Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
Biochimie ; 124: 53-64, 2016 May.
Article in English | MEDLINE | ID: mdl-26844637

ABSTRACT

Development of multidrug resistance (MDR) remains a significant problem in cancer chemotherapy and underscores the importance of using chemosensitizers. Well known MDR mechanisms include: (i) upregulation of drug-efflux; (ii) increased signaling via AKT; and (iii) decreased apoptosis. Therefore, chemosensitizers should target multiple resistance mechanisms. We investigated the efficacy of nelfinavir (NFV), a clinically approved anti-HIV drug, in increasing doxorubicin (DOX) toxicity in a MDR breast cancer cell line, MCF-7/Dox. As compared to parental MCF-7 cells, the MCF-7/Dox were 15-20 fold more resistant to DOX-induced cytotoxicity at 48 h post-exposure (DOX IC50 = 1.8 µM vs. 32.4 µM). Coexposures to NFV could significantly (p < 0.05) decrease DOX-IC50 in MCF-7/Dox cells. Multiple exposures to physiologic concentrations of NFV (2.25 µM or 6.75 µM) decreased DOX-IC50 by 21-fold and 50-fold, respectively. Interestingly, although single exposure to NFV transiently induced P-glycoprotein (P-gp) levels, multiple treatments with NFV inhibited both P-gp expression and efflux function, which increased intracellular DOX concentrations. Single exposure to NFV augmented the markers of cell-survival (AKT) and autophagy (LC3-II), whereas multiple exposures enabled suppression of both total AKT (t-AKT) and insulin like growth factor-1 (IGF-1)-induced phosphorylated AKT (p-AKT) levels. Multiple exposures to NFV also resulted in increased unfolded protein response (UPR) transducers, e.g. Grp78, p-PERK, p-eIF2α, and ATF-4; and endoplasmic reticulum (ER) stress induced death sensors, e.g. CHOP & TRIB-3. Multiple exposures to NFV also abrogated the mitogenic effects of IGF-1. In mice carrying MCF-7/Dox tumor xenografts, intraperitoneal (i.p.) injection of NFV (20 mg/kg/day) and DOX (2 mg/kg/twice/wk) decreased tumor growth more significantly (p < 0.01) than either agent alone. Immunohistochemical (IHC) analysis revealed decreased p-AKT and Ki-67 levels. Thus, NFV overcomes MDR in breast cancer cells and should be tested as an adjunct to chemotherapy.


Subject(s)
Breast Neoplasms , Doxorubicin/pharmacology , Drug Resistance, Multiple/drug effects , Drug Resistance, Neoplasm/drug effects , Nelfinavir/pharmacology , Animals , Breast Neoplasms/drug therapy , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Endoplasmic Reticulum Chaperone BiP , Female , Humans , MCF-7 Cells , Mice , Mice, Inbred BALB C , Mice, Nude , Xenograft Model Antitumor Assays
2.
Adv Healthc Mater ; 3(5): 703-13, 2014 May.
Article in English | MEDLINE | ID: mdl-24132933

ABSTRACT

Biodegradable poly(lactic-co-glycolic acid) (PLGA) porous films are developed to support mammary cell growth and function. Such porous polymer matrices of PLGA are generated using the easily implemented water-templating "breath-figure" technique that allows water droplets to penetrate the nascent polymer films to create a rough porous polymer film. Such breath figure-based micropatterned porous films show higher epithelial differentiation and growth than the corresponding flat 2D films, and represent the first instance of using them for tissue culture. Specifically, the breath figure morphology supports robust acinar growth with almost double the number of lobular-alveolar units compared to the 2D cultures. Gene profile analysis indicates that the cells grown on porous polymer films show enhanced expressions of mammary differentiation genes (GATA3, EMA, and INTEGB4) but lower the expression of mesenchymal gene (CALLA). Hormonal stimulation of these cultures dramatically increases expression of progenitor marker gene Notch1. Importantly, cells grown on porous PLGA films exhibit an enhanced resistance to doxorubicin treatment in comparison to 2D cultures. Breath-figure PLGA films show promise in mimicking in vivo mammary functions and can potentially be used to screen chemotherapeutic drugs. The simplicity and ease of fabrication of these polymer films is especially appealing to the development of effective biomaterials to support cell culture and differentiation.


Subject(s)
Antineoplastic Agents/pharmacology , Biocompatible Materials/chemistry , Lactic Acid/chemistry , Mammary Glands, Animal/cytology , Mammary Glands, Animal/growth & development , Models, Biological , Polyglycolic Acid/chemistry , Animals , Cell Culture Techniques , Cell Differentiation/physiology , Cell Proliferation/drug effects , Female , Humans , MCF-7 Cells , Morphogenesis/physiology , Polylactic Acid-Polyglycolic Acid Copolymer , Porosity
3.
Cancer Biol Ther ; 13(10): 831-5, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22785203

ABSTRACT

Biomarkers are biological agents used as indicators of biological states. In clinical applications, biomarkers reflect the presence, severity, or progression of disease states. They may also predict risk or responsiveness of a disease to a given treatment. There has been increasingly intense research interest in biomarkers, yet their translation into routine clinical use is lagging. To stimulate communication and cross-fertilization, the 2nd World Congress on Biomarkers & Clinical Research was held in Baltimore, MD, USA in 2011. The symposium covered a broad range of basic and applied biomarker research with the intent to facilitate bench-to-bedside developments. Sessions discussed DNA-based, proteomic, and blood-borne markers. The presentations covered biomarkers for cancer, other various diseases, and toxicological agents. Other topics included biomarker data assimilation, validation, standardization and quality control, as well as molecular imaging and informatics. New high-throughput assays, model systems and emerging technologies give reasons to hope for further rapid progress in the field.


Subject(s)
Biomarkers , Biomedical Research , Humans
4.
J Pineal Res ; 53(3): 307-18, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22582905

ABSTRACT

To determine whether melatonin, via its MT(1) G protein-coupled receptor, impacts mouse mammary gland development, we generated a mouse mammary tumor virus (MMTV)-MT1-Flag-mammary gland over-expressing (MT1-mOE) transgenic mouse. Increased expression of the MT(1) -Flag transgene was observed in the mammary glands of pubescent MT1-mOE transgenic female mice, with further significant increases during pregnancy and lactation. Mammary gland whole mounts from MT1-mOE mice showed significant reductions in ductal growth, ductal branching, and terminal end bud formation. Elevated MT(1) receptor expression in pregnant and lactating female MT1-mOE mice was associated with reduced lobulo-alveolar development, inhibition of mammary epithelial cell proliferation, and significant reductions in body weights of suckling pups. Elevated MT(1) expression in pregnant and lactating MT1-mOE mice correlated with reduced mammary gland expression of Akt1, phospho-Stat5, Wnt4, estrogen receptor alpha, progesterone receptors A and B, and milk proteins ß-casein and whey acidic protein. Estrogen- and progesterone-stimulated mammary gland development was repressed by elevated MT(1) receptor expression and exogenous melatonin administration. These studies demonstrate that the MT(1) melatonin receptor and its ligand melatonin play an important regulatory role in mammary gland development and lactation in mice through both growth suppression and alteration of developmental paradigms.


Subject(s)
Mammary Glands, Animal/growth & development , Melatonin/pharmacology , Receptor, Melatonin, MT1/physiology , Animals , Estrogen Receptor alpha/biosynthesis , Estrogen Receptor alpha/genetics , Female , Lactation/physiology , Mammary Glands, Animal/drug effects , Mammary Tumor Virus, Mouse/genetics , Mice , Mice, Transgenic , Pregnancy , Proto-Oncogene Proteins c-akt/biosynthesis , Proto-Oncogene Proteins c-akt/genetics , Receptor, Melatonin, MT1/genetics , STAT5 Transcription Factor/biosynthesis , STAT5 Transcription Factor/genetics
5.
Exp Biol Med (Maywood) ; 236(2): 145-55, 2011 Feb.
Article in English | MEDLINE | ID: mdl-21321311

ABSTRACT

SOX9, a high mobility group (HMG) box transcription factor, is required for development, differentiation and lineage commitment. It is known to exert its effects through nuclear translocation, such as cell cycle changes in response to retinoic acid treatment in breast cancer cells. However, it is not known whether SOX9 has prognostic significance in human breast cancer. Over-expression and cytoplasmic sequestration of nuclear proteins are implicated in tumor progression. To determine whether SOX9 has any prognostic significance in human breast cancer, its expression and subcellular localization were analyzed in more than 200 human breast carcinomas (BCs). SOX9 mRNA expression data for human BCs were computed from microarray studies available in public databases and correlated with known poor prognostic parameters of BCs. SOX9 protein expression and its correlation with Ki-67 staining in human BCs were assessed using immunohistochemistry. Higher SOX9 mRNA levels were significantly associated with estrogen receptor negative (P ≤ 0.001) and higher grade (P ≤ 0.01) human breast tumors. Patients with higher SOX9 mRNA level had significantly shorter overall survival (P ≤ 0.0001). SOX9 protein, which is normally nuclear, was instead localized in the cytoplasm of 25-30% invasive ductal carcinomas (IDCs) and lymph node metastases. Its cytoplasmic accumulation significantly correlated with enhanced proliferation in breast tumors (Kendall's tau = 0.337 with a P value < 0.0001). Cytoplasmic SOX9 can serve as a valuable prognostic marker for IDCs and metastatic breast cancer. Its significant correlation with breast tumor cell proliferation implies that SOX9 directly contributes to the poor clinical outcomes associated with invasive breast cancer.


Subject(s)
Breast Neoplasms/diagnosis , Breast Neoplasms/pathology , Carcinoma, Ductal, Breast/diagnosis , Carcinoma, Ductal, Breast/pathology , Cytoplasm/enzymology , SOX9 Transcription Factor/metabolism , Biomarkers , Breast Neoplasms/secondary , Carcinoma, Ductal, Breast/secondary , Gene Expression Profiling , Humans , Immunohistochemistry , Ki-67 Antigen/analysis , Lymph Nodes/pathology , Microarray Analysis , Neoplasm Metastasis/diagnosis , Neoplasm Metastasis/pathology , Prognosis , Severity of Illness Index , Survival Analysis
6.
Cancer Biol Ther ; 11(1): 71-83, 2011 Jan 01.
Article in English | MEDLINE | ID: mdl-21084857

ABSTRACT

We have previously reported that although SOX9 is a transcription factor, it is often localized in the cytoplasm of some invasive and metastatic breast carcinomas. To determine whether cytoplasmic compartmentalization is a common mechanism utilized by cancer cells to proliferate indefinitely, SOX9 localization was examined at different stages of development in normal mouse mammary glands and in cancer cells. We show here that SOX9 expression is nuclear in ductal epithelial cells throughout mammary gland development and differentiation but is localized in the cytoplasm of some breast cancer cell lines (BCCLs). Furthermore, cytoplasmic localization of SOX9 is associated with abrogation of the growth arrest response of breast cancer cells and results from dysregulated HDAC activity rather than defects in its nuclear export. Immuno-precipitation and immunoblot studies revealed that inhibiting HDAC activity with Trichostatin A can rescue this defect by up-regulating acetylated SOX9 and p21 expression that results in increased cell death. Our data suggests nuclear SOX9 expression parallels development and differentiation but cytoplasmic SOX9 expression is associated with abrogation of growth arrest response of breast cancer cells. Such expressions may be a common mechanism utilized by some transformed breast cancer cells to proliferate indefinitely.


Subject(s)
Breast Neoplasms/drug therapy , Breast Neoplasms/metabolism , Hydroxamic Acids/pharmacology , SOX9 Transcription Factor/metabolism , Animals , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Cell Growth Processes/physiology , Cell Line, Tumor , Cell Nucleus/metabolism , Cytoplasm/metabolism , Fatty Acids, Unsaturated/pharmacology , Female , Gene Expression , Humans , Immunohistochemistry , Mammary Glands, Animal/growth & development , Mammary Glands, Animal/metabolism , Mice , Pregnancy , SOX9 Transcription Factor/biosynthesis , SOX9 Transcription Factor/genetics , Transfection
7.
Biopolymers ; 94(5): 617-25, 2010.
Article in English | MEDLINE | ID: mdl-20564026

ABSTRACT

Cycloviolacin O2 (CyO2), a cyclotide from Viola odorata (Violaceae) has antitumor effects and causes cell death by membrane permeabilization. In the breast cancer line, MCF-7 and its drug resistant subline MCF-7/ADR, the cytotoxic effects of CyO2 (0.2-10 microM) were monitored in the presence and absence of doxorubicin (0.1-5 microM) using cell proliferation assays to establish its chemosensitizing abilities. SYTOX Green assays were Sperformed to verify membrane permeabilization and showed cellular disruption correlates with cyclotide chemosensitization. Fluorescence microscopy studies demonstrated increased cellular internalization of doxorubicin in drug resistant cells when coexposed to CyO2. Interestingly, CyO2 did not produce significant membrane disruption in primary human brain endothelial cells, which suggested cyclotide specificity toward induced pore formation in highly proliferating tumor cells. Furthermore, three novel cyclotides (psyle A, C and E) from Psychotria leptothyrsa (Rubiaceae) were also monitored for cytotoxic activity. The cyclotides displayed potent cytotoxicity (IC50 = 0.64->10 microM), and coexposure to cyclotides significantly enhanced doxorubicin induced toxicity (IC50 = 0.39-0.76 microM). This study documents several cyclotides with robust cytotoxicity that may be promising chemosensitizing agents against drug resistant breast cancer.


Subject(s)
Antineoplastic Agents/pharmacology , Cell Line, Tumor/drug effects , Cyclotides/pharmacology , Viola/chemistry , Antineoplastic Agents/chemistry , Cyclotides/chemistry , Drug Resistance, Neoplasm , Drug Screening Assays, Antitumor , Humans , Inhibitory Concentration 50 , Plant Proteins/chemistry , Plant Proteins/pharmacology
8.
Biol Pharm Bull ; 32(12): 2002-9, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19952419

ABSTRACT

The ATP binding cassette (ABC)-transporters are energy dependent efflux pumps which regulate the pharmacokinetics of both anti-cancer chemotherapeutic agents, e.g. taxol, and of human immunodeficiency virus-1 (HIV-1) protease inhibitors (HPIs), e.g. saquinavir. Increased expression of several ABC-transporters, especially P-glycoprotein (P-gp) and multidrug resistance protein 2 (MRP2), are observed in multidrug resistant (MDR) tumor cells and on HIV-1 infected lymphocytes. In addition, due to their apical expression on vascular endothelial barriers, both P-gp and MRP2 are of crucial importance towards dictating drug access into sequestered tissues. However, although a number of P-gp inhibitors are currently in clinical trials, possible inhibitors of MRP2 are not being thoroughly investigated. The experimental leukotriene receptor antagonist (LTRA), MK-571 is known to be a potent inhibitor of MRP transporters. Using the MRP2 over-expressing Madin-Darby canine kidney cell line, MDCKII-MRP2, we evaluated whether the clinically approved LTRAs, e.g. montelukast (Singulair) and zafirlukast (Accolate), can similarly suppress MRP2-mediated efflux. We compared the efficacy of increasing concentrations (20-100 microM) of MK-571, montelukast, and zafirlukast, in suppressing the efflux of calcein-AM, a fluorescent MRP substrate, and the radiolabeled [(3)H-] drugs, taxol and saquinavir. Montelukast was the most potent inhibitor (p<0.01) of MRP2-mediated efflux of all three substrates. Montelukast also increased (p<0.01) the duration of intracellular retention of both taxol and saquinavir. More than 50% of the drugs were retained in cells even after 90 min post removal of montelukast from the medium. Our findings implicate that montelukast, a relatively safe anti-asthmatic agent, may be used as an adjunct therapy to suppress the efflux of taxol and saquinavir from MRP2 overexpressing cells.


Subject(s)
Acetates/pharmacology , Antineoplastic Agents, Phytogenic/pharmacokinetics , HIV Protease Inhibitors/pharmacokinetics , Leukotriene Antagonists/pharmacology , Multidrug Resistance-Associated Proteins/metabolism , Paclitaxel/pharmacokinetics , Quinolines/pharmacology , Saquinavir/pharmacokinetics , Animals , Anti-Asthmatic Agents/pharmacology , Antineoplastic Agents, Phytogenic/therapeutic use , Biological Transport/drug effects , Cell Line , Chemotherapy, Adjuvant , Cyclopropanes , Dogs , Drug Resistance, Neoplasm/drug effects , Drug Resistance, Viral/drug effects , Fluoresceins/metabolism , HIV Infections/drug therapy , HIV Infections/metabolism , HIV Protease Inhibitors/therapeutic use , Indoles , Neoplasms/drug therapy , Neoplasms/metabolism , Paclitaxel/therapeutic use , Phenylcarbamates , Propionates/pharmacology , Saquinavir/therapeutic use , Sulfides , Sulfonamides , Time Factors , Tosyl Compounds/pharmacology
9.
Exp Biol Med (Maywood) ; 234(4): 372-86, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19176870

ABSTRACT

Overexpression of insulin-like growth factor-I receptor (IGF-IR) is seen in a multitude of human thyroid cancers and correlates with poor prognosis. However, recent studies suggest that low phospho-IGF-IR (pIGF-IR) expression rather than its overexpression may be an indicator of poorly differentiated disease. No previous study has evaluated the expression of pIGF-IR to determine if activation or loss of expression of this receptor is associated with thyroid tumor progression. Accordingly, a quantitative immunohistochemical (IHC) method was used to evaluate the clinico-pathological significance of pIGF-IR expression in archival samples of human thyroid carcinomas. Quantitative analysis of pIGF-IR levels revealed a significant difference in the median index of pIGF-IR between different histological subtypes of thyroid cancer (P < 0.001). Specifically, the median pIGF-IR index of differentiated thyroid cancers was significantly higher than the median index of other poorly differentiated thyroid cancer (P < 0.001). This was further confirmed in individual tumor sections of thyroid carcinoma where anaplastic and differentiated components co-existed. No significant difference was noted in the pIGF-IR index of tumors grouped by size or stage but a trend towards lower mean pIGF-IR index was noted in older patients. Our data indicates that pIGF-IR is upregulated in a majority of follicular thyroid carcinomas, suggesting it may be a potential target for therapy for patients with this disease. In addition, since low pIGF-IR expression was found to correlate with aggressive human thyroid carcinoma, it also suggests that IGF-IR may not be needed for progression of anaplastic thyroid carcinoma possibly because other cell signaling pathways are activated, obviating the need for IGF-IR signaling. However, more mechanistic studies would be necessary to substantiate the possibility that pIGF-IR may be important for differentiation of thyroid tissues and is lost with disease progression.


Subject(s)
Carcinoma/metabolism , Receptor, IGF Type 1/metabolism , Thyroid Neoplasms/metabolism , Adult , Age Factors , Carcinoma/genetics , Carcinoma/pathology , Gene Expression , Humans , Middle Aged , Phosphorylation , Receptor, IGF Type 1/genetics , Receptor, IGF Type 1/physiology , Thyroid Neoplasms/genetics , Thyroid Neoplasms/pathology , Up-Regulation
10.
Mol Cancer Res ; 6(1): 119-26, 2008 Jan.
Article in English | MEDLINE | ID: mdl-18234967

ABSTRACT

K7M2 mouse osteosarcoma cells form lytic tumors and are deficient in osterix (Osx), a zinc finger-containing transcription factor required for osteoblast differentiation and bone formation. Our previous studies showed that replacement of Osx suppresses lytic bone destruction. Cytokines, including interleukin (IL)-1alpha, IL-6, IL-11, and prostaglandin E2, have been shown to stimulate osteoclast activity. We showed that IL-1alpha production by K7M2 cells was significantly suppressed following Osx transfection through a transcription-mediated mechanism. Osx had no effect on IL-6, IL-11, or prostaglandin E2. Site-directed mutagenesis and chromatin immunoprecipitation indicated that Osx down-regulated IL-1alpha through an Sp1-binding site on the IL-1alpha promoter. Inhibiting Osx by small interfering RNA in two cell lines (Dunn and DLM8) that expressed high levels of Osx led to enhanced IL-1alpha promoter activity and protein production and altered the phenotype from blastic to lytic. These data suggest that Osx down-regulates IL-1alpha expression in mouse osteosarcoma cells via transcriptional repression of IL-1alpha and this may in turn affect the lytic activity of the tumor cells.


Subject(s)
Bone Neoplasms/genetics , Down-Regulation , Gene Expression Regulation, Neoplastic , Interleukin-1alpha/genetics , Osteosarcoma/genetics , Transcription Factors/metabolism , Animals , Binding Sites , Cell Line, Tumor , Male , Mice , Mice, Inbred BALB C , Mutation/genetics , Osteoprotegerin/genetics , Osteosarcoma/pathology , Phenotype , Promoter Regions, Genetic/genetics , RNA, Small Interfering , Sp1 Transcription Factor/metabolism , Sp7 Transcription Factor , Transcription Factors/genetics , Transcription, Genetic
11.
Dev Biol ; 309(1): 137-49, 2007 Sep 01.
Article in English | MEDLINE | ID: mdl-17662267

ABSTRACT

P190-B RhoGAP (p190-B, also known as ARHGAP5) has been shown to play an essential role in invasion of the terminal end buds (TEBs) into the surrounding fat pad during mammary gland ductal morphogenesis. Here we report that embryos with a homozygous p190-B gene deletion exhibit major defects in embryonic mammary bud development. Overall, p190-B-deficient buds were smaller in size, contained fewer cells, and displayed characteristics of impaired mesenchymal proliferation and differentiation. Consistent with the reported effects of p190-B deletion on IGF-1R signaling, IGF-1R-deficient embryos also displayed a similar small mammary bud phenotype. However, unlike the p190-B-deficient embryos, the IGF-1R-deficient embryos exhibited decreased epithelial proliferation and did not display mesenchymal defects. Because both IGF and p190-B signaling affect IRS-1/2, we examined IRS-1/2 double knockout embryonic mammary buds. These embryos displayed major defects similar to the p190-B-deficient embryos including smaller bud size. Importantly, like the p190-B-deficient buds, proliferation of the IRS-1/2-deficient mesenchyme was impaired. These results indicate that IGF signaling through p190-B and IRS proteins is critical for mammary bud formation and ensuing epithelial-mesenchymal interactions necessary to sustain mammary bud morphogenesis.


Subject(s)
GTPase-Activating Proteins/metabolism , Mammary Glands, Animal/embryology , Receptor, IGF Type 1/metabolism , Signal Transduction , Animals , Epithelium/embryology , Epithelium/physiology , Female , GTPase-Activating Proteins/genetics , Insulin Receptor Substrate Proteins , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Male , Mammary Glands, Animal/metabolism , Mesoderm/physiology , Mice , Mice, Knockout , Phosphoproteins/genetics , Phosphoproteins/metabolism , Receptor, IGF Type 1/genetics
12.
Clin Cancer Res ; 12(15): 4755-65, 2006 Aug 01.
Article in English | MEDLINE | ID: mdl-16899627

ABSTRACT

PURPOSE: The insulin-like growth factor-I receptor (IGF-IR) and its ligands have been implicated in the pathogenesis and progression of various cancers, including those arising in the thyroid gland. We therefore evaluated whether the IGF-IR could serve as a potential target for therapy of anaplastic thyroid carcinoma (ATC). EXPERIMENTAL DESIGN: The expression and activation of the IGF-IR and some of its downstream signaling pathway components were evaluated in both human thyroid cancer specimens and thyroid cancer cell lines. The therapeutic potential of a humanized monoclonal antibody (A12) directed against IGF-IR was assessed in vitro and in vivo in an orthotopic model of ATC. Tumor volume and overall survival time were analyzed to evaluate the efficacy of A12 in vivo. RESULTS: IGF-IR was overexpressed in 94% of the thyroid cancers. Blockade of IGF-IR with A12 was effective in attenuating IGF-IR signaling both in vitro and in vivo. However, the inhibitory effects of A12 on cell proliferation were cell line dependent, as those ATC cell lines that had detectable levels of pIGF-IR were more sensitive to A12 treatment. A12 was equally effective in vivo, where it brought approximately 57% (P = 0.041) inhibition in tumor volume. The concomitant use of A12 and irinotecan produced additive effects and resulted in a 93% (P < 0.001) reduction in tumor volume. Blocking IGF-IR blocked Akt phosphorylation and decreased proliferation and microvessel density but increased apoptosis within the tumor xenografts. Our results also highlighted a previously undefined IGF-IR-mediated antiangiogenic effect on tumor-associated endothelium in thyroid cancers. CONCLUSION: Blocking the IGF-IR with A12 seems to be a potential avenue for treating patients with ATC by its direct antitumor effects and its effects on the tumor vasculature.


Subject(s)
Antibodies, Monoclonal/pharmacology , Antineoplastic Agents/pharmacology , Carcinoma/drug therapy , Receptor, IGF Type 1/antagonists & inhibitors , Thyroid Neoplasms/drug therapy , Animals , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/therapeutic use , Antigen-Antibody Reactions , Antineoplastic Agents/immunology , Antineoplastic Agents/therapeutic use , Apoptosis/drug effects , Carcinoma/pathology , Cell Line, Tumor , Cell Proliferation/drug effects , Disease Models, Animal , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , Humans , Insulin-Like Growth Factor I/pharmacology , Insulin-Like Growth Factor II/pharmacology , Male , Methylation , Mice , Mice, Nude , Neovascularization, Pathologic/drug therapy , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Receptor, IGF Type 1/biosynthesis , Receptor, IGF Type 1/immunology , Signal Transduction/drug effects , Signal Transduction/immunology , Structure-Activity Relationship , Thyroid Neoplasms/pathology , Transplantation, Heterologous , Xenograft Model Antitumor Assays
13.
Mol Endocrinol ; 17(6): 1054-65, 2003 Jun.
Article in English | MEDLINE | ID: mdl-12637587

ABSTRACT

Previous studies from our laboratory have demonstrated that p190-B RhoGAP (p190-B) is differentially expressed in the Cap cells of terminal end buds (TEBs) and poorly differentiated rodent mammary tumors. Based on these observations we hypothesized that p190-B might play an essential role in invasion of the TEBs into the surrounding fat pad during ductal morphogenesis. To test this hypothesis, mammary development was studied in p190-B-deficient mice. A haploinsufficiency phenotype was observed in p190-B heterozygous mice as indicated by decreased number and rate of ductal outgrowth(s) at 3, 4, and 5 wk of age when compared with their wild-type littermates. This appeared to result from decreased proliferation in the Cap cells of the TEBs, a phenotype remarkably similar to that observed previously in IGF-I receptor null mammary epithelium. Furthermore, decreased expression of insulin receptor substrates 1 and 2 were observed in TEBs of p190-B heterozygous mice. These findings are consistent with decreased IGF signaling observed previously in p190-B-/- mouse embryo fibroblasts. To further assess if this defect was cell autonomous or due to systemic endocrine effects, the mammary anlagen from p190-B+/+, p190-B+/-, and p190-B-/- mice was rescued by transplantation into the cleared fat pad of recipient Rag1-/- mice. Surprisingly, as opposed to 75-80% outgrowths observed using wild-type donor epithelium, only 40% of the heterozygous and none of the p190-B-/- epithelial transplants displayed any outgrowths. Together, these results suggest that p190-B regulates ductal morphogenesis, at least in part, by modulating the IGF signaling axis.


Subject(s)
Guanine Nucleotide Exchange Factors/physiology , Mammary Glands, Animal/growth & development , Nuclear Proteins/physiology , Phosphoproteins/metabolism , Sexual Maturation/physiology , Somatomedins/metabolism , Analysis of Variance , Animals , Cell Division/physiology , DNA-Binding Proteins , Female , GTPase-Activating Proteins , Guanine Nucleotide Exchange Factors/genetics , Heterozygote , Insulin Receptor Substrate Proteins , Intracellular Signaling Peptides and Proteins , Mammary Glands, Animal/cytology , Mammary Glands, Animal/metabolism , Mice , Morphogenesis , Nuclear Proteins/genetics , Repressor Proteins , Sexual Maturation/genetics , Signal Transduction
SELECTION OF CITATIONS
SEARCH DETAIL
...