Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
4.
Cancer Res ; 78(12): 3233-3242, 2018 06 15.
Article in English | MEDLINE | ID: mdl-29661830

ABSTRACT

Mounting clinical and preclinical evidence supports a key role for sustained adrenergic signaling in the tumor microenvironment as a driver of tumor growth and progression. However, the mechanisms by which adrenergic neurotransmitters are delivered to the tumor microenvironment are not well understood. Here we present evidence for a feed-forward loop whereby adrenergic signaling leads to increased tumoral innervation. In response to catecholamines, tumor cells produced brain-derived neurotrophic factor (BDNF) in an ADRB3/cAMP/Epac/JNK-dependent manner. Elevated BDNF levels in the tumor microenvironment increased innervation by signaling through host neurotrophic receptor tyrosine kinase 2 receptors. In patients with cancer, high tumor nerve counts were significantly associated with increased BDNF and norepinephrine levels and decreased overall survival. Collectively, these data describe a novel pathway for tumor innervation, with resultant biological and clinical implications.Significance: Sustained adrenergic signaling promotes tumor growth and metastasis through BDNF-mediated tumoral innervation. Cancer Res; 78(12); 3233-42. ©2018 AACR.


Subject(s)
Brain-Derived Neurotrophic Factor/metabolism , Feedback, Physiological , Neoplasms/pathology , Norepinephrine/metabolism , Receptors, Adrenergic, beta-3/metabolism , Animals , Cell Line, Tumor , Cyclic AMP/metabolism , Female , Guanine Nucleotide Exchange Factors/metabolism , Humans , Membrane Glycoproteins/metabolism , Mice , Neoplasms/mortality , Peripheral Nerves/metabolism , Peripheral Nerves/pathology , Receptor, trkB/metabolism , Signal Transduction , Tumor Microenvironment/physiology , Xenograft Model Antitumor Assays
5.
JCI Insight ; 2(16)2017 Aug 17.
Article in English | MEDLINE | ID: mdl-28814667

ABSTRACT

Adrenergic signaling is known to promote tumor growth and metastasis, but the effects on tumor stroma are not well understood. An unbiased bioinformatics approach analyzing tumor samples from patients with known biobehavioral profiles identified a prominent stromal signature associated with cancer-associated fibroblasts (CAFs) in those with a high biobehavioral risk profile (high Center for Epidemiologic Studies Depression Scale [CES-D] score and low social support). In several models of epithelial ovarian cancer, daily restraint stress resulted in significantly increased CAF activation and was abrogated by a nonspecific ß-blocker. Adrenergic signaling-induced CAFs had significantly higher levels of collagen and extracellular matrix components than control tumors. Using a systems-based approach, we found INHBA production by cancer cells to induce CAFs. Ablating inhibin ß A decreased CAF phenotype both in vitro and in vivo. In preclinical models of breast and colon cancers, there were increased CAFs and collagens following daily restraint stress. In an independent data set of renal cell carcinoma patients, there was an association between high depression (CES-D) scores and elevated expression of ACTA2, collagens, and inhibin ß A. Collectively, our findings implicate adrenergic influences on tumor stroma as important drivers of CAFs and establish inhibin ß A as an important regulator of the CAF phenotype in ovarian cancer.

6.
Arterioscler Thromb Vasc Biol ; 35(12): 2647-56, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26494233

ABSTRACT

OBJECTIVE: Prenatal deletion of the type II transforming growth factor-ß (TGF-ß) receptor (TBRII) prevents normal vascular morphogenesis and smooth muscle cell (SMC) differentiation, causing embryonic death. The role of TBRII in adult SMC is less well studied. Clarification of this role has important clinical implications because TBRII deletion should ablate TGF-ß signaling, and blockade of TGF-ß signaling is envisioned as a treatment for human aortopathies. We hypothesized that postnatal loss of SMC TBRII would cause aortopathy. APPROACH AND RESULTS: We generated mice with either of 2 tamoxifen-inducible SMC-specific Cre (SMC-CreER(T2)) alleles and homozygous floxed Tgfbr2 alleles. Mice were injected with tamoxifen, and their aortas examined 4 and 14 weeks later. Both SMC-CreER(T2) alleles efficiently and specifically rearranged a floxed reporter gene and efficiently rearranged a floxed Tgfbr2 allele, resulting in loss of aortic medial TBRII protein. Loss of SMC TBRII caused severe aortopathy, including hemorrhage, ulceration, dissection, dilation, accumulation of macrophage markers, elastolysis, abnormal proteoglycan accumulation, and aberrant SMC gene expression. All areas of the aorta were affected, with the most severe pathology in the ascending aorta. Cre-mediated loss of SMC TBRII in vitro ablated both canonical and noncanonical TGF-ß signaling and reproduced some of the gene expression abnormalities detected in vivo. CONCLUSIONS: SMC TBRII plays a critical role in maintaining postnatal aortic homeostasis. Loss of SMC TBRII disrupts TGF-ß signaling, acutely alters SMC gene expression, and rapidly results in severe and durable aortopathy. These results suggest that pharmacological blockade of TGF-ß signaling in humans could cause aortic disease rather than prevent it.


Subject(s)
Aortic Diseases/metabolism , Muscle, Smooth, Vascular/metabolism , Myocytes, Smooth Muscle/metabolism , Protein Serine-Threonine Kinases/deficiency , Receptors, Transforming Growth Factor beta/deficiency , Age Factors , Animals , Aorta/metabolism , Aorta/pathology , Aortic Diseases/genetics , Aortic Diseases/pathology , Cell Proliferation , Elastin/metabolism , Extracellular Matrix/metabolism , Gene Expression Regulation , Genetic Predisposition to Disease , Macrophages/metabolism , Macrophages/pathology , Mice, Knockout , Muscle, Smooth, Vascular/drug effects , Muscle, Smooth, Vascular/pathology , Myocytes, Smooth Muscle/drug effects , Myocytes, Smooth Muscle/pathology , Phenotype , Protein Serine-Threonine Kinases/genetics , RNA, Messenger/metabolism , Receptor, Transforming Growth Factor-beta Type II , Receptors, Transforming Growth Factor beta/genetics , Signal Transduction , Transforming Growth Factor beta1/pharmacology
7.
Neoplasia ; 15(5): 502-10, 2013 May.
Article in English | MEDLINE | ID: mdl-23633922

ABSTRACT

Chronic sympathetic nervous system activation results in increased angiogenesis and tumor growth in orthotopic mouse models of ovarian carcinoma. However, the mechanistic effects of such activation on the tumor vasculature are not well understood. Dopamine (DA), an inhibitory catecholamine, regulates the functions of normal and abnormal blood vessels. Here, we examined whether DA, an inhibitory catecholamine, could block the effects of chronic stress on tumor vasculature and tumor growth. Exogenous administration of DA not only decreased tumor microvessel density but also increased pericyte coverage of tumor vessels following daily restraint stress in mice. Daily restraint stress resulted in significantly increased tumor growth in the SKOV3ip1 and HeyA8 ovarian cancer models. DA treatment blocked stress-mediated increases in tumor growth and increased pericyte coverage of tumor endothelial cells. Whereas the antiangiogenic effect of DA is mediated by dopamine receptor 2 (DR2), our data indicate that DA, through DR1, stimulates vessel stabilization by increasing pericyte recruitment to tumor endothelial cells. DA significantly stimulated migration of mouse 10T1/2 pericyte-like cells in vitro and increased cyclic adenosine mono-phosphate (cAMP) levels in these cells. Moreover, DA or the DR1 agonist SKF 82958 increased platinum concentration in SKOV3ip1 tumor xenografts following cisplatin administration. In conclusion, DA stabilizes tumor blood vessels through activation of pericyte cAMP-protein kinase A signaling pathway by DR1. These findings could have implications for blocking the stimulatory effects of chronic stress on tumor growth.


Subject(s)
Angiogenesis Inhibitors/pharmacology , Dopamine/pharmacology , Neovascularization, Pathologic/drug therapy , Ovarian Neoplasms/drug therapy , Animals , Antineoplastic Agents/pharmacokinetics , Antineoplastic Agents/pharmacology , Benzazepines/pharmacology , Catecholamines/pharmacology , Catecholamines/physiology , Cell Line, Tumor , Cisplatin/pharmacokinetics , Cisplatin/pharmacology , Dopamine/physiology , Dopamine Agents/pharmacology , Drug Synergism , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Female , Humans , Mice , Mice, Nude , Neovascularization, Pathologic/metabolism , Ovarian Neoplasms/blood supply , Ovarian Neoplasms/metabolism , Pericytes/drug effects , Pericytes/metabolism , Receptors, Adrenergic, beta/metabolism , Receptors, Dopamine/metabolism , Second Messenger Systems , Stress, Physiological , Xenograft Model Antitumor Assays
8.
Nat Commun ; 4: 1403, 2013.
Article in English | MEDLINE | ID: mdl-23360994

ABSTRACT

Noradrenaline can modulate multiple cellular functions important for cancer progression; however, how this single extracellular signal regulates such a broad array of cellular processes is unknown. Here we identify Src as a key regulator of phosphoproteomic signalling networks activated in response to beta-adrenergic signalling in cancer cells. These results also identify a new mechanism of Src phosphorylation that mediates beta-adrenergic/PKA regulation of downstream networks, thereby enhancing tumour cell migration, invasion and growth. In human ovarian cancer samples, high tumoural noradrenaline levels were correlated with high pSrc(Y419) levels. Moreover, among cancer patients, the use of beta blockers was significantly associated with reduced cancer-related mortality. Collectively, these data provide a pivotal molecular target for disrupting neural signalling in the tumour microenvironment.


Subject(s)
Ovarian Neoplasms/enzymology , Ovarian Neoplasms/pathology , Receptors, Adrenergic, beta/metabolism , src-Family Kinases/metabolism , Adrenergic beta-Antagonists/pharmacology , Adrenergic beta-Antagonists/therapeutic use , Animals , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Cyclic AMP/metabolism , Cyclic AMP-Dependent Protein Kinases/metabolism , Enzyme Activation/drug effects , Female , Humans , Mice , Models, Molecular , Neoplasm Invasiveness , Neoplasm Metastasis , Norepinephrine/pharmacology , Ovarian Neoplasms/drug therapy , Ovarian Neoplasms/mortality , Phosphorylation/drug effects , Phosphoserine/metabolism , Signal Transduction/drug effects , Stress, Physiological/drug effects , Survival Analysis , Tyrosine/metabolism , src-Family Kinases/chemistry
9.
Clin Cancer Res ; 17(11): 3649-59, 2011 Jun 01.
Article in English | MEDLINE | ID: mdl-21531818

ABSTRACT

PURPOSE: Increased adrenergic activity in response to chronic stress is known to promote tumor growth by stimulating the tumor microenvironment. The focus of the current study was to determine whether dopamine, an inhibitory catecholamine, could block the effects of chronic stress on tumor growth. EXPERIMENTAL DESIGN: Expression of dopamine receptors (DR1-DR5) was analyzed by reverse transcriptase-PCR and by Western blotting. In vitro effects of dopamine on cell viability, apoptosis, and migration were examined. For in vivo therapy, murine and human DR2-siRNAs were incorporated into chitosan nanoparticles (CH-NP). RESULTS: In this model of chronic stress, tumoral norepinephrine levels remained elevated whereas dopamine levels were significantly decreased compared with nonstressed animals. Daily restraint stress resulted in significantly increased tumor growth in both immunodeficient (SKOV3ip1 and HeyA8) and immunocompetent (ID8) ovarian cancer models. This increase was completely blocked with daily dopamine treatment. Dopamine treatment also blocked the stress-induced increase in angiogenesis. Endothelial and ovarian cancer cells expressed all dopamine receptors except for the lack of DR3 expression in ovarian cancer cells. DR2 was responsible for the inhibitory effects of dopamine on tumor growth and microvessel density as well as the stimulatory effect on apoptosis, as the DR2 antagonist eticlopride reversed these effects. Dopamine significantly inhibited cell viability and stimulated apoptosis in vitro. Moreover, dopamine reduced cyclic AMP levels and inhibited norepinephrine and vascular permeability factor/VEGF-induced Src kinase activation. CONCLUSIONS: Dopamine depletion under chronic stress conditions creates a permissive microenvironment for tumor growth that can be reversed by dopamine replacement.


Subject(s)
Dopamine/pharmacology , Ovarian Neoplasms/pathology , Stress, Physiological/drug effects , Animals , Cell Line, Tumor , Cell Proliferation/drug effects , Dopamine/biosynthesis , Female , Humans , Mice , Mice, Inbred C57BL , Mice, Nude , Neovascularization, Pathologic , Ovarian Neoplasms/blood supply , Ovarian Neoplasms/metabolism , Receptors, Dopamine/biosynthesis , Receptors, Dopamine/genetics , Tumor Microenvironment
10.
J Clin Invest ; 120(5): 1515-23, 2010 May.
Article in English | MEDLINE | ID: mdl-20389021

ABSTRACT

Chronic stress is associated with hormonal changes that are known to affect multiple systems, including the immune and endocrine systems, but the effects of stress on cancer growth and progression are not fully understood. Here, we demonstrate that human ovarian cancer cells exposed to either norepinephrine or epinephrine exhibit lower levels of anoikis, the process by which cells enter apoptosis when separated from ECM and neighboring cells. In an orthotopic mouse model of human ovarian cancer, restraint stress and the associated increases in norepinephrine and epinephrine protected the tumor cells from anoikis and promoted their growth by activating focal adhesion kinase (FAK). These effects involved phosphorylation of FAKY397, which was itself associated with actin-dependent Src interaction with membrane-associated FAK. Importantly, in human ovarian cancer patients, behavioral states related to greater adrenergic activity were associated with higher levels of pFAKY397, which was in turn linked to substantially accelerated mortality. These data suggest that FAK modulation by stress hormones, especially norepinephrine and epinephrine, can contribute to tumor progression in patients with ovarian cancer and may point to potential new therapeutic targets for cancer management.


Subject(s)
Adrenergic Agents/metabolism , Anoikis , Focal Adhesion Protein-Tyrosine Kinases/metabolism , Ovarian Neoplasms/metabolism , Actins/metabolism , Animals , Cell Line, Tumor , Cell Proliferation , Disease Models, Animal , Epinephrine/metabolism , Female , Humans , Mice , Mice, Nude , Norepinephrine/metabolism , Phosphorylation
11.
Cancer Biol Ther ; 9(3): 176-82, 2010 Feb.
Article in English | MEDLINE | ID: mdl-20009575

ABSTRACT

PURPOSE: On the basis of the known role of platelet-derived growth factor (PDGF)-BB/PDGF receptor (PDGFR) beta in pericyte regulation, highly specific inhibitors of this target are needed. We tested the efficacy of a highly selective aptamer against PDGF-B with or without anti-VEGF therapy in ovarian cancer models. RESULTS: Bevacizumab inhibited tumor growth by 45% and 48% in the HeyA8 and SKOV3ip1 models, respectively. AX102 had minimal effect on the HeyA8 model, but increased tumor growth in the SKOV3ip1 model. However, bevacizumab plus AX102 was more effective than bevacizumab alone, and resulted in 76-88% inhibition of tumor growth in both models. A longitudinal study in the HeyA8 model using bioluminescence imaging showed that combination of bevacizumab, AX102 and paclitaxel caused tumor reduction by 65% (based on bioluminescence imaging). In the HeyA8 model, MVD and PCNA counts were significantly reduced in the bevacizumab treatment groups, and pericyte coverage was significantly decreased in the AX102 treatment groups. In the SKOV3ip1 model, MVD and PCNA was significantly reduced in the bevacizumab treatment group, and even lower in the bevacizumab and AX102 combination treatment group. EXPERIMENTAL DESIGN: The therapeutic efficacy of targeting endothelial cells (bevacizumab) and/or pericytes (PDGF-aptamer, AX102) was examined using HeyA8 and SKOV3ip1 orthotopic models of ovarian cancer metastasis. Following therapy, tumors were examined for microvessel density (MVD), proliferating cell nuclear antigen (PCNA) and vascular maturation (pericyte coverage). CONCLUSIONS: Dual targeting of endothelial cells and pericytes holds potential as an anti-vascular therapeutic approach in ovarian carcinoma.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Aptamers, Nucleotide/therapeutic use , Neovascularization, Pathologic/drug therapy , Ovarian Neoplasms/blood supply , Pericytes/drug effects , Proto-Oncogene Proteins c-sis/antagonists & inhibitors , Angiogenesis Inhibitors/therapeutic use , Animals , Antibodies, Monoclonal/therapeutic use , Antibodies, Monoclonal, Humanized , Bevacizumab , Combined Modality Therapy , Endothelium, Vascular/drug effects , Female , Fluorescent Antibody Technique , Humans , Immunoenzyme Techniques , Longitudinal Studies , Mice , Mice, Nude , Neovascularization, Pathologic/metabolism , Neovascularization, Pathologic/pathology , Ovarian Neoplasms/metabolism , Ovarian Neoplasms/pathology , Paclitaxel/administration & dosage , Pericytes/metabolism , Tumor Cells, Cultured , Vascular Endothelial Growth Factor A/antagonists & inhibitors , Xenograft Model Antitumor Assays
SELECTION OF CITATIONS
SEARCH DETAIL