da Aromatase/uso terapêutico , Neoplasias da Mama/patologia , Sobrevivência Celular/efeitos dos fármacos , Receptor alfa de Estrogênio/genética , Feminino , Regulação Neoplásica da Expressão Gênica , Humanos , Células MCF-7 , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Inibidores de Fosfoinositídeo-3 Quinase/farmacologia , Receptor ErbB-2/metabolismo , Tiazóis/farmacologia , Transfecção
14.
Oncotarget ; 10(47): 4907-4918, 2019 Aug 06.
Artigo em Inglês | MEDLINE | ID: mdl-31448056

RESUMO

Although cyclin-dependent kinase (CDK) 4/6 inhibitors have exhibited remarkable results for patients with estrogen receptor (ER)-positive breast cancer in clinical trials, the mechanism of CDK4/6 inhibitor resistance remains unclear. Thus, this study aimed to investigate the mechanism of CDK4/6 inhibitor resistance using two CDK4/6 inhibitor resistant breast cancer cell lines. We established CDK6 overexpressed cell lines (MCF7-C6) from MCF-7 cells using the stably transfected CDK6 expression vector. Additionally, acquired ribociclib-resistant (RIBR) cell lines were created using ER-positive hormone-resistant cell lines by long-term exposure to ribociclib. CDK6 overexpression and the knockdown of CDK4 experiments highlight the significance of high levels of CDK4 and low levels of CDK6 in CDK4/6 inhibitor sensitivity. Moreover, RIBR cell lines did not exhibit incremental CDK6 compared with ER-positive hormone-resistant cell lines. In MCF7-C6 and RIBR cell lines, p21 levels decreased, and p21 levels were proportional to CDK4/6 inhibitor sensitivity. This study suggests that overexpression of CDK6 is one of the many possible mechanisms of resistance to CDK4/6 inhibitors. Furthermore, p21 levels have the potential to serve as a marker for CDK4/6 inhibitors independent of the resistance mechanism.

15.
Breast Cancer ; 26(4): 459-470, 2019 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-30610551

RESUMO

BACKGROUND: Presently, hormonal therapy targeting estrogen receptors is the most effective treatment available for luminal breast cancer. However, many patients relapse after the therapy. It has been suggested that cancer stem-like cells are involved with hormonal therapy resistance; in the present study, we evaluated this hypothesis. METHODS: In the present study, we used our previously established hormonal therapy-resistant cell lines, including aromatase inhibitor (AI)-resistant cells (Type 1 and Type 2) and fulvestrant-resistant cells (MFR). RESULTS: AI-resistant cell lines expressing ER (Type 1 V1 and V2) showed high cancer stemness in terms of their CD44/CD24 expression and side populations, which were stimulated by the addition of estrogen and inhibited by fulvestrant. However, ALDH activity was lower than in the ER-negative resistant cells, suggesting that the stemness of luminal cells is distinct from that of basal-like breast cancer cells. The migration and invasion activity of the ER-positive Type 1 V1 and V2 cells were higher than in the ER-negative cell lines, Type 2 and MFR. CONCLUSIONS: Fractionation of parental cells based on CD44/CD24 expression and colony formation assay indicated that CD44+/CD24+ cells might be the origin of hormonal therapy-resistant cells. This population reconstituted various other subpopulations under estrogen deprivation. These results indicate that hormonal therapy resistance is closely related to the cancer stem cell-like properties of luminal breast cancer.


Assuntos
Antineoplásicos Hormonais/farmacologia , Neoplasias da Mama/tratamento farmacológico , Neoplasias da Mama/patologia , Inibidores da Aromatase/farmacologia , Neoplasias da Mama/metabolismo , Antígeno CD24/metabolismo , Linhagem Celular Tumoral , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Resistencia a Medicamentos Antineoplásicos/genética , Estradiol/metabolismo , Antagonistas do Receptor de Estrogênio/farmacologia , Feminino , Fulvestranto , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Humanos , Receptores de Hialuronatos/metabolismo , Células MCF-7 , Células-Tronco Neoplásicas/patologia , Receptores de Estrogênio/metabolismo
16.
Steroids ; 73(14): 1485-99, 2008 Dec 22.
Artigo em Inglês | MEDLINE | ID: mdl-18824188

RESUMO

A single-chain Fv fragment (scFv) against estradiol-17beta (E(2)) was generated to begin the construction of a library of various mutated anti-steroid antibodies with an improved affinity and/or specificity. A hybridoma clone secreting a specific anti-E(2) antibody (Ab#E4-4) was established by the cell fusion using splenocytes from a mouse immunized with an immunogenic E(2)-carrier conjugate. DNA fragments encoding the variable heavy and light domains (V(H) and V(L)) of the Ab#E4-4 were cloned and combined to give the scFv gene fragment encoding the sequence 5'-V(H)-(GGGGS)(3)-V(L)-3'. Compared to the Ab#E4-4 Fab fragment, soluble scFv (scFv#E4-4) protein showed a similar affinity to E(2) (K(a)=8.6x10(7)M(-1)) and a similar cross-reaction profile. To further study the fundamentals for creating a comprehensive library of mutated scFvs, the scFvV(H) and V(L) genes were amplified using error-prone PCR conditions and the frequency and pattern of incorporated mutations were investigated. For this, regular Taq polymerase was used in the presence of unequal concentrations of dNTPs. At 1.0mM MnCl(2), the error frequency reached to 8.5% and 11% for the V(H) and V(L) respectively, although a significant transition/transversion bias was observed. ScFv#E4-4 and the mutated polyclonal scFvs were then displayed on filamentous phage under various packaging conditions. Cultivation of the transformed bacteria was more suitable at 25 degrees C than at higher temperatures for the packaging of scFv-bearing phagemid particles. Based on these experimental conditions, an scFv-displaying phage library, each scFv member in which has mutated complementarity-determining region (CDR) H2, H3, L1, and L3, was constructed. A soluble scFv clone (scFv#m1-e7) with a mutated amino acid (I-->V) in CDR L1, isolated from this library, showed threefold higher affinity (K(a)=2.6 x 10(8)M(-1)) than that of scFv#4-4.


Assuntos
Anticorpos Monoclonais/imunologia , Estradiol/genética , Estradiol/imunologia , Fragmentos de Imunoglobulinas/isolamento & purificação , Região Variável de Imunoglobulina/isolamento & purificação , Biblioteca de Peptídeos , Sequência de Aminoácidos , Animais , Sequência de Bases , Ensaio de Imunoadsorção Enzimática , Estrogênios/imunologia , Feminino , Hibridomas , Fragmentos de Imunoglobulinas/genética , Fragmentos de Imunoglobulinas/imunologia , Região Variável de Imunoglobulina/genética , Região Variável de Imunoglobulina/imunologia , Camundongos , Camundongos Endogâmicos BALB C , Dados de Sequência Molecular , Mutagênese , Reação em Cadeia da Polimerase , Homologia de Sequência de Aminoácidos , Homologia de Sequência do Ácido Nucleico
17.
Oncotarget ; 9(30): 21468-21477, 2018 Apr 20.
Artigo em Inglês | MEDLINE | ID: mdl-29765553

RESUMO

We previously reported the establishment of several types of long-term estrogen-depleted-resistant (EDR) cell lines from MCF-7 breast cancer cells. Type 1 EDR cells exhibited the best-studied mechanism of aromatase inhibitor (AI) resistance, in which estrogen receptor (ER) expression remained positive and PI3K signaling was upregulated. Type 2 EDR cells showed reduced ER activity and upregulated JNK-related signaling. The mTOR inhibitor everolimus reduced growth in cells similar to Type 1 EDR cells. The present study generated everolimus-resistant (EvR) cells from Types 1 and 2 EDR cells following long-term exposure to everolimus in vitro. These EvR cells modeled resistance to AI and everolimus combination therapies following first-line AI treatment failure. In Type 1 EvR cells, everolimus resistance was dependent on MAPK signaling; single agents were not effective, but hormonal therapy combined with a kinase inhibitor effectively reduced cell growth. In Type 2 EvR cells, ER expression remained negative and a JNK inhibitor was ineffective, but a Src inhibitor reduced cell growth. The mechanism of acquired everolimus resistance appears to vary depending on the mechanism of AI resistance. Strategies targeting resistant tumors should be tailored based on the resistance mechanisms, as these mechanisms impact therapeutic efficacy.

18.
J Steroid Biochem Mol Biol ; 165(Pt B): 448-457, 2017 01.
Artigo em Inglês | MEDLINE | ID: mdl-27632897

RESUMO

The relationship between tobacco smoke and breast cancer incidence has been studied for many years, but the effect of smoking on hormonal therapy has not been previously reported. We investigated the effect of smoking on hormonal therapy by performing in vitro experiments. We first prepared tobacco smoke condensate (TSC) and examined its effect on estrogen receptor (ER) activity. The ER activity was analyzed using MCF-7-E10 cells into which the estrogen-responsive element (ERE)-green fluorescent protein (GFP) reporter gene had been stably introduced (GFP assay) and performing an ERE-luciferase assay. TSC significantly activated ERs, and upregulated its endogenous target genes. This activation was inhibited by fulvestrant but more weakly by tamoxifen. These results suggest that the activation mechanism may be different from that for estrogen. Furthermore, using E10 estrogen depletion-resistant cells (EDR cells) established as a hormonal therapy-resistant model showing estrogen-independent ER activity, ER activation and induction of ER target genes were significantly higher following TSC treatment than by estradiol (E2). These responses were much higher than those of the parental E10 cells. In addition, the phosphorylation status of signaling factors (ERK1/2, Akt) and ER in the E10-EDR cells treated with TSC increased. The gene expression profile induced by estrogenic effects of TSC was characterized by microarray analysis. The findings suggested that TSC activates ER by both ligand-dependent and -independent mechanisms. Although TSC constituents will be metabolized in vivo, breast cancer tissues might be exposed for a long period along with hormonal therapy. Tobacco smoke may have a possibility to interfere with hormonal therapy for breast cancer, which may have important implications for the management of therapy.


Assuntos
Resistencia a Medicamentos Antineoplásicos , Receptor alfa de Estrogênio/metabolismo , Fumaça , Antineoplásicos Hormonais/farmacologia , Proliferação de Células , Estradiol/análogos & derivados , Estradiol/farmacologia , Estrogênios/farmacologia , Fulvestranto , Regulação Neoplásica da Expressão Gênica , Genes Reporter , Proteínas de Fluorescência Verde/metabolismo , Hormônios/farmacologia , Humanos , Ligantes , Células MCF-7 , Análise de Sequência com Séries de Oligonucleotídeos , Fosforilação , Transdução de Sinais/efeitos dos fármacos , Tamoxifeno/farmacologia , Nicotiana
19.
J Steroid Biochem Mol Biol ; 167: 115-125, 2017 03.
Artigo em Inglês | MEDLINE | ID: mdl-27888136

RESUMO

Approximately 70% of breast cancers express estrogen receptor α (ERα), which plays critical roles in breast cancer development. Fulvestrant has been effectively used to treat ERα-positive breast cancer, although resistance remains a critical problem. To elucidate the mechanism of resistance to fulvestrant, we established fulvestrant-resistant cell-lines named MFR (MCF-7 derived fulvestrant resistance) and TFR (T-47D derived fulvestrant resistance) from the ERα-positive luminal breast cancer cell lines MCF-7 and T-47D, respectively. Both fulvestrant-resistant cell lines lost sensitivity to estrogen and anti-estrogens. We observed diminished ERα expression at both the protein and mRNA levels. To address the mechanism of gene expression regulation, we examined epigenetic alteration, especially the DNA methylation level of ERα gene promoters. MFR cells displayed high methylation levels upstream of the ERα gene, whereas no change in DNA methylation was observed in TFR cells. Hence, we examined the gene expression plasticity of ERα, as there are differences in its reversibility following fulvestrant withdrawal. ERα gene expression was not restored in MFR cells, and alternative intracellular phosphorylation signals were activated. By contrast, TFR cells exhibited plasticity of ERα gene expression and ERα-dependent growth; moreover, these cells were resensitized to estrogen and anti-estrogens. The difference in epigenetic regulation among individual cells might explain the difference in the plasticity of ERα expression. We also identified an MFR cell-activating HER/Src-Akt/MAPK pathway; thus, the specific inhibitors effectively blocked MFR cell growth. This finding implies the presence of multiple fulvestrant resistance mechanisms and suggests that the optimal therapies differ among individual tumors as a result of differing epigenetic mechanisms regulating ERα gene expression.


Assuntos
Neoplasias da Mama/tratamento farmacológico , Neoplasias da Mama/metabolismo , Resistencia a Medicamentos Antineoplásicos , Epigênese Genética/efeitos dos fármacos , Estradiol/análogos & derivados , Receptor alfa de Estrogênio/metabolismo , Linhagem Celular Tumoral , Proliferação de Células , Metilação de DNA , Relação Dose-Resposta a Droga , Estradiol/farmacologia , Estrogênios/farmacologia , Feminino , Fulvestranto , Regulação Neoplásica da Expressão Gênica , Humanos , Sistema de Sinalização das MAP Quinases , Células MCF-7 , Regiões Promotoras Genéticas , Transdução de Sinais
20.
J Steroid Biochem Mol Biol ; 171: 209-217, 2017 07.
Artigo em Inglês | MEDLINE | ID: mdl-28412323

RESUMO

Hormone therapy is the most effective treatment for patients with estrogen receptor α-positive breast cancers. However, although resistance occurs during treatment in some cases and often reflects changed estrogen receptor α status, the relationship between changes in estrogen receptor α expression and resistance to therapy are poorly understood. In this study, we identified a mechanism for altered estrogen receptor α expression during disease progression and acquired hormone therapy resistance in aromatase inhibitor-resistant breast cancer cell lines. Subsequently, we investigated promoter switching and DNA methylation status of the estrogen receptor α promoter, and found marked changes of methylation at a single CpG site (CpG4) in resistant cells. In addition, luciferase reporter assays showed reduced transcriptional activity from this methylated CpG site. This CpG region was also completely conserved among species, suggesting that it acts as a methylation-sensitive Ets-2 transcription factor binding site, as confirmed using chromatin immunoprecipitation assays. In estrogen receptor α-positive tumors, CpG4 methylation levels were inversely correlated with estrogen receptor α expression status, suggesting that single CpG site plays an important role in the regulation of estrogen receptor α transcription.


Assuntos
Neoplasias da Mama/metabolismo , Metilação de DNA , Fosfatos de Dinucleosídeos/metabolismo , Receptor alfa de Estrogênio/metabolismo , Regiões Promotoras Genéticas , Proteína Proto-Oncogênica c-ets-2/metabolismo , Transcrição Gênica , Antineoplásicos Hormonais/farmacologia , Inibidores da Aromatase/farmacologia , Sequência de Bases , Neoplasias da Mama/tratamento farmacológico , Sequência Conservada , Metilação de DNA/efeitos dos fármacos , DNA Recombinante/metabolismo , Resistencia a Medicamentos Antineoplásicos , Receptor alfa de Estrogênio/química , Receptor alfa de Estrogênio/genética , Feminino , Genes Reporter/efeitos dos fármacos , Humanos , Células MCF-7 , Pessoa de Meia-Idade , Mutação , Proteínas de Neoplasias/química , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Regiões Promotoras Genéticas/efeitos dos fármacos , Proteína Proto-Oncogênica c-ets-2/genética , Proteínas Recombinantes/metabolismo , Elementos de Resposta/efeitos dos fármacos , Transcrição Gênica/efeitos dos fármacos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA