Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
1.
Nat Rev Cancer ; 7(9): 659-72, 2007 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-17721431

RESUMO

Breast cancer is not a single disease, but is instead a collection of diseases that have distinct histopathological features, genetic and genomic variability, and diverse prognostic outcomes. Thus, no individual model would be expected to completely recapitulate this complex disease. Here, the models commonly used to investigate breast cancer including cell lines, xenografts and genetically engineered mice, are discussed to help address the question: what is the most powerful way to investigate this heterogeneous disease?


Assuntos
Neoplasias da Mama , Modelos Animais de Doenças , Animais , Linhagem Celular Tumoral , Previsões , Camundongos , Camundongos Transgênicos , Transplante de Neoplasias , Ratos
2.
Breast Cancer Res ; 15(5): R91, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-24074261

RESUMO

INTRODUCTION: The Rho GTPase Cdc42 is overexpressed and hyperactivated in breast tumors compared to normal breast tissue. Cdc42 regulates key processes that are critical for mammary gland morphogenesis and become disrupted during the development, progression, and metastasis of breast cancer. However, the contribution of Cdc42 to normal and neoplastic mammary gland development in vivo remains poorly understood. We were therefore interested in investigating the effects of Cdc42 overexpression on mammary gland morphogenesis as a first step toward understanding how its overexpression may contribute to mammary tumorigenesis. METHODS: We developed a tetracycline-regulatable Cdc42 overexpression mouse model in which Cdc42 can be inducibly overexpressed in the developing mammary gland. The effects of Cdc42 overexpression during postnatal mammary gland development were investigated using in vivo and in vitro approaches, including morphometric analysis of wholemounted mammary glands, quantification of histological markers, and primary mammary epithelial cell (MEC) functional and biochemical assays. RESULTS: Analysis of Cdc42-overexpressing mammary glands revealed abnormal terminal end bud (TEB) morphologies, characterized by hyperbudding and trifurcation, and increased side branching within the ductal tree. Quantification of markers of proliferation and apoptosis suggested that these phenotypes were not due to increased cell proliferation or survival. Rather, Cdc42 overexpressing MECs were more migratory and contractile and formed dysmorphic, invasive acini in three-dimensional cultures. Cdc42 and RhoA activities, phosphorylated myosin light chain, and MAPK signaling, which contribute to migration and invasion, were markedly elevated in Cdc42 overexpressing MECs. Interestingly, Cdc42 overexpressing mammary glands displayed several features associated with altered epithelial-stromal interactions, which are known to regulate branching morphogenesis. These included increased stromal thickness and collagen deposition, and stromal cells isolated from Cdc42 overexpressing mammary glands exhibited elevated mRNA expression of extracellular matrix proteins and remodeling enzymes. CONCLUSIONS: These data suggest that Cdc42 overexpression disrupts mammary gland branching morphogenesis by altering Rho GTPase and MAPK signaling, leading to increased MEC contractility and migration in association with stromal alterations. Our studies provide insight into how aberrant Cdc42 expression may contribute to mammary tumorigenesis.


Assuntos
Expressão Gênica , Glândulas Mamárias Animais/metabolismo , Glândulas Mamárias Animais/patologia , Proteína cdc42 de Ligação ao GTP/genética , Animais , Neoplasias da Mama/genética , Neoplasias da Mama/patologia , Linhagem Celular , Movimento Celular/genética , Proliferação de Células , Sobrevivência Celular/genética , Transformação Celular Neoplásica/genética , Células Epiteliais/metabolismo , Células Epiteliais/patologia , Feminino , Humanos , Sistema de Sinalização das MAP Quinases , Camundongos , Camundongos Transgênicos , Células Estromais/metabolismo , Proteínas rac1 de Ligação ao GTP/metabolismo , Proteína rhoA de Ligação ao GTP/metabolismo
3.
Cancer Cell ; 7(5): 485-96, 2005 May.
Artigo em Inglês | MEDLINE | ID: mdl-15894268

RESUMO

We developed a rodent model that mimics the osteoblastic and osteolytic changes associated with human metastatic prostate cancer. Microarray analysis identified MMP-7, cathepsin-K, and apolipoprotein D as being upregulated at the tumor-bone interface. MMP-7, which was produced by osteoclasts at the tumor-bone interface, was capable of processing RANKL to a soluble form that promoted osteoclast activation. MMP-7-deficient mice demonstrated reduced prostate tumor-induced osteolysis and RANKL processing. This study suggests that inhibition of MMP-7 will have therapeutic benefit in the treatment of prostate cancer-induced osteolysis.


Assuntos
Proteínas de Transporte/metabolismo , Metaloproteinase 7 da Matriz/metabolismo , Glicoproteínas de Membrana/metabolismo , Osteólise/patologia , Neoplasias da Próstata/metabolismo , Fosfatase Ácida/metabolismo , Actinas/metabolismo , Animais , Proteínas de Transporte/genética , Proteínas de Transporte/farmacologia , Modelos Animais de Doenças , Regulação para Baixo/genética , Expressão Gênica/genética , Perfilação da Expressão Gênica , Glicoproteínas/genética , Humanos , Isoenzimas/metabolismo , Masculino , Metaloproteinase 7 da Matriz/genética , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/farmacologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Modelos Biológicos , Monócitos/efeitos dos fármacos , Monócitos/metabolismo , Osteoclastos/metabolismo , Osteoclastos/patologia , Osteólise/etiologia , Osteólise/metabolismo , Osteoprotegerina , Proteína Relacionada ao Hormônio Paratireóideo/genética , Neoplasias da Próstata/complicações , Ligante RANK , Ratos , Ratos Endogâmicos F344 , Receptor Ativador de Fator Nuclear kappa-B , Receptores Citoplasmáticos e Nucleares/genética , Receptores do Fator de Necrose Tumoral/genética , Crânio/patologia , Fosfatase Ácida Resistente a Tartarato , Regulação para Cima/genética
4.
Curr Drug Targets ; 23(2): 220, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35139781

RESUMO

Meet the editorial board member page has been retracted at the request of editorial board member of the journal "Current Drug Targets". Bentham Science apologizes to the readers of the journal for any inconvenience this may have caused. The Bentham Editorial Policy on Article Withdrawal can be found at https://benthamscience.com/editorial-policies-main.php. BENTHAM SCIENCE DISCLAIMER: It is a condition of publishers that manuscripts submitted to this journal should not be simultaneously submitted or published elsewhere. Furthermore, any data, illustration, structure or table that has been published elsewhere must be reported, and copyright permission for reproduction must be obtained. Plagiarism is strictly forbidden, and while submitting the article for publication, the authors agree that the publishers have the legal right to take appropriate action against the authors if plagiarism or fabricated information is discovered. By submitting a manuscript, the authors agree that the copyright of their article is transferred to the publishers, if and when the article is accepted for publication.

5.
Curr Drug Targets ; 23(1): 1, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35048801

RESUMO

Meet the editorial board member page has been retracted at the request of editorial board member of the journal "Current Drug Targets". Bentham Science apologizes to the readers of the journal for any inconvenience this may have caused. The Bentham Editorial Policy on Article Withdrawal can be found at https://benthamscience.com/editorial-policies-main.php. BENTHAM SCIENCE DISCLAIMER: It is a condition of publishers that manuscripts submitted to this journal should not be simultaneously submitted or published elsewhere. Furthermore, any data, illustration, structure or table that has been published elsewhere must be reported, and copyright permission for reproduction must be obtained. Plagiarism is strictly forbidden, and while submitting the article for publication, the authors agree that the publishers have the legal right to take appropriate action against the authors if plagiarism or fabricated information is discovered. By submitting a manuscript, the authors agree that the copyright of their article is transferred to the publishers, if and when the article is accepted for publication.

6.
Breast Cancer Res ; 12(5): R73, 2010.
Artigo em Inglês | MEDLINE | ID: mdl-20860838

RESUMO

INTRODUCTION: Rho GTPases are overexpressed and hyperactivated in human breast cancers. Deficiency of p190B RhoGAP, a major inhibitor of the Rho GTPases, inhibits mouse mammary tumor virus long terminal repeat (MMTV)-Neu/ErbB2 mammary tumor formation and progression in part through effects within the stromal environment, suggesting that p190B function is pro-tumorigenic. To further investigate the potential pro-tumorigenic actions of p190B, we examined the effects of exogenous p190B expression within the mammary epithelium on MMTV-Neu tumor formation and progression. METHODS: Tetracycline-regulatable p190B transgenic mice were bred to MMTV-Neu mice, and the effects of exogenous p190B expression on tumor latency, multiplicity, growth rates, angiogenesis, and metastasis were examined. The effects of exogenous p190B expression on cell-matrix adhesion and invasion were tested using non-transformed primary mammary epithelial cells (MECs). Rho GTPase activity, oxidative stress as an indicator of reactive oxygen species (ROS) production, and downstream signaling pathways were analyzed. RESULTS: Altered p190B expression resulted in a 2-fold increase in tumor multiplicity and a 3-fold increase in metastases compared to control mice indicating that exogenous p190B expression in the mammary epithelium promotes MMTV-Neu mammary tumor formation and progression. Interestingly, non-transformed primary MECs expressing exogenous p190B displayed increased adhesion to laminin and type IV collagen and formed invasive structures in a three-dimensional culture assay. Ras related C3 botulinum toxin 1 (Rac1)-GTP levels were elevated in p190B transgenic tumors whereas Ras homologous A (RhoA) and cell division cycle 42 (Cdc42)-GTP levels were not significantly altered. Rac1 activity affects production of ROS, which regulate transformation, metastasis, and oxidative stress. Protein carbonylation, which is indicative of oxidative stress, was elevated 1.75-fold in p190B transgenic tumors as compared to control tumors suggesting that exogenous p190B expression may affect Rac1-dependent ROS production. CONCLUSIONS: These studies indicate that paradoxically, p190B RhoGAP, a major inhibitor of the Rho GTPases in vitro, has pro-tumorigenic functions that enhance MMTV-Neu induced mammary tumor formation and metastasis. Furthermore, exogenous p190B expression enhances cell adhesion and invasion, which may facilitate metastasis. Rac1 activity and oxidative stress are elevated in tumors expressing exogenous p190B suggesting that p190B may promote tumorigenesis through a Rac1/ROS dependent mechanism.


