Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 55
Filtrar
1.
Neurogastroenterol Motil ; 30(10): e13378, 2018 10.
Artigo em Inglês | MEDLINE | ID: mdl-29797382

RESUMO

BACKGROUND: Serum levels of pro-inflammatory cytokines tend to be increased in irritable bowel syndrome (IBS) patients, or subgroups thereof. Still, the link between cytokine levels and IBS symptoms is unclear. We aim to determine systemic cytokine levels in IBS patients and healthy subjects (HS), confirm the presence of a subset of patients with an increased immune activity and to establish if cytokines are linked to IBS symptoms and pathophysiological factors. METHODS: Serum levels of interleukin (IL)-1ß, IL-6, IL-8, tumor necrosis factor (TNF), and IL-10 were measured. All subjects reported IBS symptoms using validated questionnaires and underwent colonic sensorimotor testing. Multivariate supervised orthogonal partial least squares-discriminant analysis (OPLS-DA) and unsupervised principal component analysis (PCA) and hierarchical cluster analysis (HCA) were implemented. KEY RESULTS: Irritable bowel syndrome patients (n = 246) had higher serum levels of IL-1ß, IL-6, IL-8, TNF, and IL-10 compared to HS (n = 21); however, serum cytokine profiles could not discriminate patients from HS. Moreover, cytokine levels were not correlated with symptoms among patients. Supervised OPLS-DA identified 104 patients (40% of patients) and unsupervised HCA analysis identified 49 patients (20%) with an increased immune activity indicated by elevated levels of serum cytokines compared to HS and the other patients. However, irrespective of how patients with increased immune activity were identified they were symptomatically similar to patients with no indication of increased immune activity. CONCLUSIONS & INFERENCES: Serum cytokines are elevated in IBS patients compared to HS. Immune activation characterizes a subset of patients, but modest associations between cytokine profile and symptoms suggest immune activity does not directly influence symptoms in IBS.


Assuntos
Citocinas/sangue , Síndrome do Intestino Irritável/sangue , Síndrome do Intestino Irritável/imunologia , Adulto , Feminino , Humanos , Masculino , Pessoa de Meia-Idade
2.
Br J Cancer ; 95(2): 172-80, 2006 Jul 17.
Artigo em Inglês | MEDLINE | ID: mdl-16819546

RESUMO

Resistance to antiepidermal growth factor (EGFR) strategies is an emerging clinical problem. Using human colorectal cancer (CRC) cells, we evaluated the involvement of the insulin receptor isoform-A (InsR-A) in de novo resistance to gefitinib, an EGFR tyrosine kinase inhibitor. Challenging the EGFR positive LoVo cells with gefitinib (1 microM) resulted in a small ( approximately 18%) inhibition of cell growth and although a modest reduction in phospho (p)EGFR Tyr845 was seen, pEGFR at residues -Tyr1068 and -Tyr1173 were unchanged. LoVo cells produced unprocessed pro-IGF-1R protein, substantial levels of IGF-II mRNA and mature InsR protein, consisting mainly of the InsR-A isoform. Insulin and IGF-II promoted cell growth and pEGFR Tyr845, Tyr1068 and Tyr1173 activity and conversely, the insulin-like growth factor-1 receptor (IGF-1R)/InsR inhibitor ABDP (1 muM) inhibited growth and reduced pEGFR activity at all three tyrosine residues. pInsR and pAkt levels were increased after gefitinib treatment. Blocking of pInsR with ABDP enabled gefitinib to markedly reduce pEGFR Tyr845, Tyr1068 and Tyr1173. Short-term gefitinib/ABDP dual treatment was more effective than either agent alone and chronic exposure to this combination resulted in total cell loss after 9 weeks, preventing acquisition of resistance to ABDP. LoVo cells with acquired resistance to ABDP were acutely sensitive to gefitinib. We concluded that InsR-A reduces sensitivity to gefitinib in LoVo CRC cells, thus its co-targeting alongside EGFR can improve the anti-tumour effect of gefitinib.


Assuntos
Neoplasias do Colo/tratamento farmacológico , Resistencia a Medicamentos Antineoplásicos , Quinazolinas/farmacologia , Receptor de Insulina/antagonistas & inibidores , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Difosfonatos/farmacologia , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Receptores ErbB/antagonistas & inibidores , Receptores ErbB/metabolismo , Gefitinibe , Humanos , Insulina/farmacologia , Fator de Crescimento Insulin-Like II/farmacologia , Fosforilação , Isoformas de Proteínas/antagonistas & inibidores , Isoformas de Proteínas/biossíntese , RNA Mensageiro/biossíntese , Receptor IGF Tipo 1/biossíntese , Receptor de Insulina/biossíntese , Transdução de Sinais/efeitos dos fármacos , Células Tumorais Cultivadas
3.
Breast Cancer Res Treat ; 96(2): 131-46, 2006 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-16261397

RESUMO

We have previously demonstrated that oestrogen receptor alpha (ERalpha) modulates epidermal growth factor receptor (EGFR)/mitogen-activated protein kinase (MAPK) signalling efficiency in a tamoxifen-resistant MCF-7 breast cancer cell line (Tam-R). In the present study we have investigated whether this cross-talk between EGFR/MAPK and ERalpha signalling pathways is bidirectional by examining the effects of EGFR/MAPK activity on ER functionality in the same cell line. Elevated expression levels of phosphorylated serine 118 (S118) ERalpha were observed in the Tam-R compared to the parental wild type MCF-7 cell line (WT-MCF-7) under basal growth conditions. Phosphorylation of ERalpha at S118 was regulated by the EGFR/MAPK pathway in Tam-R cells being increased in response to amphiregulin (AR) and inhibited by the selective EGFR tyrosine kinase inhibitor, gefitinib and the MEK1/2 inhibitor, PD184352. Recruitment of the co-activators p68 RNA helicase and SRC1 to ERalpha, oestrogen response element (ERE) activity and Tam-R cell growth were similarly EGFR/MAPK-regulated. Chromatin immunoprecipitation (ChIP) studies revealed that in Tam-R cells the ERalpha assembled on the AR gene promoter and this was associated with elevated basal expression of AR mRNA. Furthermore, AR mRNA expression was under the regulation of the EGFR/MAPK and ERalpha signalling pathways. Neutralising antibodies to AR inhibited EGFR/ERK1/2 activity, reduced S118 ERalpha phosphorylation and reduced AR mRNA expression in TAM-R cells. These findings suggest that ERalpha function in Tam-R cells is maintained as a consequence of EGFR/MAPK-mediated phosphorylation at serine residue 118 resulting in the generation of a self-propogating autocrine growth-regulatory loop through the ERalpha-mediated production of AR.


Assuntos
Antineoplásicos Hormonais/farmacologia , Neoplasias da Mama/patologia , Resistencia a Medicamentos Antineoplásicos , Receptores ErbB/metabolismo , Moduladores de Receptor Estrogênico/farmacologia , Receptores de Estrogênio/metabolismo , Transdução de Sinais , Tamoxifeno/farmacologia , Western Blotting , Neoplasias da Mama/tratamento farmacológico , Linhagem Celular Tumoral , Feminino , Humanos , Imuno-Histoquímica , Reação em Cadeia da Polimerase Via Transcriptase Reversa
4.
Endocr Relat Cancer ; 12 Suppl 1: S173-82, 2005 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-16113094

RESUMO

Aberrant signalling through the epidermal growth factor receptor (EGFR) is associated with increased cancer cell proliferation, reduced apoptosis, invasion and angiogenesis. Over-expression of the EGFR is seen in a variety of tumours and is a rational target for antitumour strategies. Among the classes of agent targeting the EGFR are small-molecule inhibitors, which include gefitinib (IRESSA), which acts by preventing EGFR phosphorylation and downstream signal transduction. De novo and acquired resistance, however, have been reported to gefitinib and here we describe evidence which indicates that the type II receptor tyrosine kinases (RTKs) insulin-like growth factor-I receptor (IGF-IR) and/or insulin receptor (InsR) play important roles in the mediation of responses to gefitinib in the de novo- and acquired-resistance phenotypes in several cancer types. Moreover, combination strategies that additionally target the IGF-IR/InsR can enhance the antitumour effects of gefitinib.


Assuntos
Antineoplásicos/uso terapêutico , Receptores ErbB/antagonistas & inibidores , Neoplasias/tratamento farmacológico , Inibidores de Proteínas Quinases/uso terapêutico , Quinazolinas/uso terapêutico , Antineoplásicos/farmacologia , Proliferação de Células , Resistencia a Medicamentos Antineoplásicos , Gefitinibe , Humanos , Inibidores de Proteínas Quinases/farmacologia , Quinazolinas/farmacologia , Transdução de Sinais/efeitos dos fármacos
5.
Endocr Relat Cancer ; 12 Suppl 1: S29-36, 2005 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-16113097

RESUMO

De novo insensitivity and acquired resistance to the selective oestrogen receptor modulator tamoxifen and the pure anti-oestrogen fulvestrant (faslodex) severely limit their effectiveness in breast cancer patients. This is a major clinical problem, since each year upward of 1 million women are dispensed anti-oestrogenic drugs. In order to investigate the phenomenon of anti-oestrogen resistance and to rapidly screen drugs that target the resistance mechanism(s), we have previously established several in vitro breast cancer models that have acquired resistance to anti-hormones. Such cells commonly develop an ability to proliferate after approximately 3 months of exposure to 4-hydroxytamoxifen or fulvestrant, despite an initial endocrine-responsive (i.e. growth-suppressive) phase. The current paper explores the role that growth factor signalling plays in the transition of oestrogen receptor-positive endocrine-responsive breast cancer cells to anti-oestrogen resistance or insensitivity and how we might, in the future, most effectively use anti-growth factor therapies to treat or delay endocrine-resistant states.


Assuntos
Neoplasias da Mama/tratamento farmacológico , Antagonistas de Estrogênios/uso terapêutico , Inibidores do Crescimento/uso terapêutico , Moduladores Seletivos de Receptor Estrogênico/uso terapêutico , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Quimioterapia Combinada , Feminino , Substâncias de Crescimento/metabolismo , Humanos , Fosforilação , Receptores de Superfície Celular/efeitos dos fármacos , Receptores de Superfície Celular/metabolismo , Transdução de Sinais/efeitos dos fármacos
6.
Endocrinology ; 146(11): 4609-18, 2005 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-16037379

RESUMO

There is considerable evidence that the epidermal growth factor receptor (EGFR) and IGF-I receptor (IGF-IR) cross-talk in breast cancer cells. In the present study, we have examined whether EGFR/IGF-IR cross-talk exists in EGFR-positive tamoxifen-resistant variants of MCF-7 (Tam-R) and T47D (T47D-R) breast cancer cell lines. Although Tam-R cells expressed reduced IGF-IR protein levels compared with their wild-type MCF-7 counterparts, phosphorylated IGF-IR protein levels were equivalent in the two cell lines under basal growth conditions, possibly as a consequence of increased IGF-II expression in Tam-R cells. IGF-II activated both IGF-IR and EGFR in Tam-R cells, whereas only activation of IGF-IR was observed in wild-type cells. In contrast, epidermal growth factor rapidly induced EGFR, but not IGF-IR, phosphorylation in Tam-R cells. IGF-II promoted direct association of c-SRC with IGF-IR, phosphorylated c-SRC, and increased EGFR phosphorylation at tyrosine 845, a c-SRC-dependent phosphorylation site. Pretreatment with either AG1024 (IGF-IR-specific inhibitor) or an IGF-II neutralizing antibody inhibited basal IGF-IR, c-SRC, and EGFR phosphorylation, and AG1024 significantly reduced Tam-R basal cell growth. The c-SRC inhibitor SU6656 also inhibited growth, reduced basal and IGF-II-induced c-SRC and EGFR phosphorylation, and blocked EGFR activation by TGFalpha. Similarly, in T47D-R cells, AG1024 and SU6656 inhibited basal and IGF-II-induced phosphorylation of c-SRC and EGFR, and SU6656 reduced TGFalpha-induced EGFR activity. These results suggest the existence of a unidirectional IGF-IR/EGFR cross-talk mechanism whereby IGF-II, acting through the IGF-IR, regulates basal and ligand-activated EGFR signaling and cell proliferation in a c-SRC-dependent manner in Tam-R cells. This cross-talk between IGF-IR and EGFR is not unique to Tam-R cells because this mechanism is also active in a tamoxifen-resistant T47D-R cell line.


Assuntos
Antineoplásicos Hormonais/farmacologia , Neoplasias da Mama/fisiopatologia , Resistencia a Medicamentos Antineoplásicos , Receptores ErbB/metabolismo , Receptor IGF Tipo 1/metabolismo , Transdução de Sinais , Tamoxifeno/farmacologia , Linhagem Celular Tumoral , Receptores ErbB/antagonistas & inibidores , Receptores ErbB/efeitos dos fármacos , Feminino , Humanos , Fator de Crescimento Insulin-Like II/farmacologia , Fosforilação/efeitos dos fármacos , Proteínas Proto-Oncogênicas pp60(c-src)/metabolismo , Receptor Cross-Talk , Receptor IGF Tipo 1/antagonistas & inibidores , Fator de Crescimento Transformador alfa/farmacologia
7.
J Steroid Biochem Mol Biol ; 93(2-5): 257-62, 2005 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-15860268

RESUMO

Recent evidence demonstrates that growth factor networks are highly interactive with the estrogen receptor (ER) in the control of breast cancer growth and development. As such, tumor responses to anti-hormones are likely to be a composite of the ER and growth factor inhibitory activity of these agents, with alterations/aberrations in growth factor signalling providing a mechanism for the development of anti-hormone resistance. In this light, the current article focuses on illustrating the relationship between growth factor signalling and anti-hormone failure in our in-house tumor models of breast cancer and describes how we are now beginning to successfully target their actions to improve the effects of anti-hormonal drugs and to block aggressive disease progression.


Assuntos
Neoplasias da Mama/tratamento farmacológico , Neoplasias da Mama/metabolismo , Substâncias de Crescimento/metabolismo , Resistencia a Medicamentos Antineoplásicos , Quimioterapia Combinada , Receptores ErbB/metabolismo , Feminino , Antagonistas de Hormônios/uso terapêutico , Humanos , Invasividade Neoplásica , Neoplasias Hormônio-Dependentes/tratamento farmacológico , Neoplasias Hormônio-Dependentes/metabolismo , Receptores de Estrogênio/metabolismo , Transdução de Sinais , Tamoxifeno/uso terapêutico , Falha de Tratamento
8.
Endocr Relat Cancer ; 11(4): 793-814, 2004 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-15613453

RESUMO

De novo and acquired resistance to the anti-tumour drug gefitinib (ZD1839; Iressa), a specific epidermal growth factor receptor (EGFR) tyrosine kinase inhibitor (TKI) has been reported. We have determined whether signalling through the IGF-I receptor (IGF-1R) pathway plays a role in the gefitinib-acquired resistance phenotype. Continuous exposure of EGFR-positive MCF-7-derived tamoxifen resistant breast cancer cells (TAM-R) to 1 microM gefitinib resulted in a sustained growth inhibition (90%) for 4 months before the surviving cells resumed proliferation. A stable gefitinib-resistant subline (TAM/TKI-R) was established after a further 2 months and this showed no detectable basal phosphorylated EGFR activity. Compared with the parental TAM-R cells, the TAM/ TKI-R cells demonstrated (a) elevated levels of activated IGF-1R, AKT and protein kinase C (PKC)delta, (b) an increased sensitivity to growth inhibition by the IGF-1R TKI AG1024 and (c) an increased migratory capacity that was reduced by AG1024 treatment. Similarly, the EGFR-positive androgen-independent human prostate cancer cell line DU145 was also continuously challenged with 1 microM gefitinib and, although substantial growth inhibition (60%) was seen initially, a gefitinib-resistant variant (DU145/TKI-R) developed after 3 months. Like their breast cancer counterparts, the DU145/TKI-R cells showed increases in the levels of components of the IGF-1R signalling pathway and an elevated sensitivity to growth inhibition by AG1024 compared with the parent DU145 cell line. Additionally, DU145/TKI-R cell migration was also decreased by this inhibitor. We have therefore concluded that in breast and prostate cancer cells acquired resistance to gefitinib is associated with increased signalling via the IGF-1R pathway, which also plays a role in the invasive capacity of the gefitinib-resistant phenotype.


Assuntos
Antineoplásicos/farmacologia , Resistencia a Medicamentos Antineoplásicos , Quinazolinas/farmacologia , Receptor IGF Tipo 1/metabolismo , Linhagem Celular Tumoral , Movimento Celular , Proliferação de Células , DNA Complementar/metabolismo , Receptores ErbB/antagonistas & inibidores , Receptores ErbB/genética , Receptores ErbB/metabolismo , Gefitinibe , Regulação Neoplásica da Expressão Gênica , Substâncias de Crescimento/genética , Substâncias de Crescimento/metabolismo , Substâncias de Crescimento/farmacologia , Humanos , Receptor IGF Tipo 1/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transdução de Sinais
9.
Endocrinology ; 144(11): 5105-17, 2003 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-12960029

RESUMO

Although many estrogen receptor-positive breast cancers initially respond to antihormones, responses are commonly incomplete with resistance ultimately emerging. Delineation of signaling mechanisms underlying these phenomena would allow development of therapies to improve antihormone response and compromise resistance. This in vitro investigation in MCF-7 breast cancer cells examines whether epidermal growth factor receptor (EGFR) signaling limits antiproliferative and proapoptotic activity of antihormones and ultimately supports development of resistance. It addresses whether the anti-EGFR agent gefitinib (ZD1839/Iressa; TKI: 1 mum) combined with the antihormones 4-hydroxytamoxifen (TAM: 0.1 mum) or fulvestrant (Faslodex; 0.1 mum) enhances growth inhibition and prevents resistance. TAM significantly suppressed MCF-7 growth over wk 2-5, reducing proliferation detected by immunocytochemistry and fluorescence-activated cell sorter cell cycle analysis. A modest apoptotic increase was observed by fluorescence-activated cell sorter and fluorescence microscopy, with incomplete bcl-2 suppression. EGFR induction occurred during TAM response, as measured by immunocytochemistry and Western blotting, with EGFR-positive, highly proliferative resistant growth subsequently emerging. Although TKI alone was ineffective on growth, TAM plus TKI cotreatment exhibited superior antigrowth activity vs. TAM, with no viable cells by wk 12. Cotreatment was more effective in inhibiting proliferation, promoting apoptosis, and eliminating bcl-2. Cotreatment blocked EGFR induction, markedly depleted ERK1/2 MAPK and protein kinase B phosphorylation, and prevented emergence of EGFR-positive resistance. Faslodex plus TKI cotreatment was also a superior antitumor strategy. Thus, increased EGFR evolves during treatment with antihormones, limiting their efficacy and promoting resistance. Gefitinib addition to antihormonal therapy could prove more effective in treating estrogen receptor-positive breast cancer and may combat development of resistance.


Assuntos
Antineoplásicos Hormonais/farmacologia , Neoplasias da Mama/patologia , Neoplasias da Mama/fisiopatologia , Fator de Crescimento Epidérmico/antagonistas & inibidores , Estradiol/análogos & derivados , Estradiol/farmacologia , Antagonistas de Estrogênios/farmacologia , Proteínas Serina-Treonina Quinases , Quinazolinas/farmacologia , Tamoxifeno/análogos & derivados , Tamoxifeno/farmacologia , Apoptose/efeitos dos fármacos , Divisão Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Combinação de Medicamentos , Resistência a Medicamentos/efeitos dos fármacos , Sinergismo Farmacológico , Receptores ErbB/metabolismo , Receptores ErbB/fisiologia , Feminino , Fulvestranto , Gefitinibe , Humanos , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Fosforilação/efeitos dos fármacos , Proteínas Proto-Oncogênicas/metabolismo , Proteínas Proto-Oncogênicas c-akt , Transdução de Sinais/efeitos dos fármacos
10.
Ann N Y Acad Sci ; 963: 104-15, 2002 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-12095935

RESUMO

An increasing body of evidence demonstrates that growth factor networks are highly interactive with estrogen receptor signaling in the control of breast cancer growth. As such, tumor responses to antihormones are likely to be a composite of the estrogen receptor and growth factor inhibitory activity of these agents. The modulation of growth factor networks during endocrine response is examined, and in vitro and clinical evidence is presented that epidermal growth factor receptor signaling, maintained in either an estrogen receptor-dependent or a receptor-independent manner, is critical to antihormone-resistant breast cancer cell growth. The considerable potential of the epidermal growth factor receptor-selective tyrosine kinase inhibitor Iressa (ZD 1839) to efficiently treat, and perhaps even prevent, endocrine-resistant breast cancer is highlighted.


Assuntos
Antineoplásicos/farmacologia , Neoplasias da Mama/metabolismo , Resistencia a Medicamentos Antineoplásicos/fisiologia , Receptores ErbB/metabolismo , Estradiol/análogos & derivados , Receptores de Estrogênio/metabolismo , Transdução de Sinais/efeitos dos fármacos , Anticorpos Monoclonais/farmacologia , Anticorpos Monoclonais Humanizados , Divisão Celular/efeitos dos fármacos , Glândulas Endócrinas , Estradiol/farmacologia , Moduladores de Receptor Estrogênico/farmacologia , Feminino , Fulvestranto , Gefitinibe , Humanos , Proteínas Tirosina Quinases/antagonistas & inibidores , Quinazolinas/farmacologia , Receptor ErbB-2/fisiologia , Transdução de Sinais/fisiologia , Tamoxifeno/farmacologia , Trastuzumab , Células Tumorais Cultivadas
11.
Endocr Relat Cancer ; 8(3): 175-82, 2001 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-11566608

RESUMO

There is an increasing body of evidence demonstrating that growth factor networks are highly interactive with oestrogen receptor (ER) signalling in the control of breast cancer growth. As such, tumour responses to anti- hormones are likely to be a composite of the ER and growth factor inhibitory activity of these agents. The current article examines the modulation of growth factor networks during endocrine response, and presents in vitro and clinical evidence that epidermal growth factor receptor signalling, maintained in either an ER-dependent or -independent manner, is critical to anti- hormonal-resistant breast cancer cell growth. The considerable potential of the epidermal growth factor receptor-selective tyrosine kinase inhibitor, ZD 1839 (Iressa; AstraZeneca) to efficiently treat, and perhaps even prevent, endocrine-resistant breast cancer is highlighted.


Assuntos
Neoplasias da Mama/metabolismo , Resistencia a Medicamentos Antineoplásicos , Receptores ErbB/metabolismo , Receptores de Estrogênio/metabolismo , Animais , Antineoplásicos/uso terapêutico , Antagonistas de Estrogênios/uso terapêutico , Feminino , Humanos , Receptor ErbB-2/metabolismo , Transdução de Sinais , Células Tumorais Cultivadas/efeitos dos fármacos , Células Tumorais Cultivadas/metabolismo
12.
Prostate ; 49(1): 38-47, 2001 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-11550209

RESUMO

BACKGROUND: The effect of an EGF-R selective tyrosine kinase (EGF-RTK) quinazoline inhibitor ZM252868 was determined on the androgen-sensitive human prostatic tumour cell line LNCaP, which can also respond via the EGF-R-regulated growth pathway for cell proliferation. Potential interaction or 'cross-talk' between steroid and the growth factor mitogen-activated protein kinase (MAPK) signalling pathway was also investigated. METHODS: The responses of LNCaP cells to various growth factors in the absence and presence of the EGF-RTK inhibitor and/or steroid and anti-androgen Casodex, was determined using cell population analysis. The effect of the inhibitor on the expression of androgen receptor, EGF-R and activated MAPK was assessed immunocytochemically and changes in the MAPK signalling cascade were also determined using Western blotting techniques. RESULTS: The ZM252868 inhibitor had no effect on LNCaP basal growth. At 100 nM and 1 microM concentrations, the inhibitor reduced the marked EGF- and TGF-alpha-stimulated LNCaP cell growth by 60% and to basal levels, respectively. Both bFGF- and 5alpha-DHT-stimulated growth were unaffected in this concentration range. The inhibitor (1 microM) decreased the expression of immunoreactive EGF-R but had no effect on androgen receptor levels. Activation of MAPK by EGF was noted, being down-regulated by the inhibitor at a concentration of 1 microM. MAPK was not activated by 5alpha-DHT. The anti-androgen Casodex reduced 5alpha-DHT-stimulated cell growth but had no effect on EGF-R mediated LNCaP growth or EGF-stimulated activated MAPK activity. Treatment with EGF and 5alpha-DHT in combination produced an additive effect on cell proliferation, with the anti-androgen and the EGF-RTK inhibitor only reducing the 5alpha-DHT- or EGF-stimulated portion of growth, respectively. CONCLUSIONS: The study demonstrated the efficacy and selectivity of the ZM252868 inhibitor in inhibiting EGF-R mediated LNCaP cell growth. Additionally, no interaction between androgen and EGF-R mediated growth pathways was determined.


Assuntos
Antagonistas de Androgênios/farmacologia , Anilidas/farmacologia , Divisão Celular/efeitos dos fármacos , Inibidores Enzimáticos/farmacologia , Receptores ErbB/fisiologia , Neoplasias da Próstata/patologia , Quinazolinas/farmacologia , Western Blotting , Interações Medicamentosas , Receptores ErbB/efeitos dos fármacos , Humanos , Imuno-Histoquímica , Masculino , Nitrilas , Transdução de Sinais , Compostos de Tosil , Células Tumorais Cultivadas
13.
Endocrinology ; 142(7): 2776-88, 2001 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-11415996

RESUMO

This paper describes the establishment of an antiestrogen-resistant MCF7 breast cancer cell subline (FASMCF) by continuous culture of the estrogen-responsive parental line in steroid-depleted, ICI 182,780 (Faslodex; 10(-7) M)-supplemented medium. After a 3-month period of growth suppression, cells began to proliferate in ICI 182,780 at rates similar to those of untreated wild-type cells. Immunocytochemistry showed these cells to have reduced estrogen receptor and an absence of progesterone receptor proteins. RT-PCR and transient transfection studies with estrogen response element-reporter constructs confirmed that ICI 182,780-suppressed estrogen response element-mediated signaling. FASMCF cells show increased dependence upon epidermal growth factor receptor (EgfR)/mitogen-activated protein kinase (MAPK)-mediated signaling. Thus, EgfR protein and messenger RNA, growth responses to transforming growth factor-alpha, and extracellular signal-regulated kinase 1/2 MAPK activation levels are all increased. Unlike wild-type cells, FASMCF cells are highly sensitive to growth inhibition by an EgfR-specific tyrosine-kinase inhibitor (TKI), ZD1839 (Iressa), and an inhibitor of the activation of MEK1 (MAPKK), PD098059. Short-term ( approximately 3 weeks) withdrawal of cells from antiestrogen had no effect on growth or phenotype, whereas longer withdrawal (>10 weeks) appeared to partially reverse the cellular phenotype with increasing estrogen receptor and decreasing EgfR levels. In subsequent studies FASMCF cells were maintained in TKI, where their growth was again suppressed and secondary TKI resistance failed to develop within the 3-month period in which initial ICI 182,780 resistance arose. Furthermore, wild-type cells similarly maintained in combination ICI 182,780 and TKI treatment conditions remained growth arrested (>6 months), with notable cell loss through both reduced rates of cellular proliferation and increased cell death.


Assuntos
Antineoplásicos/farmacologia , Neoplasias da Mama/fisiopatologia , Receptores ErbB/fisiologia , Estradiol/análogos & derivados , Estradiol/farmacologia , Moduladores de Receptor Estrogênico/farmacologia , Transdução de Sinais , Neoplasias da Mama/patologia , Técnicas Citológicas , Resistência a Medicamentos , Inibidores Enzimáticos/farmacologia , Feminino , Fulvestranto , Gefitinibe , Humanos , Proteínas Quinases Ativadas por Mitógeno/fisiologia , Proteínas Tirosina Quinases/antagonistas & inibidores , Quinazolinas/farmacologia , Fatores de Tempo , Células Tumorais Cultivadas/efeitos dos fármacos
14.
Science ; 291(5509): 1755-9, 2001 Mar 02.
Artigo em Inglês | MEDLINE | ID: mdl-11230685

RESUMO

The ability of intestinal mucosa to absorb dietary ferric iron is attributed to the presence of a brush-border membrane reductase activity that displays adaptive responses to iron status. We have isolated a complementary DNA, Dcytb (for duodenal cytochrome b), which encoded a putative plasma membrane di-heme protein in mouse duodenal mucosa. Dcytb shared between 45 and 50% similarity to the cytochrome b561 family of plasma membrane reductases, was highly expressed in the brush-border membrane of duodenal enterocytes, and induced ferric reductase activity when expressed in Xenopus oocytes and cultured cells. Duodenal expression levels of Dcytb messenger RNA and protein were regulated by changes in physiological modulators of iron absorption. Thus, Dcytb provides an important element in the iron absorption pathway.


Assuntos
Grupo dos Citocromos b/metabolismo , Duodeno/metabolismo , Compostos Férricos/metabolismo , Absorção Intestinal , Mucosa Intestinal/metabolismo , Ferro da Dieta/metabolismo , Oxirredutases/metabolismo , Transfecção , Sequência de Aminoácidos , Anemia/enzimologia , Animais , Linhagem Celular , Clonagem Molecular , Grupo dos Citocromos b/química , Grupo dos Citocromos b/genética , DNA Complementar , Duodeno/enzimologia , Enterócitos/enzimologia , Enterócitos/metabolismo , Indução Enzimática , Hipóxia , Mucosa Intestinal/enzimologia , Ferro da Dieta/administração & dosagem , Masculino , Camundongos , Microvilosidades/enzimologia , Microvilosidades/metabolismo , Dados de Sequência Molecular , Nitroazul de Tetrazólio/metabolismo , Oócitos , Oxirredução , Oxirredutases/química , Oxirredutases/genética , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Sais de Tetrazólio/metabolismo , Tiazóis/metabolismo , Regulação para Cima , Xenopus
15.
Mol Cell Endocrinol ; 171(1-2): 129-35, 2001 Jan 22.
Artigo em Inglês | MEDLINE | ID: mdl-11165021

RESUMO

The development of inhibitors to block the formation of estrone and 5-androstenediol from sulfated precursors is an important new strategy for the treatment of breast cancer. In this study a series of tricyclic coumarin sulfamates (665-668 COUMATE) and a tricyclic oxepin sulfamate have been synthesised and tested for their ability to inhibit estrone sulfatase activity (E1-STS). In addition the effect of the steroid-based E1-STS inhibitor, 2-methoxyestrone-3-O-sulfamate (2-MeOEMATE) on the morphology of MDA-MB-231 cells and breast tumour-derived fibroblasts was also examined. The tricyclic coumarin sulfamates and oxepin sulfamate were potent inhibitors of E1-STS activity with IC(50)s ranging from 8 to 250 nM. Of this series 667 COUMATE was the most potent inhibitor (IC(50)=8 nM) and was three-times more potent than estrone-3-O-sulfamate (EMATE, IC(50)=25 nM). 667 COUMATE did not stimulate the growth of MCF-7 breast cancer cells and is therefore devoid of estrogenicity. In vivo, 667 COUMATE inhibited E1-STS activity in rat liver tissue to a similar extent to that of EMATE. 2-MeOEMATE had a marked effect on the morphology of MDA-MB-231 cells and breast tumour-derived fibroblasts causing a significant increase in the number of rounded cells. 667 COUMATE and 2-MeOEMATE therefore offer considerable potential for development for cancer therapy.


Assuntos
Antineoplásicos/síntese química , Antineoplásicos/farmacologia , Neoplasias da Mama/tratamento farmacológico , Inibidores Enzimáticos/farmacologia , Estrona/análogos & derivados , Ácidos Sulfônicos/química , Ácidos Sulfônicos/farmacologia , Animais , Neoplasias da Mama/patologia , Cumarínicos/farmacologia , Inibidores Enzimáticos/síntese química , Estrogênios/farmacologia , Estrona/farmacologia , Feminino , Humanos , Cinética , Oxepinas , Ratos , Ratos Wistar , Sulfatases/antagonistas & inibidores , Sulfatases/metabolismo , Sulfonamidas/farmacologia , Células Tumorais Cultivadas
16.
Eur J Cancer ; 36 Suppl 4: S34-5, 2000 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-11056309

RESUMO

Selective oestrogen receptor downregulators (SERDs) are a class of highly effective steroidal antitumour agents that reduce cellular levels of the oestrogen receptor (ER). In this study, we compared the efficacy by which three novel molecular approaches: (1) antisense oligonucleotides; (2) antisense RNA; and (3) dominant negative mutants are able to act as SERDs. Using transient and, where appropriate, stable gene transfection experiments we found that constitutive overexpression of ER antisense RNA and a hormone-binding domain compromised dominant-negative ER mutant (DNER-1), were most effective at downregulating ER expression and/or activity in vitro.


Assuntos
Antineoplásicos/uso terapêutico , Estradiol/análogos & derivados , Estradiol/uso terapêutico , Antagonistas de Estrogênios/uso terapêutico , Receptores de Estrogênio/genética , Moduladores Seletivos de Receptor Estrogênico/uso terapêutico , Northern Blotting , Divisão Celular , Células Cultivadas , Fulvestranto , Genes Reporter , Humanos , Oligonucleotídeos Antissenso/metabolismo
18.
Mol Cell ; 5(2): 299-309, 2000 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-10882071

RESUMO

Iron absorption by the duodenal mucosa is initiated by uptake of ferrous Fe(II) iron across the brush border membrane and culminates in transfer of the metal across the basolateral membrane to the portal vein circulation by an unknown mechanism. We describe here the isolation and characterization of a novel cDNA (Ireg1) encoding a duodenal protein that is localized to the basolateral membrane of polarized epithelial cells. Ireg1 mRNA and protein expression are increased under conditions of increased iron absorption, and the 5' UTR of the Ireg1 mRNA contains a functional iron-responsive element (IRE). IREG1 stimulates iron efflux following expression in Xenopus oocytes. We conclude that IREG1 represents the long-sought duodenal iron export protein and is upregulated in the iron overload disease, hereditary hemochromatosis.


Assuntos
Proteínas de Transporte/metabolismo , Proteínas de Transporte de Cátions , Polaridade Celular , Duodeno/metabolismo , Absorção Intestinal/fisiologia , Mucosa Intestinal/metabolismo , Ferro/metabolismo , Proteínas de Membrana/metabolismo , Veia Porta/metabolismo , Sequência de Aminoácidos , Animais , Sequência de Bases , Transporte Biológico , Proteínas de Transporte/genética , Proteínas de Transporte/isolamento & purificação , Compartimento Celular , Clonagem Molecular , Compostos Férricos/metabolismo , Regulação da Expressão Gênica , Proteínas de Membrana/genética , Proteínas de Membrana/isolamento & purificação , Camundongos , Modelos Biológicos , Dados de Sequência Molecular , RNA Mensageiro/genética , Elementos de Resposta , Homologia de Sequência de Aminoácidos , Homologia de Sequência do Ácido Nucleico , Transferrina/deficiência
19.
Neurosurgery ; 46(4): 910-6; discussion 916-7, 2000 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-10764264

RESUMO

OBJECTIVE: To determine the protective effects of various periprocedural interventions in the prevention of cerebral ischemia as a complication of endovascular carotid sacrifice (ECS). METHODS: Thirty-two cases of ECS performed at our institution, between October 1987 and July 1998, were reviewed. Fifteen patients underwent superficial temporal artery-to-middle cerebral artery bypass surgery. In 21 patients, the carotid artery was occluded proximal to the target lesion; and in 11 patients, a lesion trapping procedure was performed. Six patients were prophylactically anticoagulated, 14 received antiplatelet agents prophylactically, and 12 received no pharmacoprophylaxis. RESULTS: Among the six patients who were anticoagulated, there were no embolic events. Embolic events affected 4 of 14 patients receiving prophylactic antiplatelet agents, 2 of 12 patients receiving no pharmacoprophylaxis, 1 of 11 patients who underwent a trapping procedure, and 5 of 21 patients whose carotid artery was occluded proximal to the target lesion. Postocclusion cerebral ischemia developed in 7 of 15 patients who underwent bypass surgery, and in 1 of the remaining 17. CONCLUSION: Superficial temporal artery-to-middle cerebral artery bypass did not protect against postocclusion cerebral ischemia after ECS in this series (P = 0.01). Although the small number of patients studied precludes statistical validity (P = 0.29), the trends suggest that antiplatelet agents provide no protection against postocclusion cerebral emboli after ECS. Prophylactic anticoagulation (P = 0.32) and lesion trapping (P = 0.12) may reduce the frequency of postocclusion embolic events after ECS; however, because of the small number of patients, statistical significance could not be demonstrated.


Assuntos
Isquemia Encefálica/prevenção & controle , Artérias Carótidas/cirurgia , Revascularização Cerebral , Complicações Pós-Operatórias/prevenção & controle , Idoso , Anticoagulantes/uso terapêutico , Isquemia Encefálica/etiologia , Doenças das Artérias Carótidas/cirurgia , Doenças das Artérias Carótidas/terapia , Feminino , Humanos , Aneurisma Intracraniano/cirurgia , Aneurisma Intracraniano/terapia , Embolia Intracraniana/etiologia , Masculino , Pessoa de Meia-Idade , Inibidores da Agregação Plaquetária/uso terapêutico , Medicina Preventiva/métodos , Estudos Retrospectivos , Falha de Tratamento
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA