Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
Add more filters










Publication year range
1.
Ann Diagn Pathol ; 67: 152214, 2023 Dec.
Article in English | MEDLINE | ID: mdl-37783147

ABSTRACT

There are immunohistochemistry (IHC) and immunofluorescence (IF) panels described in the literature and established by personal and institutional experiences that are in common use by pathologists in their daily practice. Stewardship is a difficult discussion because IHC utilization is influenced by many factors including the pathologist's experience, background, practice setting, personal bias, and medicolegal culture. We developed the methodology to audit the IHC/IF utilization in our academic subspecialty practice. We aim to share this methodology and to provide our data that can be used for consideration by other subspecialized academic practices. This analysis included a total of 63,157 specimens that were accessioned during 2022, representing 38,612 cases. The likelihood of ordering IHC/IF ranged from 1 % (in genitourinary pathology) to 59 % (in renal pathology). The average percentage of specimens with IHC/IF was 21 % for the entire practice. In cases where IHC/IF was ordered, the number of stained slides averaged 4.9 per specimen for the entire practice. The number of IHC/IF slides per specimen ranged from 1.9 (in gastrointestinal pathology) to 12.2 (in renal pathology). The highest number of antibodies ordered for a single specimen by subspecialty ranged from 11 (in cardiac pathology) to 63 (in dermatopathology). Renal pathology was the only subspecialty that had an average number of IHC/IF slides that was statistically significantly different from all other subspecialties. We described the various patterns of utilization by subspecialty and rationalized their subtle differences. We also analyzed the types of cases that exceeded the reimbursement limits set by the Centers for Medicare and Medicaid Services (CMS).


Subject(s)
Medicare , Pathologists , Aged , Humans , United States , Immunohistochemistry , Fluorescent Antibody Technique
2.
Am J Clin Pathol ; 158(5): 639-645, 2022 11 03.
Article in English | MEDLINE | ID: mdl-36208153

ABSTRACT

OBJECTIVES: We aimed to better understand the histologic changes in vaginectomy specimens in transgender and gender-diverse (TGD) individuals after prolonged androgen administration. METHODS: After obtaining institutional review board approval, we reviewed clinical records for all TGD individuals who underwent vaginal tissue resection at our institution between January 2002 and July 2020. RESULTS: Ten transgender males who underwent vaginectomy for gender affirmation were identified. All patients had been assigned female gender at birth, and the median age at surgery was 41 years (range, 22-74 years). All 10 patients had received androgen for 2 to 10 years preoperatively. The corresponding pathology specimens were examined grossly and microscopically, including with immunohistochemical stains for NKX3.1, prostate-specific antigen (PSA), p501s, and androgen receptor (AR). No gross lesions were identified. Microscopically, prostate-like glands (8/10), urothelial metaplasia (4/10), and vaginal atrophy (8/10) were identified. Seven cases with prostate-like glands showed positive staining with PSA, NKX3.1, p501s, and AR in both squamous and glandular components. CONCLUSIONS: Recognition of these androgen-related changes enables pathologist to avoid the overdiagnosis of dysplasia. Long-term follow-up is needed to thoroughly understand any potential future implications of these androgen-related changes.


Subject(s)
Transgender Persons , Transsexualism , Male , Infant, Newborn , Pregnancy , Humans , Female , Young Adult , Adult , Middle Aged , Aged , Prostate-Specific Antigen , Androgens , Colpotomy , Transcription Factors
3.
Int J Mol Sci ; 23(1)2021 Dec 22.
Article in English | MEDLINE | ID: mdl-35008514

ABSTRACT

High ERß/HER oncogenic signaling defines lung tumors with an aggressive biology. We previously showed that combining the anti-estrogen fulvestrant with the pan-HER inhibitor dacomitinib reduced ER/HER crosstalk and produced synergistic anti-tumor effects in immunocompromised lung cancer models, including KRAS mutant adenocarcinoma. How this combination affects the tumor microenvironment (TME) is not known. We evaluated the effects of fulvestrant and dacomitinib on murine bone marrow-derived macrophages (BMDMs) and CD8+ T cells, and tested the efficacy of the combination in vivo, using the KRAS mutant syngeneic lung adenocarcinoma model, FVBW-17. While this combination synergistically inhibited proliferation of FVBW-17 cells, it had unwanted effects on immune cells, by reducing CD8+ T cell activity and phagocytosis in BMDMs and inducing PD-1. The effects were largely attributed to dacomitinib, which caused downregulation of Src family kinases and Syk in immune cells. In a subcutaneous flank model, the combination induced an inflamed TME with increased myeloid cells and CD8+ T cells and enhanced PD-1 expression in the splenic compartment. Concomitant administration of anti-PD-1 antibody with fulvestrant and dacomitinib was more efficacious than fulvestrant plus dacomitinib alone. Administering anti-PD-1 sequentially after fulvestrant plus dacomitinib was synergistic, with a two-fold greater tumor inhibitory effect compared to concomitant therapy, in both the flank model and in a lung metastasis model. Sequential triple therapy has potential for treating lung cancer that shows limited response to current therapies, such as KRAS mutant lung adenocarcinoma.


Subject(s)
Estrogen Receptor beta/genetics , Lung Neoplasms/genetics , Proto-Oncogene Proteins p21(ras)/genetics , Receptor, ErbB-2/genetics , Tumor Microenvironment/genetics , Animals , CD8-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/immunology , Carcinogenesis/genetics , Carcinogenesis/immunology , Cell Line, Tumor , Estrogen Receptor beta/immunology , Female , Humans , Immunotherapy/methods , Lung Neoplasms/immunology , Lung Neoplasms/therapy , Lymphocytes, Tumor-Infiltrating/drug effects , Lymphocytes, Tumor-Infiltrating/immunology , Macrophages/drug effects , Macrophages/immunology , Mice , Oncogenes/genetics , Oncogenes/immunology , Programmed Cell Death 1 Receptor/genetics , Proto-Oncogene Proteins p21(ras)/immunology , Quinazolinones/pharmacology , Receptor, ErbB-2/immunology , Tumor Microenvironment/immunology
4.
Cancer Prev Res (Phila) ; 13(9): 735-746, 2020 09.
Article in English | MEDLINE | ID: mdl-32655003

ABSTRACT

The STAT3 pathway is frequently overactive in non-small cell lung cancer (NSCLC), an often fatal disease with known risk factors including tobacco and chemical exposures. Whether STAT3 can be downmodulated to delay or prevent development of lung cancer resulting from an environmental exposure has not been previously tested. A circular oligonucleotide STAT3 decoy (CS3D) was used to treat mice previously exposed to the tobacco carcinogen nitrosamine 4-(methylnitrosamino)-1-(3-pyridyl)-1-butanone. CS3D contains a double-stranded STAT3 DNA response element sequence and interrupts STAT3 signaling by binding to STAT3 dimers, rendering them unable to initiate transcription at native STAT3 DNA binding sites. An intermittent course of CS3D decreased the development of airway preneoplasias by 42% at 1 week posttreatment, reduced the progression of preneoplasia to adenomas by 54% at 8 weeks posttreatment, and reduced the size and number of resulting lung tumors by 49.7% and 29.5%, respectively, at 20 weeks posttreatment. No toxicity was detected. A mutant cyclic oligonucleotide with no STAT3 binding ability was used as a control. Chemopreventive effects were independent of the KRAS mutational status of the tumors. In lungs harvested during and after the treatment course with CS3D, airway preneoplasias had reduced STAT3 signaling. Chemopreventive effects were accompanied by decreased VEGFA expression, ablated IL6, COX-2, and p-NF-κB, and decreased pulmonary M2 macrophages and myeloid-derived suppressor cells. Thus, downmodulation of STAT3 activity using a decoy molecule both reduced oncogenic signaling in the airway epithelium and favored a lung microenvironment with reduced immunosuppression.


Subject(s)
Anticarcinogenic Agents/pharmacology , Carcinoma, Non-Small-Cell Lung/prevention & control , Lung Neoplasms/prevention & control , Nicotiana/toxicity , STAT3 Transcription Factor/antagonists & inhibitors , Animals , Anticarcinogenic Agents/therapeutic use , Butanones/toxicity , Carcinogenesis/chemically induced , Carcinogenesis/drug effects , Carcinogens/toxicity , Carcinoma, Non-Small-Cell Lung/chemically induced , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/immunology , Cell Proliferation/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Humans , Lung/drug effects , Lung/pathology , Lung Neoplasms/chemically induced , Lung Neoplasms/genetics , Lung Neoplasms/immunology , Mice , Mutation , Neoplasms, Experimental/chemically induced , Neoplasms, Experimental/genetics , Neoplasms, Experimental/pathology , Neoplasms, Experimental/prevention & control , Nitrosamines/toxicity , Oligonucleotides/pharmacology , Oligonucleotides/therapeutic use , Proto-Oncogene Proteins p21(ras)/genetics , STAT3 Transcription Factor/genetics , STAT3 Transcription Factor/metabolism , Signal Transduction/drug effects , Nicotiana/chemistry , Transcriptional Activation/drug effects , Tumor Microenvironment/drug effects , Tumor Microenvironment/immunology
5.
J Thorac Oncol ; 15(1): 62-79, 2020 01.
Article in English | MEDLINE | ID: mdl-31606604

ABSTRACT

INTRODUCTION: Mounting evidence supports a role for estrogen signaling in NSCLC progression. We previously reported a seven-gene signature that predicts prognosis in estrogen receptor ß positive (ERß+) NSCLC. The signature defines a network comprised of ER and human EGFR-2/3 (HER2/HER3) signaling. METHODS: We tested the efficacy of combining the pan-HER inhibitor, dacomitinib, with the estrogen antagonist, fulvestrant, in ERß+ NSCLC models with differing genotypes. We assessed the potency of this combination on xenograft growth and survival of host mice, and the ability to reverse the gene signature associated with poor outcome. RESULTS: Synergy was observed between dacomitinib and fulvestrant in three human ERß+ NSCLC models: 201T (wild-type EGFR), A549 (KRAS mutant), and HCC827 (EGFR 19 deletion) with combination indices of 0.1-0.6. The combination, but not single agents, completely reversed the gene signature associated with poor prognosis in a mechanism that is largely mediated by activator protein 1 downregulation. In vivo, the combination also induced tumor regression and reversed the gene signature. In HCC827 xenografts treated with the combination, survival of mice was prolonged after therapy discontinuation, tumors that recurred were less aggressive, and two mechanisms of HER inhibitor resistance involving c-Met activation and PTEN loss were blocked. CONCLUSIONS: The combination of an ER blocker and a pan-HER inhibitor provides synergistic efficacy in different models of ERß+ NSCLC. Our data support the use of this combination clinically, considering its ability to induce potent antitumor effects and produce a gene signature that predicts better clinical outcomes.


Subject(s)
Carcinoma, Non-Small-Cell Lung , Lung Neoplasms , Animals , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/genetics , Cell Line, Tumor , Drug Resistance, Neoplasm , Fulvestrant/pharmacology , Lung Neoplasms/drug therapy , Lung Neoplasms/genetics , Mice , Neoplasm Recurrence, Local , Signal Transduction , Xenograft Model Antitumor Assays
6.
Cancers (Basel) ; 10(9)2018 Aug 21.
Article in English | MEDLINE | ID: mdl-30134579

ABSTRACT

Hepatocyte growth factor (HGF) is the ligand for the tyrosine kinase receptor c-Met (Mesenchymal Epithelial Transition Factor also known as Hepatocyte Growth Factor Receptor, HGFR), a receptor with expression throughout epithelial and endothelial cell types. Activation of c-Met enhances cell proliferation, invasion, survival, angiogenesis, and motility. The c-Met pathway also stimulates tissue repair in normal cells. A body of past research shows that increased levels of HGF and/or overexpression of c-Met are associated with poor prognosis in several solid tumors, including lung cancer, as well as cancers of the head and neck, gastro-intestinal tract, breast, ovary and cervix. The HGF/c-Met signaling network is complex; both ligand-dependent and ligand-independent signaling occur. This article will provide an update on signaling through the HGF/c-Met axis, the mechanism of action of HGF/c-Met inhibitors, the lung cancer patient populations most likely to benefit, and possible mechanisms of resistance to these inhibitors. Although c-Met as a target in non-small cell lung cancer (NSCLC) showed promise based on preclinical data, clinical responses in NSCLC patients have been disappointing in the absence of MET mutation or MET gene amplification. New therapeutics that selectively target c-Met or HGF, or that target c-Met and a wider spectrum of interacting tyrosine kinases, will be discussed.

7.
Mol Cancer Ther ; 17(9): 1917-1926, 2018 09.
Article in English | MEDLINE | ID: mdl-29891486

ABSTRACT

Constitutively activated STAT3 plays a critical role in non-small cell lung carcinoma (NSCLC) progression by mediating proliferation and survival. STAT3 activation in normal cells is transient, making it an attractive target for NSCLC therapy. The therapeutic potential of blocking STAT3 in NSCLC was assessed utilizing a decoy approach by ligating a double-stranded 15-mer oligonucleotide that corresponds to the STAT3 response element of STAT3-target genes, to produce a cyclic STAT3 decoy (CS3D). The decoy was evaluated using NSCLC cells containing either wild-type EGFR (201T) or mutant EGFR with an additional EGFRi resistance mutation (H1975). These cells are resistant to EGFR inhibitors and require an alternate therapeutic approach. CS3D activity was compared with an inactive cyclic control oligonucleotide (CS3M) that differs by a single base pair, rendering it unable to bind to STAT3 protein. Transfection of 0.3 µmol/L of CS3D caused a 50% inhibition in proliferation in 201T and H1975 cells, relative to CS3M, and a 2-fold increase in apoptotic cells. Toxicity was minimal in normal cells. CS3D treatment caused a significant reduction of mRNA and protein expression of the STAT3 target gene c-Myc and inhibited colony formation by 70%. The active decoy decreased the nuclear pool of STAT3 compared with the mutant. In a xenograft model, treatments with CS3D (5 mg/kg) caused a potent 96.5% and 81.7% reduction in tumor growth in 201T (P < 0.007) and H1975 models (P < 0.0001), respectively, and reduced c-Myc and p-STAT3 proteins. Targeting STAT3 with the cyclic decoy could be an effective therapeutic strategy for NSCLC. Mol Cancer Ther; 17(9); 1917-26. ©2018 AACR.


Subject(s)
Carcinoma, Non-Small-Cell Lung/metabolism , ErbB Receptors/metabolism , Lung Neoplasms/metabolism , STAT3 Transcription Factor/metabolism , Animals , Apoptosis/drug effects , Apoptosis/genetics , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/genetics , Cell Line, Tumor , ErbB Receptors/genetics , Female , Gene Expression Regulation, Neoplastic/drug effects , Humans , Lung Neoplasms/drug therapy , Lung Neoplasms/genetics , Mice, Nude , Mutation , Oligonucleotides, Antisense/genetics , Oligonucleotides, Antisense/pharmacology , STAT3 Transcription Factor/genetics , Xenograft Model Antitumor Assays/methods
8.
J Thorac Oncol ; 13(3): 399-412, 2018 03.
Article in English | MEDLINE | ID: mdl-29233790

ABSTRACT

INTRODUCTION: A hormonal role in NSCLC development is well documented. We previously showed that the aromatase inhibitor (AI) anastrozole decreased development of tobacco carcinogen-induced lung tumors in a murine lung cancer prevention model and that aromatase and estrogen receptor were expressed in pulmonary inflammatory cells. METHODS: We utilized a tobacco carcinogen-induced lung tumor mouse model by treatment with 4-(methylnitrosoamino)-1-(3-pyridyl)-1-butanone (NNK), to determine whether an AI combined with nonsteroidal anti-inflammatory drugs results in greater lung tumor prevention effects compared to single-agent treatment. RESULTS: Combination of anastrozole (0.1 mg/kg/d) with aspirin (25 mg/kg/d) after NNK exposure resulted in significantly fewer and smaller lung tumors than did single-agent treatments and was accompanied by maximum decreases in circulating ß-estradiol (E2) and interleukin-6, tumor-infiltrating macrophages, and tumoral Ki67, phospho-mitogen-activated protein kinase, phospho-signal transducer and activator of transcription 3, and interleukin-17A expression. Preneoplasia arising after combination treatment showed the lowest Sox-2 expression, suggesting an inhibitory effect on proliferative capacity in the airways by blocking both E2 and inflammation. Anastrozole combined with ibuprofen instead of aspirin also showed enhanced antitumor effects. Moreover, male mice treated with NNK that received E2 in their drinking water showed greater levels of pulmonary macrophages and inflammatory markers than did the control, confirming an E2 effect on inflammation in the microenvironment. CONCLUSIONS: Our results suggest a benefit to joint targeting of the estrogen and inflammatory pathways for NSCLC prevention. Combining AIs with nonsteroidal anti-inflammatory drugs reduces circulating E2, proinflammatory cytokines, and macrophage recruitment in the lung microenvironment after tobacco exposure. This strategy could be particularly effective in women who have underlying pulmonary inflammatory diseases.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/therapeutic use , Aromatase Inhibitors/therapeutic use , Lung Neoplasms/drug therapy , Nicotiana/adverse effects , Animals , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Aromatase Inhibitors/pharmacology , Carcinogens , Disease Models, Animal , Female , Humans , Lung Neoplasms/pathology , Mice
9.
Cancer ; 123(15): 2936-2944, 2017 Aug 01.
Article in English | MEDLINE | ID: mdl-28472537

ABSTRACT

BACKGROUND: Activation of the mesenchymal-epidermal transition factor (MET) tyrosine kinase and its ligand, hepatocyte growth factor (HGF), is implicated in resistance to epidermal growth factor receptor (EGFR) inhibitors. In this phase 1/2 trial, rilotumumab (an anti-HGF antibody) combined with erlotinib was evaluated in patients with metastatic, previously treated non-small cell lung cancer. METHODS: In phase 1, a dose de-escalation design was adopted with rilotumumab starting at 15 mg/kg intravenously every 3 weeks and oral erlotinib 150 mg daily. In phase 2, the disease control rate (DCR) (according to Response Evaluation Criteria in Solid Tumors) of the combination was evaluated using a Simon 2-stage design. The biomarkers examined included 10 plasma-circulating molecules associated with the EGFR and MET pathways. RESULTS: Without indications for de-escalation, the recommended phase 2 dose was dose level 0. Overall, 45 response-evaluable patients were enrolled (13 with squamous carcinoma, 32 with adenocarcinoma; 2 had confirmed EGFR mutations, 33 had confirmed wild-type [WT] EGFR, and 7 had KRAS mutations). The DCR for all patients was 60% (90% confidence interval [CI], 47.1%-71.3%). Median progression-free survival was 2.6 months (90% CI, 1.4-2.7 months), and median overall survival was 6.6 months (90% CI, 5.6-8.9 months). Among patients with WT EGFR, the DCR was 60.6% (90% CI, 46.3%-73.3%), median progression-free survival was 2.6 months (90% CI, 1.4-2.7 months), and median overall survival was 7.0 months (90% CI, 5.6-13.4 months). Elevated baseline levels of neuregulin 1 were associated with longer progression-free survival (hazard ratio, 0.41; 95% CI, 0.19-0.87), whereas elevated amphiregulin levels were associated with more rapid progression (hazard ratio, 2.14; 95% CI, 1.48-3.08). CONCLUSIONS: Combined rilotumumab and erlotinib had an acceptable safety profile, and the DCR met the prespecified criteria for success. In the EGFR WT group, the DCR exceeded published reports for erlotinib alone. High circulating levels of neuregulin 1 may indicate sensitivity to this combination. Cancer 2017;123:2936-44. © 2017 American Cancer Society.


Subject(s)
Adenocarcinoma/drug therapy , Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Squamous Cell/drug therapy , Lung Neoplasms/drug therapy , Adenocarcinoma/genetics , Adenocarcinoma/secondary , Adult , Aged , Aged, 80 and over , Antibodies, Monoclonal/administration & dosage , Antibodies, Monoclonal, Humanized , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/secondary , Carcinoma, Squamous Cell/genetics , Carcinoma, Squamous Cell/secondary , Disease-Free Survival , ErbB Receptors/genetics , Erlotinib Hydrochloride/administration & dosage , Female , Humans , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Male , Middle Aged , Mutation , Proportional Hazards Models , Treatment Outcome
10.
Transl Res ; 185: 13-23, 2017 07.
Article in English | MEDLINE | ID: mdl-28554003

ABSTRACT

We have previously shown that topical opioids including morphine and its congeners promote healing of full thickness ischemic wounds in rats. We examined the contribution of mu opioid receptor (MOPr)-mediated healing of full thickness ischemic wounds using MOPr and delta or kappa opioid receptor knockout (KO) mice. Wound closure in the early (day 5) as well as later phases was delayed in topical morphine or PBS-treated MOPr-KO mice compared with reciprocal treatments of wounds in wild-type (WT) mice. MOPr expression was significantly upregulated at 30 min in the wound margins and colocalized with wound margins and vasculature in the epidermal and dermal layers of the skin. We next examined whether neuropeptide expression was involved in the mechanism of MOPr-mediated wound closure. Substance P (SP) and calcitonin gene-related peptide immunoreactivity (ir) was significantly increased in the skin of MOPr-KO mice as compared with WT mice. Neuropeptide-ir was increased significantly in PBS-treated wounds of MOPr and WT mice, but morphine treatment reduced neuropeptide immunoreactivity in both as compared with PBS. Wounding of keratinocytes led to the release of opioid peptide beta-endorphin (ß-END) in conditioned medium, which stimulated the proliferation of endothelial cells. MOPr-selective (D-Phe-Cys-Tyr-D-Trp-Orn-Thr-Pen-Thr-NH2, CTOP) and nonselective OPr antagonist naloxone-inhibited endothelial proliferation induced by wounded keratinocyte-conditioned medium. In addition, accelerated wound area closure in vitro by morphine was suppressed by methylnaltrexone, a nonselective OPr antagonist with high affinity for MOPr. Morphine and its congeners stimulated the proliferation of endothelial cells from WT mice but not those from MOPr-KO mice. Furthermore, morphine-induced mitogen-activated protein kinase/extracellular signal-regulated kinase phosphorylation in endothelial cells was significantly decreased in MOPr-KO mice as compared with WT mice. Collectively, these data suggest that MOPr plays a critical role in the proliferation phase with the formation of granulation tissue during wound healing.


Subject(s)
Analgesics, Opioid/therapeutic use , Ischemia/pathology , Morphine/therapeutic use , Receptors, Opioid/metabolism , Wound Healing/physiology , Administration, Topical , Analgesics, Opioid/administration & dosage , Animals , Gene Expression Regulation/physiology , Humans , Keratinocytes/drug effects , Mice , Mice, Knockout , Morphine/administration & dosage , Receptors, Opioid/genetics , Up-Regulation
11.
Oncotarget ; 8(15): 24063-24076, 2017 Apr 11.
Article in English | MEDLINE | ID: mdl-28445992

ABSTRACT

The estrogen receptor (ER) promotes non-small cell lung cancer (NSCLC) proliferation. Since fibroblast growth factors (FGFs) are known regulators of stem cell markers in ER positive breast cancer, we investigated whether a link between the ER, FGFs, and stem cell markers exists in NSCLC. In lung preneoplasias and adenomas of tobacco carcinogen exposed mice, the anti-estrogen fulvestrant and/or the aromatase inhibitor anastrozole blocked FGF2 and FGF9 secretion, and reduced expression of the stem cell markers SOX2 and nanog. Mice administered ß-estradiol during carcinogen exposure showed increased FGF2, FGF9, SOX2, and Nanog expression in airway preneoplasias. In normal FGFR1 copy number NSCLC cell lines, multiple FGFR receptors were expressed and secreted several FGFs. ß-estradiol caused enhanced FGF2 release, which was blocked by fulvestrant. Upon co-inhibition of ER and FGFRs using fulvestrant and the pan-FGFR inhibitor AZD4547, phosphorylation of FRS2, the FGFR docking protein, was maximally reduced, and enhanced anti-proliferative effects were observed. Combined AZD4547 and fulvestrant enhanced lung tumor xenograft growth inhibition and decreased Ki67 and stem cell marker expression. To verify a link between ERß, the predominant ER in NSCLC, and FGFR signaling in patient tumors, mRNA analysis was performed comparing high versus low ERß expressing tumors. The top differentially expressed genes in high ERß tumors involved FGF signaling and human embryonic stem cell pluripotency. These results suggest interaction between the ER and FGFR pathways in NSCLC promotes a stem-like state. Combined FGFR and ER inhibition may increase the efficacy of FGFR inhibitors for NSCLC patients lacking FGFR genetic alterations.


Subject(s)
Carcinoma, Non-Small-Cell Lung/metabolism , Lung Neoplasms/metabolism , Receptors, Estrogen/metabolism , Receptors, Fibroblast Growth Factor/metabolism , Signal Transduction , Animals , Antineoplastic Agents, Hormonal/pharmacology , Benzamides/pharmacology , Biomarkers , Carcinoma, Non-Small-Cell Lung/pathology , Cell Line, Tumor , Cell Proliferation/drug effects , Disease Models, Animal , Estradiol/analogs & derivatives , Estradiol/pharmacology , Estrogens/metabolism , Female , Fibroblast Growth Factors/biosynthesis , Fibroblast Growth Factors/metabolism , Fulvestrant , Humans , Ligands , Lung Neoplasms/pathology , Mice , Neoplastic Stem Cells/metabolism , Piperazines/pharmacology , Protein Binding , Pyrazoles/pharmacology , Receptors, Estrogen/antagonists & inhibitors , Receptors, Fibroblast Growth Factor/antagonists & inhibitors , Signal Transduction/drug effects , Xenograft Model Antitumor Assays
12.
J Cardiovasc Pharmacol ; 69(5): 314-325, 2017 May.
Article in English | MEDLINE | ID: mdl-28207427

ABSTRACT

OBJECTIVE: Although atherosclerosis is described in New Zealand White rabbit's iliac artery, yet details of time-dependent atherosclerosis progression are not well known. Further, a well characterized accelerated model of atherosclerosis is also required for the screening of candidate drugs to target specific steps of atherosclerosis development. The present study extensively characterizes the time-dependent plaque composition and functional responses of the atherosclerosis in rabbit iliac artery and its modification by simvastatin. METHODS: Atherosclerosis was induced with a combination of balloon injury and atherogenic diet (AD) (1% cholesterol, 6% peanut oil) in rabbit's iliac artery. Atherosclerosis progression was evaluated on days 8, 10, 15, 21, 35, and 56 after AD feeding. The plaque characterization was done using histology, real-time reverse transcription-polymerase chain reaction, and vasoreactivity experiments. The standard anti-hyperlipidemic drug, simvastatin (5 mg·kg·d), was used to investigate its effect on atherosclerotic changes. RESULTS: Plasma lipids were elevated in a progressive manner after AD feeding from days 8 to 56. Similarly, arterial lipids, Monocyte Chemoattractant Protein-1 (MCP-1) level along with infiltration of macrophages in the lesion area were also increased from day 15 onward. This resulted in a significant increase in the plaque area and intimal-medial thickness ratio in contrast to normal animals. Inflammatory milieu was observed with a significant increase in expression of pro-inflammatory regulators like MCP-1, Tumor Necrosis Factor-α (TNF-α) and Vascular Cell Adhesion Molecule-1 (VCAM-1), whereas anti-inflammatory cytokine interleukin 10 decreased as disease progressed. Endothelial dysfunction was also observed, specifically Acetylcholine (ACh)-induced vasorelaxation was reduced from day 8 onward, whereas the phenylephrine-induced vasoconstriction response was progressively reduced from day 15 in the iliac artery. Ground substances including proteoglycans, α-actin, and collagen content along with metalloproteinase-9 and Tissue inhibitor of metalloproteinases-1 (TIMP-1) inhibitors were significantly augmented at later time points, day 21 onward. Simvastatin treatment for 35 days, at a dose having no significant effect on plasma lipid levels, significantly reduced atherosclerotic progression as evident by reduced macrophage content, inflammatory burden, and extracellular matrix component like proteoglycans and metalloproteinase-9. CONCLUSIONS: The authors observed that AD feeding with balloon injury in the rabbit iliac artery accelerated the progression of atherosclerosis and exhibited predominant features of type III human lesion within 8 weeks (56 days). Simvastatin treatment for 35 days exhibited anti-atherosclerotic efficacy without significantly lowering the circulating lipids. The current study thus provides an insight into the time-dependent atherosclerotic progression in rabbit iliac artery and highlights its utility for anti-atherosclerotic evaluation of the candidate drugs.


Subject(s)
Atherosclerosis/drug therapy , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology , Iliac Artery/drug effects , Plaque, Atherosclerotic , Simvastatin/pharmacology , Angioplasty, Balloon , Animals , Atherosclerosis/blood , Atherosclerosis/pathology , Atherosclerosis/physiopathology , Biomarkers/blood , Diet, High-Fat , Disease Models, Animal , Disease Progression , Endothelium, Vascular/drug effects , Endothelium, Vascular/physiopathology , Extracellular Matrix/metabolism , Iliac Artery/metabolism , Iliac Artery/pathology , Iliac Artery/physiopathology , Inflammation Mediators/blood , Lipids/blood , Macrophages/drug effects , Macrophages/metabolism , Male , Rabbits , Time Factors , Vascular Remodeling/drug effects , Vasoconstriction/drug effects , Vasodilation/drug effects
13.
Mol Cancer ; 14: 138, 2015 Jul 29.
Article in English | MEDLINE | ID: mdl-26215578

ABSTRACT

BACKGROUND: The epidermal growth factor (EGF) family of ligands has been implicated in promoting breast cancer initiation, growth and progression. The contributions of EGF family ligands and their receptors to breast cancer are complex, and the specific mechanisms through which different ligands regulate breast tumor initiation and growth are not well-defined. These studies focus on the EGF family member epiregulin (EREG) as a mediator of early stage breast tumorigenesis. METHODS: EREG expression levels were assessed in both cell lines and human samples of ductal carcinoma in situ (DCIS) using quantitative RT-PCR, ELISA and immunohistochemistry. Gene knock-down approaches using shRNA-based strategies were used to determine the requirement of EREG for growth of MCF10DCIS cells in vivo, and for identifying mechanisms through which EREG promotes tumor cell survival. Experiments were performed using a combination of two-dimensional culture, three-dimensional culture and tumor growth in vivo. RESULTS: In comparison with other EGF family members, EREG was induced in MCF10DCIS cells compared with MCF10A and MCF10AT cells and its expression was partially regulated by fibroblast growth factor receptor (FGFR) activity. Reduced EREG expression in MCF10DCIS cells led to decreased tumor growth in vivo, which was associated with reduced cell survival. Furthermore, treatment of MCF10A cells with exogenous EREG enhanced cell survival both in three-dimensional culture and in response to chemotherapeutic agents. Examination of EREG-induced signaling pathways demonstrated that EREG promoted survival of MCF10A cells through regulating expression of matrix metalloproteinase-1 (MMP-1). To determine the relevance of these findings in human tumors, samples of DCIS were analyzed for EREG and MMP-1 expression. EREG was induced in DCIS lesions compared to normal breast epithelium, and EREG and MMP-1 were correlated in a subset of DCIS samples. CONCLUSIONS: Together, these studies lead to identification of a novel pathway involving EREG and MMP-1 that contributes to the formation of early stage breast cancer. Understanding these complex pathways could ultimately lead to the development of novel biomarkers of neoplastic progression and/or new therapeutic strategies for patients with early stage cancer.


Subject(s)
Breast Neoplasms/genetics , Carcinogenesis/genetics , Carcinoma, Intraductal, Noninfiltrating/genetics , Epiregulin/biosynthesis , Matrix Metalloproteinase 1/biosynthesis , Breast Neoplasms/pathology , Carcinogenesis/pathology , Carcinoma, Intraductal, Noninfiltrating/pathology , Cell Line, Tumor , Epiregulin/genetics , Female , Gene Expression Regulation, Neoplastic , Humans , Matrix Metalloproteinase 1/genetics , Neoplasm Staging , Receptor, Fibroblast Growth Factor, Type 1/genetics
14.
J Diabetes ; 7(4): 573-583, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25266258

ABSTRACT

BACKGROUND: Topically applied opioids promote angiogenesis and healing of ischemic wounds in rats. We examined if topical fentanyl stimulates wound healing in diabetic rats by stimulating growth-promoting signaling, angiogenesis, lymphangiogenesis and nerve regeneration. METHODS: We used Zucker diabetic fatty rats that develop obesity and diabetes on a high fat diet due to a mutation in the Leptin receptor. Fentanyl blended with hydrocream was applied topically on ischemic wounds twice daily, and wound closure was analyzed regularly. Wound histology was analyzed by hematoxylin and eosin staining. Angiogenesis, lymphangiogenesis, nerve fibers and phospho-platelet derived growth factor receptor-ß (PDGFR-ß) were visualized by CD31-, lymphatic vessel endothelium-1, protein gene product 9.5- and anti-phospho PDGFR-ß-immunoreactivity, respectively. Nitric oxide synthase (NOS) and PDGFR-ß signaling were analyzed using Western immunoblotting. RESULTS: Fentanyl significantly promoted wound closure as compared to phosphate-buffered saline (PBS). Histology scores were significantly higher in fentanyl-treated wounds, indicative of increased granulation tissue formation, reduced edema and inflammation, and increased matrix deposition. Fentanyl treatment resulted in increased wound angiogenesis, lymphatic vasculature, nerve fibers, nitric oxide, NOS and PDGFR-ß signaling as compared to PBS. Phospho-PDGFR-ß co-localized with CD31 co-staining for vasculature. CONCLUSIONS: Topically applied fentanyl promotes closure of ischemic wounds in diabetic rats. Increased angiogenesis, lymphangiogenesis, peripheral nerve regeneration, NO and PDGFR-ß signaling are associated with fentanyl-induced tissue remodeling and wound healing.


Subject(s)
Analgesics, Opioid/pharmacology , Diabetes Mellitus, Experimental/physiopathology , Fentanyl/pharmacology , Ischemia/physiopathology , Wound Healing/drug effects , Animals , Rats , Rats, Zucker
15.
Cancer ; 117(19): 4493-505, 2011 Oct 01.
Article in English | MEDLINE | ID: mdl-21437888

ABSTRACT

BACKGROUND: The carbohydrate sialyl Lewis X (sLeX) is expressed on leukocytes and carcinoma cells and binds to selectins during inflammatory processes and early metastasis. Synthesis of sLeX depends on activity of enzymes, including α(1,3/1,4) fucosyltransferase (FucT-III). Tumor necrosis factor-α (TNF-α) up-regulates FucT-III, resulting in increased sLeX in the airways of patients with respiratory disease; however, the mechanisms that regulate sLeX in the inflammatory tumor microenvironment are not well understood. METHODS: The authors stably transfected human lung carcinoma cell lines with the FucT-III gene and exposed them to TNF-α to investigate its role in regulation of sLeX expression and selectin-binding ability using semiquantitative real-time polymerase chain reaction and flow cytometry. Cytokine expression was examined in transfected cells using chemiluminescent arrays and enzyme-linked immunosorbent assays, and invasion was studied using Matrigel assays and alterations in morphology. Human lung tissue arrays were analyzed for immunohistochemical detection of sLeX and neutrophils. RESULTS: Stimulation of FucT-III-transfected cells with recombinant human (rh) TNF-α up-regulated sLeX expression and increased E-selectin binding. Transfected cells secreted high levels of interleukin 8, growth-regulated oncogene-α, and mast cell proteinase-1. Cells exposed to rhTNF-α, neutrophil-conditioned media, and cultures with a 5:1 ratio of neutrophils to cancer cells had significantly increased sLeX expression and invasiveness and underwent nonadherent morphologic changes. In lung carcinomas, but not in normal lung tissues, 71% of tumors were highly positive for sLeX expression in areas of increased neutrophil infiltration. CONCLUSIONS: The current results indicated that neutrophils may be recruited to areas of FucT-III activity and sLeX expression in lung carcinomas to enhance the invasive and metastatic potential of lung cancer cells.


Subject(s)
Carcinoma, Non-Small-Cell Lung/immunology , Carcinoma, Non-Small-Cell Lung/pathology , Lung Neoplasms/immunology , Lung Neoplasms/pathology , Neutrophils/immunology , Oligosaccharides/metabolism , Blotting, Western , Carcinoma, Non-Small-Cell Lung/metabolism , E-Selectin/genetics , E-Selectin/metabolism , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Fucosyltransferases/genetics , Fucosyltransferases/metabolism , Humans , Immunoenzyme Techniques , Lung Neoplasms/metabolism , Neoplasm Invasiveness , Neutrophils/metabolism , Oligosaccharides/genetics , RNA, Messenger/genetics , Reverse Transcriptase Polymerase Chain Reaction , Sialyl Lewis X Antigen , Tumor Cells, Cultured , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/metabolism
16.
PLoS One ; 6(1): e16281, 2011 Jan 19.
Article in English | MEDLINE | ID: mdl-21283832

ABSTRACT

Similar to mechanisms of recruitment of activated leukocytes to inflamed tissues, selectins mediate adhesion and extravasation of circulating cancer cells. Our objective was to determine whether sialyl Lewis X modified core 2 O-glycans (C2-O-sLe(X)) present on colon and hepatic carcinoma cells promote their adhesion and invasion. We examined membrane expression of C2-O-sLe(X), selectin binding, invasion of human colon and hepatic carcinoma cell lines, and mRNA levels of alpha-2,3 fucosyltransferase (FucT-III) and core 2 beta-1,6 N-acetylglucosaminyltransferase (C2GnT1) genes, necessary for C2-O-sLe(X) synthesis, by quantitative reverse-transcriptase (RT) PCR. Synthesis of core 2 branched O-glycans decorated by sLe(X) is dependent on C2GnT1 function and thus we determined enzyme activity of C2GnT1. The cell lines that expressed C2GnT1 and FucT-III mRNA by quantitative RT-PCR were highly positive for C2-O-sLe(X) by flow cytometry, and colon carcinoma cells possessed highly active C2GnT1 enzyme. Cells bound avidly to E-selection but not to P- and L-selectin. Gene knock-down of C2GnT1 in colon and hepatic carcinoma cells using short hairpin RNAs (shRNA) resulted in a 40-90% decrease in C2-O-sLe(X) and a 30-50% decrease in E-selectin binding compared to control cells. Invasion of hepatic and colon carcinoma cells containing C2GnT1 shRNA was significantly reduced compared to control cells in Matrigel assays and C2GnT1 activity was down-regulated in the latter cells. The sLe(X) epitope was predominantly distributed on core 2 O-glycans on colon and hepatic carcinoma cells. Our findings indicate that C2GnT1 gene expression and the resulting C2-O-sLe(X) carbohydrates produced mediate the adhesive and invasive behaviors of human carcinomas which may influence their metastatic potential.


Subject(s)
Carcinoma, Hepatocellular/pathology , Colonic Neoplasms/pathology , E-Selectin/metabolism , Neoplasm Invasiveness/pathology , Oligosaccharides/physiology , Carcinoma, Hepatocellular/chemistry , Cell Adhesion , Cell Line, Tumor , Colonic Neoplasms/chemistry , Epitopes/analysis , Glycoproteins/analysis , Glycoproteins/physiology , Humans , Ligands , N-Acetylglucosaminyltransferases/genetics , Oligosaccharides/analysis , RNA, Messenger/analysis
17.
BMC Cancer ; 9: 79, 2009 Mar 06.
Article in English | MEDLINE | ID: mdl-19267921

ABSTRACT

BACKGROUND: The metastasis of cancer cells and leukocyte extravasation into inflamed tissues share common features. Specialized carbohydrates modified with sialyl Lewis x (sLex) antigens on leukocyte membranes are ligands for selectin adhesion molecules on activated vascular endothelial cells at inflammatory sites. The activity of the enzyme core 2 beta1,6 N-acetylglucosaminyltransferase (C2GnT1) in leukocytes greatly increases their ability to bind to endothelial selectins. C2GnT1 is essential for the synthesis of core 2-branched O-linked carbohydrates terminated with sLex (C2-O-sLex). Our goal was to determine the expression profiles of C2-O-sLex in the malignant progression and metastasis of colorectal adenocarcinomas. The well characterized CHO-131 monoclonal antibody (mAb) specifically recognizes C2-O-sLex present in human leukocytes and carcinoma cells. Using CHO-131 mAb, we investigated whether C2-O-sLex was present in 113 human primary colorectal adenocarcinomas, 10 colorectal adenomas, 46 metastatic liver tumors, 28 normal colorectal tissues, and 5 normal liver tissues by immunohistochemistry. We also examined mRNA levels of the enzyme core 2 beta1,6-N-acetylglucosaminyltransferase (C2GnT1) in 20 well, 15 moderately, and 2 poorly differentiated colorectal adenocarcinomas, and in 5 normal colorectal tissues by using quantitative real-time polymerase chain reactions (RT-PCR). RESULTS: We observed high reactivity with CHO-131 mAb in approximately 70% of colorectal carcinomas and 87% of metastatic liver tumors but a lack of reactivity in colorectal adenomas and normal colonic and liver tissues. Positive reactivity with CHO-131 mAb was very prominent in neoplastic colorectal glands of well to moderately differentiated adenocarcinomas. The most intense staining with CHO-131 mAb was observed at the advancing edge of tumors with the deepest invasive components.Finally, we analyzed C2GnT1 mRNA levels in 37 colorectal adenocarcinomas and 5 normal colorectal tissues by RT-PCR. Significantly, we observed a greater than 15-fold increase in C2GnT1 mRNA levels in colorectal adenocarcinomas compared to normal colorectal tissues. CONCLUSION: C2-O-sLex, detected by the CHO-131 mAb, is a tumor associated antigen whose expression is highly upregulated in colorectal adenocarcinomas and metastatic liver tumors compared to normal tissues. C2-O-sLex is a potentially useful early predictor of metastasis.


Subject(s)
Adenocarcinoma/metabolism , Antigens, Neoplasm/metabolism , Biomarkers, Tumor/metabolism , Colorectal Neoplasms/metabolism , Liver Neoplasms/metabolism , N-Acetylglucosaminyltransferases/metabolism , Adenocarcinoma/diagnosis , Adenocarcinoma/genetics , Adenocarcinoma/pathology , Antigens, Neoplasm/genetics , Biomarkers, Tumor/genetics , Carbohydrate Metabolism , Colon/cytology , Colon/metabolism , Colorectal Neoplasms/diagnosis , Colorectal Neoplasms/genetics , Colorectal Neoplasms/pathology , E-Selectin/metabolism , Humans , Immunohistochemistry , Liver/cytology , Liver/metabolism , Liver Neoplasms/diagnosis , Liver Neoplasms/genetics , Liver Neoplasms/pathology , N-Acetylglucosaminyltransferases/genetics , Neoplasm Metastasis , Neoplasm Staging , Polysaccharides/metabolism , Prognosis , Protein Binding , RNA, Messenger/analysis , Reverse Transcriptase Polymerase Chain Reaction
18.
Am J Physiol Renal Physiol ; 294(6): F1388-97, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18385270

ABSTRACT

Morphine sulfate (MS) stimulates mesangial cell (MC) proliferation, a process central to development of glomerular disease. The purpose of this study was to examine whether specific opioid receptors (OR) and signal transducer and activators of transcription 3 (STAT3) signaling are associated with MS-induced MC proliferation. C57Bl/6J and OR-specific knockout (KO) mice were treated for up to 6 wk with PBS, MS (0.7-2.14 mg/kg), naloxone (equimolar to MS), or MS+naloxone (n = 6 per group). Glomerular volume and expression of PCNA, Thy1, and ED1/CD68 were analyzed in kidney sections. Cell proliferation and STAT3 phosphorylation were analyzed by bromodeoxyuridine (BrdU) ELISA and Western blot, respectively, in MCs in vitro. MS treatment led to enlarged kidneys and glomerulopathy and naloxone reversed these effects. MS treatment increased glomerular volume in both mu-OR (MOR) KO and delta-OR (DOR) KO mice, but not in kappa-OR (KOR) KO mice. To ascertain that MS-induced glomerulopathy in vivo was due to MC proliferation, we further examined the OR-specific effects of MS in MCs in vitro. MS-induced MC proliferation in vitro was inhibited by KOR-specific nor-BNI, but not by DOR or MOR-specific antagonists naltrindol or CTOP, respectively. KOR-specific agonist U50488H stimulated proliferation of MCs, but DOR-specific agonist DPDPE and MOR-specific agonist DAMGO did not. MS failed to stimulate proliferation of MCs from KOR KO mice. MS and KOR agonists induced STAT3 phosphorylation, and STAT3 inhibitor blocked KOR agonist-induced MC proliferation. We show that MS stimulates glomerulopathy and MC proliferation via KOR and STAT3 signaling.


Subject(s)
Analgesics, Opioid/toxicity , Kidney Diseases/chemically induced , Mesangial Cells/drug effects , Mesangial Cells/pathology , Morphine/toxicity , Receptors, Opioid, kappa/metabolism , Animals , Cell Division/drug effects , Cells, Cultured , Female , Glomerular Filtration Rate/drug effects , Kidney Diseases/pathology , Kidney Diseases/physiopathology , Male , Mesangial Cells/physiology , Mice , Mice, Inbred C57BL , Mice, Knockout , Phosphorylation/drug effects , Receptors, Opioid, kappa/genetics , STAT3 Transcription Factor/metabolism , Signal Transduction/drug effects
19.
Nucleic Acids Res ; 35(7): 2269-82, 2007.
Article in English | MEDLINE | ID: mdl-17389641

ABSTRACT

Orphan nuclear receptor TR2 is a preadipocyte proliferator. Knockdown of TR2 in 3T3-L1 preadipocytes reduced their proliferation efficiency, whereas specific elevation of TR2 in these cells facilitated their proliferation. All-trans retinoic acid (RA) stimulates cellular proliferation in 3T3-L1 preadipocytes by activating TR2 through an IR0-type RA response element, which further activates c-Myc expression. In post-differentiated adipocytes, RA becomes a repressive signal for TR2 and rapidly down-regulates its expression. The biphasic effect of RA on TR2 expression in 3T3-L1 is mediated by differential RA-dependent coregulator recruitment to the receptor/Glucocorticoid Receptor-Interacting Protein 1 (GRIP1) complex that binds IR0 on the TR2 promoter. RA induces the recruitment of histone acetyl transferase-containing/GRIP1/p300/CBP-associated factor (PCAF) complex to the TR2 promoter in undifferentiated cells, whereas it triggers recruitment of histone deacetylase-containing/GRIP1/receptor-interacting protein 140 (RIP140) complex in differentiated cells. GRIP1 directly interacts with RIP140 through its carboxyl terminal AD2 domain. GRIP1 interacts with PCAF and RIP140 directly and differentially, functioning as a platform molecule to mediate differential RA-induced coregulator recruitment to TR2 promoter target. This results in a biphasic effect of RA on the expression of TR2 in undifferentiated and differentiated cells, which is required for RA-stimulated preadipocyte proliferation.


Subject(s)
Adipocytes/metabolism , Receptors, Thyroid Hormone/metabolism , Stem Cells/metabolism , Transcription Factors/metabolism , Tretinoin/pharmacology , 3T3-L1 Cells , Adaptor Proteins, Signal Transducing/chemistry , Adaptor Proteins, Signal Transducing/metabolism , Adipocytes/cytology , Adipocytes/drug effects , Animals , Binding Sites , Cell Cycle Proteins/metabolism , Cell Differentiation , Cell Proliferation/drug effects , Histone Acetyltransferases/metabolism , Mice , Nuclear Proteins/chemistry , Nuclear Proteins/metabolism , Nuclear Receptor Coactivator 2/chemistry , Nuclear Receptor Coactivator 2/metabolism , Nuclear Receptor Interacting Protein 1 , Nuclear Receptor Subfamily 2, Group C, Member 1 , Promoter Regions, Genetic , Protein Structure, Tertiary , Receptors, Thyroid Hormone/antagonists & inhibitors , Receptors, Thyroid Hormone/physiology , Repressor Proteins/chemistry , Repressor Proteins/metabolism , Stem Cells/cytology , Stem Cells/drug effects , p300-CBP Transcription Factors
20.
Curr Neurovasc Res ; 3(3): 171-80, 2006 Aug.
Article in English | MEDLINE | ID: mdl-16918381

ABSTRACT

Go/Gi coupled G-protein receptor mediated transactivation is critical in the activation of receptor tyrosine kinases (RTK). Here we show that mu opioid receptor (MOR) transactivates Flk1 and platelet-derived growth factor-beta (PDGF-beta) receptors and its agonist morphine stimulates pro-angiogenic and survival-promoting signaling in mouse retinal endothelial cells (mREC). Morphine stimulates mREC proliferation in a dose dependent fashion and promotes survival to the same extent as vascular endothelial growth factor164 (VEGF164). Morphine stimulates mitogen-activated protein kinase/extracellular signal-regulated kinase (MAPK/ERK) and Akt phosphorylation in a time dependent manner like VEGF in mREC. Moreover, analogous to VEGF, morphine stimulates oncogenic signal transducer and activator of transcription 3 (STAT3) signaling. Morphine as well as VEGF-induced phospho-STAT3 and phospho-Flk1 immunoprecipitated with MOR-associated proteins. In addition morphine also stimulated MOR associated PDGF-beta receptor phosphorylation. Consistent with the relationship between VEGF and MOR we found that VEGF upregulates MOR protein and RNA expression in mREC. These data suggest that MOR associates and transactivates RTKs for Flk1 and PDGF-beta, which may have a compounding effect on angiogenic signaling in endothelium. Therefore, G-Protein coupled receptors including MOR provide novel targets to develop anti-angiogenic agents.


Subject(s)
Analgesics, Opioid/pharmacology , Endothelial Cells/drug effects , Morphine/pharmacology , Receptors, Opioid, mu/metabolism , Retinal Vessels/metabolism , Signal Transduction/drug effects , Animals , Apoptosis/drug effects , Blotting, Western , Cell Proliferation/drug effects , Dose-Response Relationship, Drug , Fluorescent Antibody Technique , Immunoprecipitation , Mice , Mice, Inbred C57BL , Mitogen-Activated Protein Kinases/metabolism , Organ Culture Techniques , Phosphorylation , Proto-Oncogene Proteins c-akt/metabolism , RNA, Messenger/analysis , Receptor, Platelet-Derived Growth Factor beta/drug effects , Receptor, Platelet-Derived Growth Factor beta/metabolism , Reverse Transcriptase Polymerase Chain Reaction , STAT3 Transcription Factor/drug effects , Transcriptional Activation , Vascular Endothelial Growth Factor A/drug effects , Vascular Endothelial Growth Factor A/metabolism , Vascular Endothelial Growth Factor Receptor-2/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...