Assuntos
Transformação Celular Neoplásica/metabolismo , Proteínas Ativadoras de GTPase/metabolismo , Neoplasias Mamárias Animais/metabolismo , Vírus do Tumor Mamário do Camundongo/metabolismo , Animais , Adesão Celular , Transformação Celular Neoplásica/genética , Junções Célula-Matriz , Células Cultivadas , Células Epiteliais/metabolismo , Células Epiteliais/patologia , Feminino , Proteínas Ativadoras de GTPase/genética , Glândulas Mamárias Animais/metabolismo , Glândulas Mamárias Animais/patologia , Neoplasias Mamárias Animais/patologia , Vírus do Tumor Mamário do Camundongo/genética , Camundongos , Metástase Neoplásica , Neovascularização Patológica , Estresse Oxidativo , Espécies Reativas de Oxigênio , Proteína cdc42 de Ligação ao GTP/biossíntese , Proteínas rac1 de Ligação ao GTP/biossíntese , Proteína rhoA de Ligação ao GTP/biossíntese , Proteína rhoA de Ligação ao GTP/metabolismo
7.
J Cell Biol ; 171(4): 663-73, 2005 Nov 21.
Artigo em Inglês | MEDLINE | ID: mdl-16301332

RESUMO

Members of the fibroblast growth factor (FGF) family and the FGF receptors (FGFRs) have been implicated in mediating various aspects of mammary gland development and transformation. To elucidate the molecular mechanisms of FGFR1 action in a context that mimics polarized epithelial cells, we have developed an in vitro three-dimensional HC11 mouse mammary epithelial cell culture model expressing a drug-inducible FGFR1 (iFGFR1). Using this conditional model, iFGFR1 activation in these growth-arrested and polarized mammary acini initially led to reinitiation of cell proliferation, increased survival of luminal cells, and loss of cell polarity, resulting in the disruption of acinar structures characterized by the absence of an empty lumen. iFGFR1 activation also resulted in a gain of invasive properties and the induction of matrix metalloproteinase 3 (MMP-3), causing the cleavage of E-cadherin and increased expression of smooth muscle actin and vimentin. The addition of a pan MMP inhibitor abolished these phenotypes but did not prevent the effects of iFGFR1 on cell proliferation or survival.


Assuntos
Células Epiteliais/citologia , Glândulas Mamárias Animais/citologia , Receptor Tipo 1 de Fator de Crescimento de Fibroblastos/fisiologia , Animais , Apoptose , Membrana Basal/metabolismo , Caderinas/metabolismo , Linhagem Celular , Movimento Celular , Proliferação de Células , Sobrevivência Celular , Dimerização , Epitélio/metabolismo , Técnica Indireta de Fluorescência para Anticorpo , Imageamento Tridimensional , Immunoblotting , Metaloproteinase 3 da Matriz/biossíntese , Metaloproteinase 3 da Matriz/metabolismo , Mesoderma/metabolismo , Camundongos , Microscopia Confocal , Modelos Biológicos , Fenótipo , Plasmídeos/metabolismo , RNA Interferente Pequeno/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transdução de Sinais , Fatores de Tempo , Cicatrização
8.
Breast Cancer Res ; 11(4): R61, 2009.
Artigo em Inglês | MEDLINE | ID: mdl-19703301

RESUMO

INTRODUCTION: Rho signaling regulates key cellular processes including proliferation, survival, and migration, and it has been implicated in the development of many types of cancer including breast cancer. P190B Rho GTPase activating protein (RhoGAP) functions as a major inhibitor of the Rho GTPases. P190B is required for mammary gland morphogenesis, and overexpression of p190B in the mammary gland induces hyperplastic lesions. Hence, we hypothesized that p190B may play a pivotal role in mammary tumorigenesis. METHODS: To investigate the effects of loss of p190B function on mammary tumor progression, p190B heterozygous mice were crossed with an MMTV-Neu breast cancer model. Effects of p190B deficiency on tumor latency, multiplicity, growth, preneoplastic progression and metastasis were evaluated. To investigate potential differences in tumor angiogenesis between the two groups, immunohistochemistry to detect von Willebrand factor was performed and quantified. To examine gene expression of potential mediators of the angiogenic switch, an angiogenesis PCR array was utilized and results were confirmed using immunohistochemistry. Finally, reciprocal transplantation of tumor fragments was performed to determine the impact of stromal deficiency of p190B on tumor angiogenesis. RESULTS: P190B deficiency reduced tumor penetrance (53% of p190B+/-Neu mice vs. 100% of p190B+/+Neu mice formed tumors) and markedly delayed tumor onset by an average of 46 weeks. Tumor multiplicity was also decreased, but an increase in the number of preneoplastic lesions was detected indicating that p190B deficiency inhibited preneoplastic progression. Angiogenesis was decreased in the p190B heterozygous tumors, and expression of a potent angiogenic inhibitor, thrombospondin-1, was elevated in p190B+/-Neu mammary glands. Transplantation of p190B+/-Neu tumor fragments into wild-type recipients restored tumor angiogenesis. Strikingly, p190B+/+Neu tumor fragments were unable to grow when transplanted into p190B+/-Neu recipients. CONCLUSIONS: These data suggest that p190B haploinsufficiency in the epithelium inhibits MMTV-Neu tumor initiation. Furthermore, p190B deficiency in the vasculature is responsible, in part, for the inhibition of MMTV-Neu tumor progression.


Assuntos
Adenocarcinoma/genética , Proteínas Ativadoras de GTPase/deficiência , Neoplasias Mamárias Experimentais/genética , Proteínas de Neoplasias/fisiologia , Adenocarcinoma/irrigação sanguínea , Adenocarcinoma/enzimologia , Adenocarcinoma/patologia , Animais , Cruzamentos Genéticos , Progressão da Doença , Células Epiteliais/metabolismo , Células Epiteliais/patologia , Feminino , Proteínas Ativadoras de GTPase/genética , Proteínas Ativadoras de GTPase/fisiologia , Heterozigoto , Neoplasias Mamárias Experimentais/irrigação sanguínea , Neoplasias Mamárias Experimentais/enzimologia , Neoplasias Mamárias Experimentais/patologia , Neoplasias Mamárias Experimentais/virologia , Vírus do Tumor Mamário do Camundongo , Camundongos , Camundongos Mutantes , Camundongos SCID , Proteínas de Neoplasias/deficiência , Proteínas de Neoplasias/genética , Transplante de Neoplasias , Neovascularização Patológica/genética , Neovascularização Patológica/patologia , Receptor ErbB-2 , Transdução de Sinais , Células Estromais/metabolismo , Células Estromais/patologia , Trombospondina 1/biossíntese , Trombospondina 1/genética
9.
Cancer Res ; 67(14): 6760-7, 2007 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-17638887

RESUMO

To delineate the role of matrix metalloproteinase 7 (MMP7) in mammary tumorigenesis, MMP7 was expressed in the normal murine mammary gland cell line, c57MG. MMP7 markedly enhanced the growth rate of the c57MG cells in three-dimensional culture and promoted tumor formation in vivo. Subsequent investigation showed that MMP7 (a) up-regulated ErbB4 receptor levels, (b) solubilized the ErbB4 receptor cognate ligand heparin-bound epidermal growth factor, and (c) mediated the proteolytic processing of ErbB4 to yield a soluble intracellular domain (ICD) that localized to the cytoplasm and the nucleus. Furthermore, overexpression of the ErbB4 ICD in the c57MG cell line recapitulated the proliferative effects of MMP7 in vitro and in vivo. These data indicate a novel mechanism for mammary epithelial cell transformation by MMP7.


Assuntos
Transformação Celular Neoplásica , Receptores ErbB/biossíntese , Regulação Neoplásica da Expressão Gênica , Glândulas Mamárias Animais/metabolismo , Neoplasias Mamárias Animais/metabolismo , Metaloproteinase 7 da Matriz/fisiologia , Animais , Linhagem Celular Tumoral , Núcleo Celular/metabolismo , Proliferação de Células , Células Epiteliais , Humanos , Receptores de Hialuronatos/biossíntese , Metaloproteinase 7 da Matriz/metabolismo , Camundongos , Modelos Biológicos , Receptor ErbB-4
10.
Acta Biomater ; 82: 122-132, 2018 12.
Artigo em Inglês | MEDLINE | ID: mdl-30316022

RESUMO

A preclinical murine model of hydroxyapatite (HA) breast microcalcifications (µcals), which are an important clinical biomarker for breast cancer detection, was used to investigate the independent effects of high affinity bisphosphonate (BP) ligands and a polyethylene glycol (PEG) spacer on targeted delivery of gold nanoparticles (Au NPs) for contrast-enhanced radiographic detection. The addition of BP ligands to PEGylated Au NPs (BP-PEG-Au NPs) resulted in five-fold greater binding affinity for targeting HA µcals, as expected, due to the strong binding affinity of BP ligands for calcium. Therefore, BP-PEG-Au NPs were able to target HA µcals in vivo after intramammary delivery, which enabled contrast-enhanced radiographic detection of µcals in both normal and radiographically dense mammary tissues similar to previous results for BP-Au NPs, while PEG-Au NPs did not. The addition of a PEG spacer between the BP targeting ligand and Au NP surface enabled improved in vivo clearance. PEG-Au NPs and BP-PEG-Au NPs were cleared from all mammary glands (MGs) and control MGs, respectively, within 24-48 h after intramammary delivery, while BP-Au NPs were not. PEGylated Au NPs were slowly cleared from MGs by lymphatic drainage and accumulated in the spleen. Histopathology revealed uptake of PEG-Au NPs and BP-PEG-Au NPs by macrophages in the spleen, liver, and MGs; there was no evidence of toxicity due to the accumulation of NPs in organs and tissues compared with untreated controls for up to 28 days after delivery. STATEMENT OF SIGNIFICANCE: Au NP imaging probes and therapeutics are commonly surface functionalized with PEG and/or high affinity targeting ligands for delivery. However, direct comparisons of PEGylated Au NPs with and without a targeting ligand, or ligand-targeted Au NPs with and without a PEG spacer, on in vivo targeting efficiency, biodistribution, and clearance are limited. Therefore, the results of this study are important for the rationale design of targeted NP imaging probes and therapeutics, including the translation of BP-PEG-Au NPs which enable improved sensitivity and specificity for the radiographic detection of abnormalities (e.g., µcals) in women with dense breast tissue.


Assuntos
Calcinose , Difosfonatos , Sistemas de Liberação de Medicamentos , Ouro , Neoplasias Mamárias Experimentais , Nanopartículas Metálicas , Animais , Calcinose/diagnóstico por imagem , Calcinose/tratamento farmacológico , Calcinose/metabolismo , Calcinose/patologia , Difosfonatos/química , Difosfonatos/farmacocinética , Difosfonatos/farmacologia , Feminino , Ouro/química , Ouro/farmacocinética , Ouro/farmacologia , Neoplasias Mamárias Experimentais/diagnóstico por imagem , Neoplasias Mamárias Experimentais/tratamento farmacológico , Neoplasias Mamárias Experimentais/metabolismo , Neoplasias Mamárias Experimentais/patologia , Nanopartículas Metálicas/química , Nanopartículas Metálicas/uso terapêutico , Camundongos , Polietilenoglicóis/química , Polietilenoglicóis/farmacocinética , Polietilenoglicóis/farmacologia
11.
Mol Endocrinol ; 20(6): 1391-405, 2006 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-16469769

RESUMO

p190-B Rho GTPase activating protein is essential for mammary gland development because p190-B deficiency prevents ductal morphogenesis. To investigate the role of p190-B during distinct stages of mammary gland development, tetracycline-regulatable p190-B-overexpressing mice were generated. Short-term induction of p190-B in the developing mammary gland results in abnormal terminal end buds (TEBs) that exhibit aberrant budding off the neck, histological anomalies, and a markedly thickened stroma. Overexpression of p190-B throughout postnatal development results in increased branching, delayed ductal elongation, and disorganization of the ductal tree. Interestingly, overexpression of p190-B during pregnancy results in hyperplastic lesions. Several cellular and molecular alterations detected within the aberrant TEBs may contribute to these phenotypes. Signaling through the IGF pathway is altered, and the myoepithelial cell layer is discontinuous at sites of aberrant budding. An increase in collagen and extensive infiltration of macrophages, which have recently been implicated in branching morphogenesis, is observed in the stroma surrounding the p190-B-overexpressing TEBs. We propose that the stromal response, disruption of the myoepithelial layer, and alterations in IGF signaling in the p190-B-overexpressing mice impact the TEB architecture, leading to disorganization and increased branching of the ductal tree. Moreover, we suggest that alterations in tissue architecture and the adjacent stroma as a consequence of p190-B overexpression during pregnancy leads to loss of growth control and the formation of hyperplasia. These data demonstrate that precise control of p190-B Rho GTPase-activating protein activity is critical for normal branching morphogenesis during mammary gland development.


Assuntos
Proteínas de Transporte/genética , Glândulas Mamárias Animais/patologia , Animais , Sequência de Bases , Proteínas de Transporte/metabolismo , DNA Complementar/genética , Proteínas de Ligação a DNA , Feminino , Proteínas Ativadoras de GTPase , Expressão Gênica , Fatores de Troca do Nucleotídeo Guanina , Humanos , Hiperplasia , Glândulas Mamárias Animais/crescimento & desenvolvimento , Glândulas Mamárias Animais/metabolismo , Camundongos , Camundongos Endogâmicos , Camundongos Transgênicos , Fenótipo , Gravidez , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Proteínas Repressoras , Transdução de Sinais
12.
Cancer Res ; 62(19): 5559-63, 2002 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-12359768

RESUMO

Overexpression of the matrix metalloproteinase matrilysin (matrix metalloproteinase-7) in the mouse mammary gland promotes mammary hyperplasia and accelerates the onset of oncogene-induced mammary tumors. In cell culture models, acute exposure of cells coexpressing Fas and Fas ligand (FasL) to matrilysin induces apoptosis, whereas chronic exposure to matrilysin selects for apoptosis-resistant cells. We now demonstrate that matrilysin promotes resistance to apoptosis in vivo. Matrilysin expression increased apoptosis in the involuting mammary gland of mice that had undergone a single pregnancy and lactation cycle. Premature basement membrane disruption was detected in matrilysin-expressing mice, which could account for the increase in apoptosis. However, multiparous mice, in which the involuting mammary epithelial cells have been repeatedly exposed to matrilysin, show a significant decrease in apoptosis. Mammary tissue from multiparous matrilysin-expressing mice showed decreased FasL expression, suggesting that loss of FasL is at least one mechanism of matrilysin-induced resistance to apoptosis. We propose that matrilysin promotes mammary tumor formation by enhancing the selection of cells that are resistant to apoptosis.


Assuntos
Apoptose/fisiologia , Glândulas Mamárias Animais/enzimologia , Metaloproteinase 7 da Matriz/fisiologia , Animais , Feminino , Hiperplasia/enzimologia , Hiperplasia/patologia , Lactação , Glândulas Mamárias Animais/patologia , Vírus do Tumor Mamário do Camundongo/enzimologia , Vírus do Tumor Mamário do Camundongo/genética , Metaloproteinase 7 da Matriz/biossíntese , Metaloproteinase 7 da Matriz/genética , Camundongos , Camundongos Transgênicos , Gravidez
13.
Adv Drug Deliv Rev ; 99(Pt A): 12-27, 2016 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-26482186

RESUMO

The high concentration of mineral present in bone and pathological calcifications is unique compared with all other tissues and thus provides opportunity for targeted delivery of pharmaceutical drugs, including radiosensitizers and imaging probes. Targeted delivery enables accumulation of a high local dose of a therapeutic or imaging contrast agent to diseased bone or pathological calcifications. Bisphosphonates (BPs) are the most widely utilized bone-targeting ligand due to exhibiting high binding affinity to hydroxyapatite mineral. BPs can be conjugated to an agent that would otherwise have little or no affinity for the sites of interest. This article summarizes the current state of knowledge and practice for the use of BPs as ligands for targeted delivery to bone and mineral deposits. The clinical history of BPs is briefly summarized to emphasize the success of these molecules as therapeutics for metabolic bone diseases. Mechanisms of binding and the relative binding affinity of various BPs to bone mineral are introduced, including common methods for measuring binding affinity in vitro and in vivo. Current research is highlighted for the use of BP ligands for targeted delivery of BP conjugates in various applications, including (1) therapeutic drug delivery for metabolic bone diseases, bone cancer, other bone diseases, and engineered drug delivery platforms; (2) imaging probes for scintigraphy, fluorescence, positron emission tomography, magnetic resonance imaging, and computed tomography; and (3) radiotherapy. Last, and perhaps most importantly, key structure-function relationships are considered for the design of drugs with BP ligands, including the tether length between the BP and drug, the size of the drug, the number of BP ligands per drug, cleavable tethers between the BP and drug, and conjugation schemes.


Assuntos
Osso e Ossos/metabolismo , Difosfonatos/administração & dosagem , Sistemas de Liberação de Medicamentos , Animais , Difosfonatos/química , Difosfonatos/farmacocinética , Desenho de Fármacos , Humanos , Ligantes , Minerais/metabolismo
14.
ACS Nano ; 9(9): 8923-32, 2015 Sep 22.
Artigo em Inglês | MEDLINE | ID: mdl-26308767

RESUMO

Breast density reduces the accuracy of mammography, motivating methods to improve sensitivity and specificity for detecting abnormalities within dense breast tissue, but preclinical animal models are lacking. Therefore, the objectives of this study were to investigate a murine model of radiographically dense mammary tissue and contrast-enhanced X-ray detection of microcalcifications in dense mammary tissue by targeted delivery of bisphosphonate-functionalized gold nanoparticles (BP-Au NPs). Mammary glands (MGs) in the mouse mammary tumor virus - polyomavirus middle T antigen (MMTV-PyMT or PyMT) model exhibited greater radiographic density with age and compared with strain- and age-matched wild-type (WT) controls at 6-10 weeks of age. The greater radiographic density of MGs in PyMT mice obscured radiographic detection of microcalcifications that were otherwise detectable in MGs of WT mice. However, BP-Au NPs provided enhanced contrast for the detection of microcalcifications in both radiographically dense (PyMT) and WT mammary tissues as measured by computed tomography after intramammary delivery. BP-Au NPs targeted microcalcifications to enhance X-ray contrast with surrounding mammary tissue, which resulted in improved sensitivity and specificity for detection in radiographically dense mammary tissues.


Assuntos
Calcinose/diagnóstico , Meios de Contraste/administração & dosagem , Glândulas Mamárias Humanas/ultraestrutura , Nanopartículas Metálicas/administração & dosagem , Radiografia , Animais , Calcinose/patologia , Meios de Contraste/química , Difosfonatos/administração & dosagem , Difosfonatos/química , Ouro/química , Humanos , Nanopartículas Metálicas/química , Camundongos
15.
Nanomedicine (Lond) ; 10(2): 321-41, 2015 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-25600973

RESUMO

Computed tomography enables 3D anatomic imaging at a high spatial resolution, but requires delivery of an x-ray contrast agent to distinguish tissues with similar or low x-ray attenuation. Gold nanoparticles (AuNPs) have gained recent attention as an x-ray contrast agent due to exhibiting a high x-ray attenuation, nontoxicity and facile synthesis and surface functionalization for colloidal stability and targeted delivery. Potential diagnostic applications include blood pool imaging, passive targeting and active targeting, where actively targeted AuNPs could enable molecular imaging by computed tomography. This article summarizes the current state of knowledge for AuNP x-ray contrast agents within a paradigm of key structure-property-function relationships in order to provide guidance for the design of AuNP contrast agents to meet the necessary functional requirements in a particular application. Functional requirements include delivery to the site of interest (e.g., blood, tumors or microcalcifications), nontoxicity during delivery and clearance, targeting or localization at the site of interest and contrast enhancement for the site of interest compared with surrounding tissues. Design is achieved by strategically controlling structural characteristics (composition, mass concentration, size, shape and surface functionalization) for optimized properties and functional performance. Examples from the literature are used to highlight current design trade-offs that exist between the different functional requirements.


Assuntos
Meios de Contraste/química , Diagnóstico por Imagem/métodos , Ouro/química , Nanopartículas Metálicas/química , Tomografia Computadorizada por Raios X/métodos
16.
Biomaterials ; 35(7): 2312-21, 2014 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-24360718

RESUMO

Microcalcifications are one of the most common abnormalities detected by mammography for the diagnosis of breast cancer. However, the detection of microcalcifications and correct diagnosis of breast cancer are limited by the sensitivity and specificity of mammography. Therefore, the objective of this study was to investigate the potential of bisphosphonate-functionalized gold nanoparticles (BP-Au NPs) for contrast-enhanced radiographic detection of breast microcalcifications using two models of breast microcalcifications, which allowed for precise control over levels of hydroxyapatite (HA) mineral within a low attenuating matrix. First, an in vitro imaging phantom was prepared with varying concentrations of HA uniformly dispersed in an agarose hydrogel. The X-ray attenuation of HA-agarose compositions labeled by BP-Au NPs was increased by up to 26 HU compared to unlabeled compositions for HA concentrations ranging from 1 to 10 mg/mL. Second, an ex vivo tissue model was developed to more closely mimic the heterogeneity of breast tissue by injecting varying concentrations of HA in a Matrigel carrier into murine mammary glands. The X-ray attenuation of HA-Matrigel compositions labeled by BP-Au NPs was increased by up to 289 HU compared to unlabeled compositions for HA concentrations ranging from 0.5 to 25 mg/mL, which included an HA concentration (0.5 mg/mL) that was otherwise undetectable by micro-computed tomography. Cumulatively, both models demonstrated the ability of BP-Au NPs to enhance contrast for radiographic detection of microcalcifications, including at a clinically-relevant imaging resolution. Therefore, BP-Au NPs may have potential to improve clinical detection of breast microcalcifications by mammography.


Assuntos
Calcinose/diagnóstico , Meios de Contraste , Difosfonatos/química , Ouro/química , Glândulas Mamárias Animais/patologia , Mamografia/métodos , Nanopartículas Metálicas , Animais , Feminino , Camundongos , Imagens de Fantasmas
17.
ACS Nano ; 8(7): 7486-96, 2014 Jul 22.
Artigo em Inglês | MEDLINE | ID: mdl-24992365

RESUMO

Microcalcifications are deposits of hydroxyapatite (HA) mineral within breast tissue and the most common abnormality detected by mammography when screening for breast cancer due to exhibiting greater X-ray attenuation than the surrounding tissue. However, the detection of microcalcifications is limited by the sensitivity and specificity of mammography. Therefore, the objective of this study was to investigate in vivo targeted delivery of bisphosphonate-functionalized gold nanoparticles (BP-Au NPs) for contrast-enhanced detection of microcalcifications using computed tomography (CT). A murine model was developed for precise, a priori control over the level of microcalcification burden by injecting varying concentrations of HA crystals in a Matrigel carrier into mammary glands. The measured X-ray attenuation of microcalcifications containing varying HA concentrations demonstrated that the model was reproducible and able to recapitulate varying levels of microcalcification burden, including levels undetectable by CT in the absence of contrast enhancement. After intramammary delivery, BP-Au NPs provided enhanced contrast for the detection of microcalcifications that were otherwise below the CT detection limit. BP-Au NPs targeted microcalcifications due to specific binding to HA crystal surfaces, resulting in contrast between the HA microcalcification site and surrounding tissue which was visibly apparent (∼30-135 HU) within 2 days after delivery. Therefore, targeted BP-Au NPs enabled improved sensitivity and specificity for the detection of microcalcifications.


Assuntos
Calcinose/diagnóstico por imagem , Meios de Contraste/química , Ouro/química , Mamografia/métodos , Nanopartículas Metálicas , Animais , Neoplasias da Mama/diagnóstico por imagem , Meios de Contraste/farmacocinética , Difosfonatos/química , Modelos Animais de Doenças , Feminino , Ouro/farmacocinética , Imageamento Tridimensional , Camundongos , Distribuição Tecidual , Tomografia Computadorizada por Raios X
18.
PLoS One ; 8(5): e65105, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23717689

RESUMO

Rho GTPases mediate stromal-epithelial interactions that are important for mammary epithelial cell (MEC) morphogenesis. Increased extracellular matrix (ECM) deposition and reorganization affect MEC morphogenesis in a Rho GTPase-dependent manner. Although the effects of altered ECM on MEC morphogenesis have been described, how MECs regulate stromal deposition is not well understood. Previously, we showed that p190B RhoGAP overexpression disrupts mammary gland morphogenesis by inducing hyperbranching in association with stromal alterations. We therefore hypothesized that MEC overexpression of p190B regulates paracrine interactions to impact fibroblast activation. Using a combination of in vivo morphometric and immunohistochemical analyses and primary cell culture assays, we found that p190B overexpression in MECs activates fibroblasts leading to increased collagen, fibronectin, and laminin production and elevated expression of the collagen crosslinking enzyme lysyl oxidase. Phosphorylation of the TGF-ß effector SMAD2 and expression of the TGF-ß target gene αSma were increased in p190B-associated fibroblasts, suggesting that elevated TGF-ß signaling promoted fibroblast activation. Mechanical tension and TGF-ß cooperate to activate fibroblasts. Interestingly, active TGF-ß was elevated in conditioned medium from p190B overexpressing MECs compared to control MECs, and p190B overexpressing MECs exhibited increased contractility in a collagen gel contraction assay. These data suggest that paracrine signaling from the p190B overexpressing MECs may activate TGF-ß signaling in adjacent fibroblasts. In support of this, transfer of conditioned medium from p190B overexpressing MECs onto wildtype fibroblasts or co-culture of p190B overexpressing MECs with wildtype fibroblasts increased SMAD2 phosphorylation and mRNA expression of ECM genes in the fibroblasts when compared to fibroblasts treated with control CM or co-cultured with control MECs. The increased ECM gene expression and SMAD2 phosphorylation were blocked by treatment with a TGF-ß receptor inhibitor. Taken together, these data suggest that p190B overexpression in the mammary epithelium induces fibroblast activation via elevated TGF-ß paracrine signaling.


Assuntos
Fibroblastos/metabolismo , Proteínas Ativadoras de GTPase/metabolismo , Glândulas Mamárias Animais/metabolismo , Proteínas Repressoras/metabolismo , Fator de Crescimento Transformador beta/fisiologia , Animais , Western Blotting , Técnicas de Cocultura , Meios de Cultivo Condicionados , Epitélio/metabolismo , Matriz Extracelular/metabolismo , Feminino , Camundongos , Reação em Cadeia da Polimerase em Tempo Real
19.
Cancers (Basel) ; 4(2): 475-89, 2012 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-22582143

RESUMO

Rho GTPases are overexpressed and hyperactivated in many cancers, including breast cancer. Rho proteins, as well as their regulators and effectors, have been implicated in mitosis, and their altered expression promotes mitotic defects and aneuploidy. Previously, we demonstrated that p190B Rho GTPase activating protein (RhoGAP) deficiency inhibits ErbB2-induced mammary tumor formation in mice. Here we describe a novel role for p190B as a regulator of mitosis. We found that p190B localized to centrosomes during interphase and mitosis, and that it is differentially phosphorylated during mitosis. Knockdown of p190B expression in MCF-7 and Hela cells increased the incidence of aberrant microtubule-kinetochore attachments at metaphase, lagging chromosomes at anaphase, and micronucleation, all of which are indicative of aneuploidy. Cell cycle analysis of p190B deficient MCF-7 cells revealed a significant increase in apoptotic cells with a concomitant decrease in cells in G1 and S phase, suggesting that p190B deficient cells die at the G1 to S transition. Chemical inhibition of the Rac GTPase during mitosis reduced the incidence of lagging chromosomes in p190B knockdown cells to levels detected in control cells, suggesting that aberrant Rac activity in the absence of p190B promotes chromosome segregation defects. Taken together, these data suggest that p190B regulates chromosome segregation and apoptosis in cancer cells. We propose that disruption of mitosis may be one mechanism by which p190B deficiency inhibits tumorigenesis.

20.
Small GTPases ; 2(5): 247-258, 2011 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-22292127

RESUMO

The Rho GTPase Cdc42 is overexpressed and hyperactivated in breast cancer, and several studies have described mechanisms by which it may promote tumor formation and progression. However, little is known about the role of Cdc42 during normal mammary epithelial cell (MEC) morphogenesis. Here we aimed to define the precise role for Cdc42 during primary mammary acinus formation in vitro. For these studies, MECs were isolated from Cdc42fl/fl conditional knockout mice, transduced with Adeno-cre-GFP virus to delete Cdc42 or Adeno-GFP control virus, and effects on morphogenesis were investigated using a three-dimensional (3D) culture assay. Interestingly, markedly fewer mammary acini developed in Cdc42 deficient cultures, and the acini that formed were significantly smaller and disorganized. Cellular proliferation and survival were reduced in the Cdc42 deficient acini. However, control and knockout MECs cultured as monolayers displayed similar cell cycle profiles, suggesting that Cdc42 is important for MEC proliferation in the context of 3D polarity. Overexpression of cyclin D1, which promotes cell cycle progression downstream of Cdc42, failed to rescue the defect in acinus size. Furthermore, lumen formation and apical-basal polarity were disrupted, and mitotic spindle orientation and Cdc42/aPKC polarity complex defects likely contributed to these phenotypes. Studies using dominant negative Cdc42 and siRNa to knockdown Cdc42 in MDcK and Caco-2 cell lines undergoing cystogenesis in 3D cultures revealed critical roles for Cdc42 in spindle orientation, polarity and lumen formation. Our studies, using complete knockout in primary epithelial cells, demonstrate that Cdc42 is not only an important regulator of polarity and lumen formation; it is also essential for proliferation and survival, which are key cellular processes that drive MEC morphogenesis in vitro and in vivo.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA