Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
Metabolites ; 12(1)2022 Jan 10.
Article in English | MEDLINE | ID: mdl-35050177

ABSTRACT

The aim of this study was to investigate whether CD74 levels in atherosclerotic lesions are associated with inflammation, apoptosis, plaque severity, and clinical symptoms among patients with carotid atherosclerosis. We further studied whether CD74 expression is associated with apoptosis in macrophages induced by 7ketocholesterol (7keto). Sixty-one carotid samples (39 males and 22 females) were immunostained with macrophages, smooth muscle cells, CD74, ferritin, TUNEL (Terminal deoxynucleotidyl transferase dUTP nick end labeling), and thrombin receptors. Double immunocytochemistry of CD74 and caspase 3 or CD74 and Annexin V was performed on THP-1 macrophages exposed to 7keto. In human carotid plaques, CD74 expression is lesion-dependently increased and is associated with necrotic core formation and plaque rupture, clinical symptoms, macrophage apoptosis, ferritin, and thrombin receptors. CD74 levels were inversely correlated to high-density lipoproteins and statin treatment, and positively correlated to triglycerides. In THP-1 macrophages, 7keto induced a significant increase in levels of CD74, ferritin, and apoptotic cell death. This study suggests that CD74 in apoptotic macrophages is linked to inflammation and thrombosis in progression of human atherosclerotic plaques, lipid metabolism, and clinical manifestation in atherosclerosis. Surface CD74 in apoptotic macrophages and ferritin production induced by oxidized lipids may contribute to inflammation and plaque vulnerability in atherosclerosis.

2.
Breast Cancer Res ; 12(6): R96, 2010.
Article in English | MEDLINE | ID: mdl-21080941

ABSTRACT

INTRODUCTION: The human epidermal growth factor receptor 2 (HER2) is a validated therapeutic target in breast cancer. Heterodimerization of HER2 with other HER family members results in enhanced tyrosine phosphorylation and activation of signal transduction pathways. HER2 overexpression increases the translation of fatty acid synthase (FASN), and FASN overexpression markedly increases HER2 signaling, which results in enhanced cell growth. However, the molecular mechanism and regulation of HER2 and FASN interaction are not well defined. Lapatinib is a small-molecule tyrosine kinase inhibitor that blocks phosphorylation of the epidermal growth factor receptor and HER2 in breast cancer cells, resulting in apoptosis. We hypothesized that FASN is directly phosphorylated by HER2, resulting in enhanced signaling and tumor progression in breast cancer cells. METHODS: Using mass spectrometry, we identified FASN as one of the proteins that is dephosphorylated by lapatinib in SKBR3 breast cancer cells. Immunofluorescence, immunoprecipitation, Western blotting, a kinase assay, a FASN enzymatic activity assay, an invasion assay, a cell viability assay and zymography were used to determine the role of FASN phosphorylation in invasion of SKBR3 and BT474 cells. The FASN inhibitor C75 and small interfering RNA were used to downregulate FASN expression and/or activity. RESULTS: Our data demonstrated that FASN is phosphorylated when it is in complex with HER2. FASN phosphorylation was induced by heregulin in HER2-overexpressing SKBR3 and BT474 breast cancer cells. Heregulin-induced FASN phosphorylation resulted in increased FASN enzymatic activity, which was inhibited by lapatinib. The FASN inhibitor C75 suppressed FASN activity by directly inhibiting HER2 and FASN phosphorylation. Blocking FASN phosphorylation and activity by lapatinib or C75 suppressed the activity of matrix metallopeptidase 9 and inhibited invasion of SKBR3 and BT474 cells. CONCLUSIONS: FASN phosphorylation by HER2 plays an important role in breast cancer progression and may be a novel therapeutic target in HER2-overexpressing breast cancer cells.


Subject(s)
4-Butyrolactone/analogs & derivatives , Antineoplastic Agents/pharmacology , Breast Neoplasms/metabolism , Fatty Acid Synthases/metabolism , Quinazolines/pharmacology , Receptor, ErbB-2/metabolism , 4-Butyrolactone/metabolism , 4-Butyrolactone/pharmacology , Apoptosis/drug effects , Blotting, Western , Breast Neoplasms/genetics , Cell Line, Tumor , Fatty Acid Synthases/antagonists & inhibitors , Female , Fluorescent Antibody Technique , Gene Expression , Humans , Lapatinib , Mass Spectrometry , Matrix Metalloproteinase 9/metabolism , Matrix Metalloproteinase Inhibitors , Neuregulin-1/metabolism , Phosphorylation/drug effects , Protein Kinase Inhibitors/pharmacology , Quinazolines/metabolism , RNA, Small Interfering , Receptor, ErbB-2/antagonists & inhibitors , Receptor, ErbB-2/genetics , Signal Transduction/drug effects
3.
Clin Cancer Res ; 16(6): 1802-11, 2010 Mar 15.
Article in English | MEDLINE | ID: mdl-20215547

ABSTRACT

PURPOSE: To determine the role of PEA-15 in breast cancer. EXPERIMENTAL DESIGN: A reverse-phase protein array was used to measure PEA-15 expression levels in 320 human breast cancers; these levels were correlated with clinical and tumor characteristics. PEA-15 was overexpressed by an adenovirus vector or by stably expressing PEA-15 in different breast cancer cell lines. The effects on breast cancer cell survival and on the downstream apoptotic signaling pathway were measured in terms of cell proliferation (trypan blue for cell viability, bromodeoxyuridine incorporation for DNA synthesis), anchorage-independent growth (soft agar colony formation), and apoptosis (fluorescence-activated cell sorter analysis). The preclinical efficacy of Ad.PEA-15 given intratumorally was evaluated in nude mice bearing tumors from s.c. implanted human MDA-MB-468 triple-negative breast cancer cells. RESULTS: In human breast cancers, low levels of PEA-15 expression correlated with high nuclear grade (P < 0.0001) and with negative hormone receptor status (P = 0.0004). Overexpression of PEA-15 in breast cancer cells resulted in growth inhibition, reduction in DNA synthesis, and onset of caspase-8-dependent apoptosis. In athymic nude mice bearing MDA-MB-468 xenografts, tumor volumes were significantly smaller in mice treated intratumorally with Ad.PEA-15 than in control mice (P < 0.0001). Tumors from mice treated with Ad.PEA-15 had increased levels of activated (phosphorylated) extracellular signal-regulated kinase and reduced levels of Ki-67 compared with tumors from nontreated or control-adenovirus-treated mice. CONCLUSION: PEA-15 has therapeutic potential in breast cancer. Further preclinical and clinical exploration of PEA-15 as a druggable target is warranted.


Subject(s)
Breast Neoplasms/metabolism , Breast Neoplasms/prevention & control , Carcinoma, Ductal, Breast/metabolism , Carcinoma, Ductal, Breast/prevention & control , Cytoplasm/enzymology , Extracellular Signal-Regulated MAP Kinases/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Phosphoproteins/metabolism , Xenograft Model Antitumor Assays , Adult , Aged , Aged, 80 and over , Animals , Apoptosis , Apoptosis Regulatory Proteins , Blotting, Western , Breast Neoplasms/pathology , Carcinoma, Ductal, Breast/pathology , Caspases/metabolism , Cell Adhesion , Cell Cycle , Cell Proliferation , Colony-Forming Units Assay , Female , Humans , Immunoenzyme Techniques , In Situ Nick-End Labeling , Mice , Mice, Inbred BALB C , Mice, Nude , Middle Aged , Protein Array Analysis , Signal Transduction , Tumor Cells, Cultured
4.
Nat Immunol ; 11(1): 55-62, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19898471

ABSTRACT

Autophagy is emerging as a crucial defense mechanism against bacteria, but the host intracellular sensors responsible for inducing autophagy in response to bacterial infection remain unknown. Here we demonstrated that the intracellular sensors Nod1 and Nod2 are critical for the autophagic response to invasive bacteria. By a mechanism independent of the adaptor RIP2 and transcription factor NF-kappaB, Nod1 and Nod2 recruited the autophagy protein ATG16L1 to the plasma membrane at the bacterial entry site. In cells homozygous for the Crohn's disease-associated NOD2 frameshift mutation, mutant Nod2 failed to recruit ATG16L1 to the plasma membrane and wrapping of invading bacteria by autophagosomes was impaired. Our results link bacterial sensing by Nod proteins to the induction of autophagy and provide a functional link between Nod2 and ATG16L1, which are encoded by two of the most important genes associated with Crohn's disease.


Subject(s)
Autophagy , Carrier Proteins/metabolism , Cell Membrane/metabolism , Nod1 Signaling Adaptor Protein/metabolism , Nod2 Signaling Adaptor Protein/metabolism , Animals , Autophagy-Related Proteins , Bacteria/metabolism , Carrier Proteins/genetics , Cell Line , Cell Membrane/microbiology , Cell Membrane/ultrastructure , Cells, Cultured , Female , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , HeLa Cells , Humans , Immunoblotting , Male , Mice , Mice, Inbred C57BL , Mice, Inbred Strains , Mice, Knockout , Microscopy, Confocal , Microscopy, Electron , Microscopy, Fluorescence , Mutation , Nod1 Signaling Adaptor Protein/genetics , Nod2 Signaling Adaptor Protein/genetics , Transfection
5.
Clin Cancer Res ; 14(23): 7909-16, 2008 Dec 01.
Article in English | MEDLINE | ID: mdl-19047121

ABSTRACT

PURPOSE: Preclinical data showed enhancement of breast cancer cell death when G3139 was combined with anthracyclines and taxanes. We evaluated the efficacy and safety of a Bcl-2 antisense oligonucleotide, G3139, in combination with doxorubicin (A) and docetaxel (T) in patients with locally advanced breast cancer (LABC). EXPERIMENTAL DESIGN: Following a brief phase I to determine the phase II dose, patients with locally advanced breast cancer received G3139 administered by continuous i.v. infusion for 5 to 7 days with bolus A (50 mg/m2) and T (75 mg/m2) administered on either day 3 or 6 of therapy with G3139. Cycles were repeated every 21 days x 6 in the neoadjuvant setting. Serial plasma samples were obtained for pharmacokinetic analysis. Tissue samples were obtained before and after therapy for pharmacodynamic analysis of Bcl-2 expression. RESULTS: Thirty patients (median age, 49 years; range, 24-71 years) received 160 cycles. During the phase I portion of the trial, the dose of G3139 was escalated from 3 to 7 mg/kg/d (i.v. for 5 days) in combination with AT. During the phase II portion of the trial, several doses and schedules of G3139 were evaluated. There were no pathologic complete responses. Pharmacodynamic studies showed limited Bcl-2 down-regulation in the primary tumors. CONCLUSIONS: G3139 in combination with doxorubicin and docetaxel is well tolerated. No pathologic complete response was seen and pharmacodynamic studies showed very little down-regulation of Bcl-2 in primary tumors, perhaps related to issues with insufficient drug delivery to the intact tumor.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Breast Neoplasms/drug therapy , Proto-Oncogene Proteins c-bcl-2/drug effects , Thionucleotides/administration & dosage , Adult , Aged , Antineoplastic Combined Chemotherapy Protocols/pharmacokinetics , Docetaxel , Doxorubicin/administration & dosage , Doxorubicin/adverse effects , Female , Humans , Middle Aged , Proto-Oncogene Proteins c-bcl-2/biosynthesis , Reverse Transcriptase Polymerase Chain Reaction , Taxoids/administration & dosage , Taxoids/adverse effects , Thionucleotides/adverse effects , Thionucleotides/pharmacokinetics
6.
Mol Cancer Ther ; 6(2): 667-74, 2007 Feb.
Article in English | MEDLINE | ID: mdl-17308062

ABSTRACT

The majority of breast cancer patients who achieve an initial therapeutic response to the HER2-targeted antibody trastuzumab will show disease progression within 1 year. Thus, the identification of novel agents that effectively inhibit survival of cancer cells that have progressed on trastuzumab is critical. In the current study, we show that the dual epidermal growth factor receptor (EGFR)/human EGFR-2 (HER2) kinase inhibitor lapatinib induces apoptosis in trastuzumab-resistant cells derived from the HER2-overexpressing SKBR3 breast cancer line. Lapatinib inhibited EGFR and HER2 signaling in resistant cells, blocking activation of downstream Akt, mitogen-activated protein kinase [corrected] Importantly, lapatinib also inhibited insulin-like growth factor I (IGF-I) signaling and growth-promoting effects in parental and resistant cells, and the cytotoxic effects of lapatinib were further enhanced by the IGF-I receptor-blocking antibody alphaIR3. As increased IGF-I receptor signaling has been implicated in trastuzumab resistance, our data strongly support further study of lapatinib as a potential therapeutic in breast cancers that have progressed on trastuzumab.


Subject(s)
Antibodies, Monoclonal/pharmacology , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Breast Neoplasms/drug therapy , Drug Resistance, Neoplasm , Insulin-Like Growth Factor I/metabolism , Quinazolines/pharmacology , Antibodies, Monoclonal, Humanized , Breast Neoplasms/pathology , Cell Cycle/drug effects , Cell Line, Tumor/drug effects , Cell Proliferation/drug effects , ErbB Receptors/antagonists & inhibitors , Female , Humans , Immunoblotting , Immunoprecipitation , Lapatinib , Receptor, ErbB-2/antagonists & inhibitors , Receptor, IGF Type 1/metabolism , Signal Transduction , Trastuzumab
7.
Mol Cancer Ther ; 5(6): 1593-601, 2006 Jun.
Article in English | MEDLINE | ID: mdl-16818519

ABSTRACT

The antiapoptotic protein Bcl-2 is overexpressed in a majority of breast cancers, and is associated with a diminished apoptotic response and resistance to various antitumor agents. Bcl-2 inhibition is currently being explored as a possible strategy for sensitizing breast cancer cells to standard chemotherapeutic agents. Antisense Bcl-2 oligonucleotides represent one method for blocking the antiapoptotic effects of Bcl-2. In this study, we show that antisense Bcl-2 efficiently blocks Bcl-2 expression, resulting in the apoptosis of breast cancer cells. Antisense Bcl-2-mediated cytotoxicity was associated with the induction of the B cell translocation gene 1 (BTG1). Importantly, knockdown of BTG1 reduced antisense Bcl-2-mediated cytotoxicity in breast cancer cells. Furthermore, BTG1 expression seems to be negatively regulated by Bcl-2, and exogenous expression of BTG1 induced apoptosis. These results suggest that BTG1 is a Bcl-2-regulated mediator of apoptosis in breast cancer cells, and that its induction contributes to antisense Bcl-2-mediated cytotoxic effects.


Subject(s)
Apoptosis , Breast Neoplasms/pathology , Neoplasm Proteins/metabolism , Oligonucleotides, Antisense/pharmacology , Proto-Oncogene Proteins c-bcl-2/metabolism , Breast Neoplasms/drug therapy , Breast Neoplasms/metabolism , Down-Regulation , Female , Gene Expression Profiling , Humans , Leukemia, Lymphocytic, Chronic, B-Cell/pathology , Neoplasm Proteins/antagonists & inhibitors , Neoplasm Proteins/genetics , Oligonucleotide Array Sequence Analysis , Proto-Oncogene Proteins c-bcl-2/antagonists & inhibitors , Proto-Oncogene Proteins c-bcl-2/genetics , Tumor Cells, Cultured
8.
Cancer Res ; 65(23): 11118-28, 2005 Dec 01.
Article in English | MEDLINE | ID: mdl-16322262

ABSTRACT

The majority of breast cancer patients who achieve an initial therapeutic response to the human epidermal growth factor receptor 2 (HER-2)-targeted antibody trastuzumab will show disease progression within 1 year. We previously reported the characterization of SKBR3-derived trastuzumab-resistant pools. In the current study, we show that HER-2 interacts with insulin-like growth factor-I receptor (IGF-IR) uniquely in these resistant cells and not in the parental trastuzumab-sensitive cells. The occurrence of cross talk between IGF-IR and HER-2 exclusively in resistant cells is evidenced by the IGF-I stimulation resulting in increased phosphorylation of HER-2 in resistant cells, but not in parental cells, and by the inhibition of IGF-IR tyrosine kinase activity leading to decreased HER-2 phosphorylation only in resistant cells. In addition, inhibition of IGF-IR tyrosine kinase activity by I-OMe-AG538 increased sensitivity of resistant cells to trastuzumab. HER-2/IGF-IR interaction was disrupted on exposure of resistant cells to the anti-IGF-IR antibody alpha-IR3 and, to a lesser extent, when exposed to the anti-HER-2 antibody pertuzumab. Heterodimer disruption by alpha-IR3 dramatically restored sensitivity to trastuzumab and resistant cells showed a slightly increased sensitivity to pertuzumab versus parental cells. Neither alpha-IR3 nor pertuzumab decreased HER-2 phosphorylation, suggesting that additional sources of phosphorylation other than IGF-IR exist when HER-2 and IGF-IR are not physically bound. Our data support a unique interaction between HER-2 and IGF-IR in trastuzumab-resistant cells such that cross talk occurs between IGF-IR and HER-2. These data suggest that the IGF-IR/HER-2 heterodimer contributes to trastuzumab resistance and justify the need for further studies examining this complex as a potential therapeutic target in breast cancers that have progressed while on trastuzumab.


Subject(s)
Antibodies, Monoclonal/pharmacology , Antineoplastic Agents/pharmacology , Breast Neoplasms/drug therapy , Breast Neoplasms/metabolism , Receptor, ErbB-2/metabolism , Receptor, IGF Type 1/metabolism , Amino Acid Sequence , Antibodies, Monoclonal, Humanized , Cell Line, Tumor , Dimerization , Drug Resistance, Neoplasm , Humans , Insulin-Like Growth Factor I/pharmacology , Molecular Sequence Data , Phosphorylation , Receptor Cross-Talk , Receptor, ErbB-2/immunology , Receptor, IGF Type 1/antagonists & inhibitors , Trastuzumab
9.
Cancer Res ; 65(18): 8406-13, 2005 Sep 15.
Article in English | MEDLINE | ID: mdl-16166319

ABSTRACT

We are currently conducting clinical trials of E1A gene therapy for patients with ovarian cancer. The adenovirus type 5 E1A gene suppresses growth of ovarian cancer cells that overexpress HER-2/neu (HER2) and growth of some--but not all--that express low HER2. In HER2-overexpressing cells, suppression by E1A is predominantly by down-regulation of HER2, but the mechanism in low HER2-expressing cells is not fully understood. The adenoviral E1B protein has sequential and functional homology to Bcl-2 and prolongs the viability of adenovirus host cells by inhibiting E1A-induced apoptosis. Bcl-2 is overexpressed in ovarian cancer and participates in chemoresistance; we hypothesized that Bcl-2 inhibits E1A-induced apoptosis leading to resistance to E1A gene therapy. E1A suppressed colony formation of ovarian cancer cells that express low levels of Bcl-2 and HER2 (OVCAR-3 and OVCA 433), but enhanced colony formation in low HER2-, high Bcl-2-expressing ovarian cancer cells (2774 and HEY). Treating 2774 or HEY cells with antisense oligonucleotide Bcl-2 (Bcl-2-ASO) did not reduce cell viability. E1A combined with Bcl-2-ASO led to significant decreases in cell viability resulting from increased apoptosis relative to cells treated with E1A alone (P < 0.05). The increase in apoptosis was partly due to cytochrome c release and subsequently caspase-9 activation by Bcl-2-ASO. Finally, in an ovarian cancer xenograft model, treatment with Bcl-2-ASO did not prolong survival, but E1A plus Bcl-2-ASO did (P < 0.001). In conclusion, ovarian tumors overexpressing Bcl-2 may not respond well to E1A gene therapy, but treatment with a combination of E1A and Bcl-2-ASO may overcome this resistance.


Subject(s)
Adenovirus E1B Proteins/genetics , Genetic Therapy/methods , Ovarian Neoplasms/therapy , Receptor, ErbB-2/biosynthesis , Thionucleotides/genetics , Animals , Apoptosis/genetics , Caspase 9 , Caspases/metabolism , Cell Line, Tumor , Cytochromes c/metabolism , Enzyme Activation , Female , Humans , Mice , Mice, Nude , Oligonucleotides, Antisense , Ovarian Neoplasms/genetics , Ovarian Neoplasms/metabolism , Receptor, ErbB-2/genetics , Thionucleotides/biosynthesis , Transfection , Xenograft Model Antitumor Assays
10.
Clin Cancer Res ; 9(15): 5652-9, 2003 Nov 15.
Article in English | MEDLINE | ID: mdl-14654548

ABSTRACT

PURPOSE: The purpose is to evaluate expression levels of Jun activation domain-binding protein 1 (JAB1) in breast cancer tissue and adjacent normal tissue, to determine whether JAB1 expression is associated with p27(Kip1) expression in invasive breast carcinomas, and to evaluate the prognostic significance of JAB1 and p27(Kip1) in node-negative breast cancer. EXPERIMENTAL DESIGN: JAB1 levels were measured in 10 matched pairs of invasive breast tumor tissue and adjacent normal tissue using Western blot analysis. We also investigated the immunoreactivity of JAB1 and p27(Kip1) levels in paraffin-embedded tissue specimens from 220 patients with node-negative breast cancer who had not received adjuvant systemic therapy. The median follow-up was 15 years. RESULTS: JAB1 was expressed at higher levels in invasive tumors than in adjacent normal tissue (P = 0.01). JAB1 overexpression was observed in 57% of invasive breast cancers. Low levels of p27(Kip1) were noted in 70% of the tumor specimens. We found an inverse correlation between JAB1 and p27(Kip1) expression levels (P = 0.01). JAB1 overexpression was associated with patient age of at least 50 years (P = 0.03) and tumor size of

Subject(s)
Breast Neoplasms/genetics , Breast Neoplasms/pathology , Cell Cycle Proteins/genetics , DNA-Binding Proteins/genetics , Gene Expression Regulation, Neoplastic/genetics , Genes, Tumor Suppressor/physiology , Transcription Factors/genetics , Tumor Suppressor Proteins/genetics , Breast Neoplasms/mortality , COP9 Signalosome Complex , Cyclin-Dependent Kinase Inhibitor p27 , Female , Humans , Immunohistochemistry , Intracellular Signaling Peptides and Proteins , Lymphatic Metastasis/genetics , Middle Aged , Peptide Hydrolases , Receptor, ErbB-2/analysis , Receptors, Estrogen/analysis , Survival Analysis , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...