Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26
Filter
1.
Biomedicines ; 12(5)2024 May 07.
Article in English | MEDLINE | ID: mdl-38790986

ABSTRACT

The poor prognosis for pancreatic ductal adenocarcinoma (PDAC) patients is due in part to the highly fibrotic nature of the tumors that impedes delivery of therapeutics, including nanoparticles (NPs). Our prior studies demonstrated that proglumide, a cholecystokinin receptor (CCKR) antagonist, reduced fibrosis pervading PanIN lesions in mice. Here, we further detail how the reduced fibrosis elicited by proglumide achieves the normalization of the desmoplastic tumor microenvironment (TME) and improves nanoparticle uptake. One week following the orthotopic injection of PDAC cells, mice were randomized to normal or proglumide-treated water for 3-6 weeks. Tumors were analyzed ex vivo for fibrosis, vascularity, stellate cell activation, vascular patency, and nanoparticle distribution. The histological staining and three-dimensional imaging of tumors each indicated a reduction in stromal collagen in proglumide-treated mice. Proglumide treatment increased tumor vascularity and decreased the activation of cancer-associated fibroblasts (CAFs). Additionally, PANC-1 cells with the shRNA-mediated knockdown of the CCK2 receptor showed an even greater reduction in collagen, indicating the CCK2 receptors on tumor cells contribute to the desmoplastic TME. Proglumide-mediated reduction in fibrosis also led to functional changes in the TME as evidenced by the enhanced intra-tumoral distribution of small (<12 nm) Rhodamine-loaded nanoparticles. The documented in vivo, tumor cell-intrinsic anti-fibrotic effects of CCK2R blockade in both an immunocompetent syngeneic murine PDAC model as well as a human PDAC xenograft model demonstrates that CCK2R antagonists, such as proglumide, can improve the delivery of nano-encapsulated therapeutics or imaging agents to pancreatic tumors.

2.
Front Neurosci ; 13: 837, 2019.
Article in English | MEDLINE | ID: mdl-31507355

ABSTRACT

PDE9A is a cGMP-specific phosphodiesterase expressed in neurons throughout the brain that has attracted attention as a therapeutic target to treat cognitive disorders. Indeed, PDE9A inhibitors are under evaluation in clinical trials as a treatment for Alzheimer's disease and schizophrenia. However, little is known about the cGMP signaling cascades regulated by PDE9A. Canonical cGMP signaling in brain follows the activation of neuronal nitric oxide synthase (nNOS) and the generation of nitric oxide, which activates soluble guanylyl cyclase and cGMP synthesis. However, we show that in mice, PDE9A regulates a pool of cGMP that is independent of nNOS, specifically, and nitric oxide signaling in general. This PDE9A-regulated cGMP pool appears to be highly compartmentalized and independent of cGMP pools regulated by several PDEs. These findings provide a new foundation for study of the upstream and downstream signaling elements regulated by PDE9A and its potential as a therapeutic target for brain disease.

3.
Biomedicines ; 6(2)2018 Jun 02.
Article in English | MEDLINE | ID: mdl-29865257

ABSTRACT

It is estimated that early detection of pancreatic ductal adenocarcinoma (PDAC) could increase long-term patient survival by as much as 30% to 40% (Seufferlein, T. et al., Nat. Rev. Gastroenterol. Hepatol.2016, 13, 74⁻75). There is an unmet need for reagents that can reliably identify early cancerous or precancerous lesions through various imaging modalities or could be employed to deliver anticancer treatments specifically to tumor cells. However, to date, many PDAC tumor-targeting strategies lack selectivity and are unable to discriminate between tumor and nontumor cells, causing off-target effects or unclear diagnoses. Although a variety of approaches have been taken to identify tumor-targeting reagents that can effectively direct therapeutics or imaging agents to cancer cells (Liu, D. et al., J. Controlled Release2015, 219, 632⁻643), translating these reagents into clinical practice has been limited, and it remains an area open to new methodologies and reagents (O'Connor, J.P. et al., Nat. Rev. Clin. Oncol. 2017, 14, 169⁻186). G protein⁻coupled receptors (GPCRs), which are key target proteins for drug discovery and comprise a large proportion of currently marketed therapeutics, hold significant promise for tumor imaging and targeted treatment, particularly for pancreatic cancer.

5.
J Med Chem ; 61(3): 1001-1018, 2018 02 08.
Article in English | MEDLINE | ID: mdl-29293004

ABSTRACT

Computational modeling was used to direct the synthesis of analogs of previously reported phosphodiesterase 2A (PDE2A) inhibitor 1 with an imidazotriazine core to yield compounds of significantly enhanced potency. The analog PF-05180999 (30) was subsequently identified as a preclinical candidate targeting cognitive impairment associated with schizophrenia. Compound 30 demonstrated potent binding to PDE2A in brain tissue, dose responsive mouse brain cGMP increases, and reversal of N-methyl-d-aspartate (NMDA) antagonist-induced (MK-801, ketamine) effects in electrophysiology and working memory models in rats. Preclinical pharmacokinetics revealed unbound brain/unbound plasma levels approaching unity and good oral bioavailability resulting in an average concentration at steady state (Cav,ss) predicted human dose of 30 mg once daily (q.d.). Modeling of a modified release formulation suggested that 25 mg twice daily (b.i.d.) could maintain plasma levels of 30 at or above targeted efficacious plasma levels for 24 h, which became part of the human clinical plan.


Subject(s)
Brain/drug effects , Brain/metabolism , Cyclic Nucleotide Phosphodiesterases, Type 2/antagonists & inhibitors , Drug Discovery , Enzyme Inhibitors/metabolism , Enzyme Inhibitors/pharmacology , Animals , Biological Availability , Brain/physiology , Cyclic Nucleotide Phosphodiesterases, Type 2/chemistry , Cyclic Nucleotide Phosphodiesterases, Type 2/metabolism , Dose-Response Relationship, Drug , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacokinetics , Humans , Imidazoles/chemistry , Imidazoles/metabolism , Imidazoles/pharmacokinetics , Imidazoles/pharmacology , Inhibitory Concentration 50 , Memory, Short-Term/drug effects , Molecular Docking Simulation , Protein Conformation
6.
J Med Chem ; 60(13): 5673-5698, 2017 07 13.
Article in English | MEDLINE | ID: mdl-28574706

ABSTRACT

Phosphodiesterase 2A (PDE2A) inhibitors have been reported to demonstrate in vivo activity in preclinical models of cognition. To more fully explore the biology of PDE2A inhibition, we sought to identify potent PDE2A inhibitors with improved brain penetration as compared to current literature compounds. Applying estimated human dose calculations while simultaneously leveraging synthetically enabled chemistry and structure-based drug design has resulted in a highly potent, selective, brain penetrant compound 71 (PF-05085727) that effects in vivo biochemical changes commensurate with PDE2A inhibition along with behavioral and electrophysiological reversal of the effects of NMDA antagonists in rodents. This data supports the ability of PDE2A inhibitors to potentiate NMDA signaling and their further development for clinical cognition indications.


Subject(s)
Cyclic Nucleotide Phosphodiesterases, Type 2/antagonists & inhibitors , Drug Design , Phosphodiesterase Inhibitors/chemistry , Phosphodiesterase Inhibitors/pharmacology , Animals , Brain/drug effects , Brain/metabolism , Crystallography, X-Ray , Cyclic Nucleotide Phosphodiesterases, Type 2/chemistry , Cyclic Nucleotide Phosphodiesterases, Type 2/metabolism , Dogs , Haplorhini , Humans , Mice , Molecular Docking Simulation , Phosphodiesterase Inhibitors/administration & dosage , Phosphodiesterase Inhibitors/pharmacokinetics , Rats
7.
Neuron ; 92(6): 1220-1237, 2016 Dec 21.
Article in English | MEDLINE | ID: mdl-27916455

ABSTRACT

Huntington's disease (HD) symptoms are driven to a large extent by dysfunction of the basal ganglia circuitry. HD patients exhibit reduced striatal phoshodiesterase 10 (PDE10) levels. Using HD mouse models that exhibit reduced PDE10, we demonstrate the benefit of pharmacologic PDE10 inhibition to acutely correct basal ganglia circuitry deficits. PDE10 inhibition restored corticostriatal input and boosted cortically driven indirect pathway activity. Cyclic nucleotide signaling is impaired in HD models, and PDE10 loss may represent a homeostatic adaptation to maintain signaling. Elevation of both cAMP and cGMP by PDE10 inhibition was required for rescue. Phosphoproteomic profiling of striatum in response to PDE10 inhibition highlighted plausible neural substrates responsible for the improvement. Early chronic PDE10 inhibition in Q175 mice showed improvements beyond those seen with acute administration after symptom onset, including partial reversal of striatal deregulated transcripts and the prevention of the emergence of HD neurophysiological deficits. VIDEO ABSTRACT.


Subject(s)
Cerebral Cortex/drug effects , Huntington Disease/physiopathology , Neostriatum/drug effects , Phosphodiesterase Inhibitors/pharmacology , Pyrazoles/pharmacology , Quinolines/pharmacology , Animals , Basal Ganglia/diagnostic imaging , Basal Ganglia/drug effects , Basal Ganglia/metabolism , Basal Ganglia/physiopathology , Cerebral Cortex/diagnostic imaging , Cerebral Cortex/metabolism , Cerebral Cortex/physiopathology , Cyclic AMP/metabolism , Cyclic GMP/metabolism , Disease Models, Animal , Huntington Disease/metabolism , Mice , Neostriatum/diagnostic imaging , Neostriatum/metabolism , Neostriatum/physiopathology , Phosphoric Diester Hydrolases , Positron-Emission Tomography , Subthalamic Nucleus/diagnostic imaging , Subthalamic Nucleus/drug effects , Subthalamic Nucleus/metabolism , Subthalamic Nucleus/physiopathology , Tritium
8.
J Neurosci ; 35(14): 5781-91, 2015 Apr 08.
Article in English | MEDLINE | ID: mdl-25855188

ABSTRACT

The striatum contains a rich variety of cyclic nucleotide phosphodiesterases (PDEs), which play a critical role in the regulation of cAMP and cGMP signaling. The dual-substrate enzyme PDE10A is the most highly expressed PDE in striatal medium-sized spiny neurons (MSNs) with low micromolar affinity for both cyclic nucleotides. Previously, we have shown that systemic and local administration of the selective PDE10A inhibitor TP-10 potently increased the responsiveness of MSNs to cortical stimulation. However, the signaling mechanisms underlying PDE10A inhibitor-induced changes in corticostriatal transmission are only partially understood. The current studies assessed the respective roles of cAMP and cGMP in the above effects using soluble guanylyl cyclase (sGC) or adenylate cyclase (AC) specific inhibitors. Cortically evoked spike activity was monitored in urethane-anesthetized rats using in vivo extracellular recordings performed proximal to a microdialysis probe during local infusion of vehicle, the selective sGC inhibitor ODQ, or the selective AC inhibitor SQ 22536. Systemic administration of TP-10 (3.2 mg/kg) robustly increased cortically evoked spike activity in a manner that was blocked following intrastriatal infusion of ODQ (50 µm). The effects of TP-10 on evoked activity were due to accumulation of cGMP, rather than cAMP, as the AC inhibitor SQ was without effect. Consistent with these observations, studies in neuronal NO synthase (nNOS) knock-out (KO) mice confirmed that PDE10A operates downstream of nNOS to limit cGMP production and excitatory corticostriatal transmission. Thus, stimulation of PDE10A acts to attenuate corticostriatal transmission in a manner largely dependent on effects directed at the NO-sGC-cGMP signaling cascade.


Subject(s)
Cerebral Cortex/cytology , Corpus Striatum/drug effects , Cyclic GMP/metabolism , Nitric Oxide Synthase Type I/metabolism , Phosphoric Diester Hydrolases/metabolism , Signal Transduction/physiology , Action Potentials/drug effects , Action Potentials/genetics , Animals , Biophysics , Corpus Striatum/cytology , Cyclic AMP/metabolism , Electric Stimulation , Enzyme Inhibitors/pharmacology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Microdialysis , Neural Pathways/drug effects , Neural Pathways/physiology , Neurons/drug effects , Nitric Oxide Synthase Type I/genetics , Rats , Rats, Sprague-Dawley
9.
J Pharmacol Exp Ther ; 341(2): 396-409, 2012 May.
Article in English | MEDLINE | ID: mdl-22328573

ABSTRACT

Cyclic nucleotides are critical regulators of synaptic plasticity and participate in requisite signaling cascades implicated across multiple neurotransmitter systems. Phosphodiesterase 9A (PDE9A) is a high-affinity, cGMP-specific enzyme widely expressed in the rodent central nervous system. In the current study, we observed neuronal staining with antibodies raised against PDE9A protein in human cortex, cerebellum, and subiculum. We have also developed several potent, selective, and brain-penetrant PDE9A inhibitors and used them to probe the function of PDE9A in vivo. Administration of these compounds to animals led to dose-dependent accumulation of cGMP in brain tissue and cerebrospinal fluid, producing a range of biological effects that implied functional significance for PDE9A-regulated cGMP in dopaminergic, cholinergic, and serotonergic neurotransmission and were consistent with the widespread distribution of PDE9A. In vivo effects of PDE9A inhibition included reversal of the respective disruptions of working memory by ketamine, episodic and spatial memory by scopolamine, and auditory gating by amphetamine, as well as potentiation of risperidone-induced improvements in sensorimotor gating and reversal of the stereotypic scratching response to the hallucinogenic 5-hydroxytryptamine 2A agonist mescaline. The results suggested a role for PDE9A in the regulation of monoaminergic circuitry associated with sensory processing and memory. Thus, PDE9A activity regulates neuronal cGMP signaling downstream of multiple neurotransmitter systems, and inhibition of PDE9A may provide therapeutic benefits in psychiatric and neurodegenerative diseases promoted by the dysfunction of these diverse neurotransmitter systems.


Subject(s)
3',5'-Cyclic-AMP Phosphodiesterases/antagonists & inhibitors , 3',5'-Cyclic-GMP Phosphodiesterases/metabolism , Cholinergic Agents/pharmacology , Cyclic GMP/metabolism , Neurons/drug effects , Neurons/metabolism , Phosphodiesterase Inhibitors/pharmacology , 3',5'-Cyclic-AMP Phosphodiesterases/genetics , 3',5'-Cyclic-AMP Phosphodiesterases/metabolism , Animals , Avoidance Learning/drug effects , Brain/drug effects , Brain/metabolism , Female , Humans , Macaca fascicularis , Male , Memory/drug effects , Mice , Mice, Inbred C57BL , Motor Activity/drug effects , Neurotransmitter Agents/pharmacology , Rats , Rats, Long-Evans , Rats, Wistar , Sensory Gating/drug effects , Stereotyped Behavior/drug effects , Synaptic Transmission/drug effects
10.
Cancer Biol Ther ; 13(3): 164-74, 2012 Feb 01.
Article in English | MEDLINE | ID: mdl-22277584

ABSTRACT

There currently are no tests available for early diagnosis or for the identification of patients at risk for development of pancreatic cancer. We report the discovery of single nucleotide polymorphism (SNP) in the cholecystokinin B receptor (CCKBR) gene predicts survival and risk of pancreatic cancer. Growth of human pancreatic cancer is stimulated by gastrin through the CCKBR and an alternatively spliced isoform of the CCKBR gene called CCKCR. One hundred and ten surgically resected benign and malignant pancreatic tissues as well as normal pancreas were prospectively evaluated for CCKBR genotype and protein expression. Analysis demonstrated the expression of the spliced isoform, CCKCR, was associated with a (SNP) (C > A) at position 32 of the intron 4 (IVS 4) of the CCKBR gene. Since the SNP is within an intron, it has not previously been identified in the GWAS studies. Only patients with the A/A or A/C genotypes, exhibited immunoreactivity to a selective CCKCR antibody. Survival among pancreatic cancer patients with the A-SNP was significantly shorter (p = 0.0001, hazard ratio = 3.63) compared with individuals with C/C genotype. Other variables such as surgical margins, lymph node status, histologic grade or adjuvant chemotherapy were not associated with survival. Furthermore, having one or two of the A-alleles was found to increase the risk of pancreatic adenocarcinoma by 174% (p = 0.0192) compared with the C/C wild type. Cancer cells transfected to overexpress the CCKCR demonstrated increased proliferation over controls. Genetic screening for this SNP may aid in early detection of pancreatic cancer in high risk subjects.


Subject(s)
Carcinoma, Pancreatic Ductal/genetics , Pancreatic Neoplasms/genetics , Receptor, Cholecystokinin B/genetics , Aged , Antibodies, Monoclonal, Murine-Derived/chemistry , Antibody Specificity , Carcinoma, Pancreatic Ductal/metabolism , Carcinoma, Pancreatic Ductal/mortality , Carcinoma, Pancreatic Ductal/pathology , Case-Control Studies , Cell Line, Tumor , Cell Proliferation , Female , Genome-Wide Association Study , Genotype , Humans , Kaplan-Meier Estimate , Male , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/mortality , Pancreatic Neoplasms/pathology , Polymorphism, Single Nucleotide , Protein Isoforms/genetics , Protein Isoforms/immunology , Protein Isoforms/metabolism , Receptor, Cholecystokinin B/immunology , Receptor, Cholecystokinin B/metabolism , Risk Factors
11.
J Pharmacol Exp Ther ; 336(1): 64-76, 2011 Jan.
Article in English | MEDLINE | ID: mdl-20923867

ABSTRACT

Inhibition of phosphodiesterase 10A (PDE10A) promotes cyclic nucleotide signaling, increases striatal activation, and decreases behavioral activity. Enhanced cyclic nucleotide signaling is a well established route to producing changes in gene expression. We hypothesized that chronic suppression of PDE10A activity would have significant effects on gene expression in the striatum. A comparison of the expression profile of PDE10A knockout (KO) mice and wild-type mice after chronic PDE10A inhibition revealed altered expression of 19 overlapping genes with few significant changes outside the striatum or after administration of a PDE10A inhibitor to KO animals. Chronic inhibition of PDE10A produced up-regulation of mRNAs encoding genes that included prodynorphin, synaptotagmin10, phosphodiesterase 1C, glutamate decarboxylase 1, and diacylglycerol O-acyltransferase and a down-regulation of mRNAs encoding choline acetyltransferase and Kv1.6, suggesting long-term suppression of the PDE10A enzyme is consistent with altered striatal excitability and potential utility as a antipsychotic therapy. In addition, up-regulation of mRNAs encoding histone 3 (H3) and down-regulation of histone deacetylase 4, follistatin, and claspin mRNAs suggests activation of molecular cascades capable of neuroprotection. We used lentiviral delivery of cAMP response element (CRE)-luciferase reporter constructs into the striatum and live animal imaging of 2-{4-[-pyridin-4-yl-1-(2,2,2-trifluoro-ethyl)-1H-pyrazol-3-yl]-phenoxymethyl}-quinoline succinic acid (TP-10)-induced luciferase activity to further demonstrate PDE10 inhibition results in CRE-mediated transcription. Consistent with potential neuroprotective cascades, we also demonstrate phosphorylation of mitogen- and stress-activated kinase 1 and H3 in vivo after TP-10 treatment. The observed changes in signaling and gene expression are predicted to provide neuroprotective effects in models of Huntington's disease.


Subject(s)
Corpus Striatum/enzymology , Huntington Disease/drug therapy , Huntington Disease/enzymology , Neurotransmitter Agents/antagonists & inhibitors , Phosphodiesterase Inhibitors/pharmacology , Phosphoric Diester Hydrolases/metabolism , Signal Transduction/drug effects , Animals , Corpus Striatum/drug effects , Corpus Striatum/metabolism , Disease Models, Animal , Gene Expression Profiling/methods , Gene Expression Regulation/drug effects , Huntington Disease/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Neuroprotective Agents/pharmacology , Neuroprotective Agents/therapeutic use , Neurotransmitter Agents/biosynthesis , Neurotransmitter Agents/genetics , Phosphodiesterase Inhibitors/therapeutic use , Phosphoric Diester Hydrolases/deficiency , Phosphoric Diester Hydrolases/genetics , Pyrazoles/pharmacology , Pyrazoles/therapeutic use , Quinolines/pharmacology , Quinolines/therapeutic use , Signal Transduction/genetics
12.
Int J Oncol ; 38(3): 593-601, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21186400

ABSTRACT

Cholecystokinin (CCK) and gastrin stimulate growth of pancreatic cancer. Although down-regulation of gastrin inhibits growth of pancreatic cancer, the contribution of endogenous CCK to tumor growth is unknown. The purpose of this study was to evaluate the role of endogenous CCK on autocrine growth of pancreatic cancer. Pancreatic cancer cell lines were analyzed for CCK mRNA and peptide expression by real-time RT-PCR and radioimmunoassay, respectively. The effect of endogenous CCK on growth was evaluated by treating cancer cells with CCK neutralizing antibodies and by down-regulating CCK mRNA by RNAi. Wild-type pancreatic cancer cells expressed significantly lower CCK mRNA and peptide levels than gastrin. Neither treatment of pancreatic cancer cells with CCK antibodies nor the down-regulation of CCK mRNA and peptide by shRNAs altered growth in vitro or in vivo. Conversely, when gastrin mRNA expression was down-regulated, the same cells failed to produce tumors in spite of having sustained levels of endogenous CCK. Pancreatic cancer cells produce CCK and gastrin; however, the autocrine production of gastrin is more important for stimulating tumor growth.


Subject(s)
Adenocarcinoma/pathology , Cell Proliferation , Cholecystokinin/physiology , Pancreatic Neoplasms/pathology , Adenocarcinoma/genetics , Adenocarcinoma/metabolism , Animals , Antibodies, Neutralizing/pharmacology , Autocrine Communication/drug effects , Autocrine Communication/genetics , Autocrine Communication/physiology , Cell Line, Tumor , Cell Proliferation/drug effects , Cholecystokinin/genetics , Cholecystokinin/immunology , Cholecystokinin/metabolism , Gastrins/analysis , Gastrins/genetics , Gastrins/metabolism , Gastrins/pharmacology , Gene Expression Regulation, Neoplastic/drug effects , Humans , Male , Mice , Mice, Nude , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/metabolism , RNA, Messenger/analysis , RNA, Messenger/metabolism , RNA, Small Interfering/pharmacology , Tissue Distribution , Xenograft Model Antitumor Assays
13.
J Med Chem ; 52(16): 5188-96, 2009 Aug 27.
Article in English | MEDLINE | ID: mdl-19630403

ABSTRACT

By utilizing structure-based drug design (SBDD) knowledge, a novel class of phosphodiesterase (PDE) 10A inhibitors was identified. The structure-based drug design efforts identified a unique "selectivity pocket" for PDE10A inhibitors, and interactions within this pocket allowed the design of highly selective and potent PDE10A inhibitors. Further optimization of brain penetration and drug-like properties led to the discovery of 2-[4-(1-methyl-4-pyridin-4-yl-1H-pyrazol-3-yl)-phenoxymethyl]-quinoline (PF-2545920). This PDE10A inhibitor is the first reported clinical entry for this mechanism in the treatment of schizophrenia.


Subject(s)
Antipsychotic Agents/chemical synthesis , Models, Molecular , Phosphoric Diester Hydrolases/metabolism , Pyrazoles/chemical synthesis , Quinolines/chemical synthesis , Schizophrenia/drug therapy , Animals , Antipsychotic Agents/pharmacokinetics , Antipsychotic Agents/pharmacology , Avoidance Learning/drug effects , Binding Sites , Brain/metabolism , Crystallography, X-Ray , Dogs , Female , Humans , Hydrogen Bonding , In Vitro Techniques , Macaca fascicularis , Male , Mice , Mice, Knockout , Microsomes, Liver/metabolism , Molecular Structure , Phosphoric Diester Hydrolases/chemistry , Phosphoric Diester Hydrolases/genetics , Protein Binding , Pyrazoles/pharmacokinetics , Pyrazoles/pharmacology , Quinolines/pharmacokinetics , Quinolines/pharmacology , Rats , Rats, Sprague-Dawley , Structure-Activity Relationship
14.
Pancreas ; 38(5): e151-61, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19465883

ABSTRACT

OBJECTIVES: This study evaluated the effects of gastrin messenger RNA (mRNA) down-regulation on growth of human pancreatic cancer. METHODS: Gastrin expression was examined in human pancreatic cancer cell lines by reverse transcriptase-polymerase chain reaction, and peptide expression was assessed by immunocytochemistry. Gastrin was down-regulated using either stable transfection of an antisense gastrin cDNA or 1 of 3 shRNA (short hairpin RNA) constructs. Tumor formation was evaluated after either subcutaneous or orthotopic injections into nude mice. The effect of nanoliposomes loaded with gastrin siRNA (small interfering RNA) was tested in mice bearing pancreatic tumors. RESULTS: Stable transfection of gastrin antisense or shRNAs into BxPC-3 cells resulted in clones with more than 90% reduction in gastrin mRNA. Tumor growth rate and incidence of metastases in both wild-type and transfected pancreatic cancer cells were directly proportional to the degrees of gastrin mRNA expression. Immunofluorescence analysis confirmed that gastrin peptide levels were decreased in antisense and shRNA tumors. Gastrin knockdown clones had lower Ki-67 and increased cleaved caspase-3 staining, consistent with known effects of gastrin on proliferation and apoptosis. Tumors in mice treated with gastrin siRNA were smaller than controls. CONCLUSIONS: These results suggest that RNAi targeting of gastrin could serve as an effective treatment for pancreatic cancer.


Subject(s)
Cell Proliferation , Gastrins/genetics , Pancreatic Neoplasms/pathology , RNA Interference , Animals , Cell Line, Tumor , Chromatography, High Pressure Liquid , Fluorescent Antibody Technique , Gastrins/metabolism , Humans , Ki-67 Antigen/analysis , Liposomes , Mass Spectrometry , Mice , Mice, Nude , Nanotechnology , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/therapy , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Small Interfering/genetics , Reverse Transcriptase Polymerase Chain Reaction , Transfection , Tumor Burden , Xenograft Model Antitumor Assays
15.
J Immunotoxicol ; 5(2): 179-87, 2008 Apr.
Article in English | MEDLINE | ID: mdl-18569389

ABSTRACT

Inflammatory bowel disease (IBD) is a condition of the intestine with significant morbidity. Although hereditary, environmental, immunologic, and bacterial factors have been implicated, the etiology of IBD remains unknown. Since opioid peptides modulate inflammatory cytokine production and opioid antagonists promote tissue growth and repair, we hypothesized the opioid antagonist naltrexone could reduce inflammation of the bowel. Using a chemically-induced mouse model of IBD, C57BL/6J mice received either untreated drinking water or water containing 2% dextran sulfate sodium (DSS) in two parallel regimens modeling moderate and severe colitis. After colitis was established, animals in the moderate colitis study were administered either saline (control) or naltrexone (NTX; 8 or 400 microg/kg) daily, while those in the severe colitis study received 0.1 or 10 mg/kg NTX. DSS-treated animals had significant weight loss (p = 0.006) and higher disease activity index (DAI) scores (p < 0.001) compared to water controls. However, NTX treatment of mice with moderate colitis resulted in less weight loss, lower DAI scores, and less histologic evidence of inflammation compared to controls. Significantly, elevated levels of colonic RNA for pro-inflammatory cytokines interleukin (IL)-6 and IL-12 were also decreased toward normal with NTX. Similar to patients with severe and unresponsive disease, animals in the severe colitis study did not significantly respond to treatment. Thus, NTX therapy reverses physical symptoms, histologic evidence, and molecular markers of inflammation in moderate colitis. The mechanism by which NTX acts to reverse colitis is related in part to the decreased expression of pro-inflammatory cytokines.


Subject(s)
Inflammation Mediators/immunology , Inflammatory Bowel Diseases/drug therapy , Naltrexone/pharmacology , Narcotic Antagonists/pharmacology , Animals , Biomarkers , Dextran Sulfate/toxicity , Disease Models, Animal , Gene Expression Regulation/drug effects , Gene Expression Regulation/immunology , Humans , Inflammatory Bowel Diseases/chemically induced , Inflammatory Bowel Diseases/immunology , Inflammatory Bowel Diseases/pathology , Interleukin-12/immunology , Interleukin-6/immunology , Male , Mice , Weight Loss/drug effects
16.
Regul Pept ; 151(1-3): 35-42, 2008 Nov 29.
Article in English | MEDLINE | ID: mdl-18499282

ABSTRACT

We have previously demonstrated that inactivation of capsaicin-sensitive sensory neurons enhances lung and heart metastases of breast carcinoma. Because a significant part of sensory innervation of lung tissue is supplied by the vagus nerve, we here examined the effects of unilateral mid-cervical vagotomy in the metastases of 4THMpc breast carcinoma and tissue Substance P (SP) levels. Balb-c mice were injected orthotopically with 4THMpc cells 1 week after vagotomy. Animals were sacrificed 27-30 days after injection of 4THMpc cells and the extent of metastases was determined. Unilateral vagotomy, right or left significantly increased the lung, liver and kidney metastases without altering the growth rate of the primary tumor. Heart metastases were increased only following left vagotomy. The changes in SP levels were somewhat surprising such that vagotomy actually increased while sham-operation decreased SP levels in lung. The effect of sham-operation was reversed by unilateral vagotomy demonstrating that vagal activity decreases total SP levels in the lung. Increased SP levels might be due to decreased degradation of the peptide. Presence of the tumor markedly increased SP level in the lung, which was more prominent in vagotomized animals. These results provide evidence that vagal activity may protect against metastatic disease.


Subject(s)
Mammary Neoplasms, Experimental/metabolism , Mammary Neoplasms, Experimental/secondary , Substance P/metabolism , Vagus Nerve/physiopathology , Animals , Cell Line, Tumor , Female , Heart Neoplasms/secondary , Kidney Neoplasms/secondary , Liver Neoplasms, Experimental/secondary , Lung/metabolism , Lung Neoplasms/secondary , Mammary Neoplasms, Experimental/pathology , Mammary Neoplasms, Experimental/physiopathology , Mice , Mice, Inbred BALB C , Vagotomy
17.
Neuropharmacology ; 54(2): 417-27, 2008 Feb.
Article in English | MEDLINE | ID: mdl-18061215

ABSTRACT

The phenotype of genetically modified animals is strongly influenced by both the genetic background of the animal as well as environmental factors. We have previously reported the behavioral and neurochemical characterization of PDE10A knockout mice maintained on a DBA1LacJ (PDE10A(DBA)) genetic background. The aim of the present studies was to assess the behavioral and neurochemical phenotype of PDE10A knockout mice on an alternative congenic C57BL/6N (PDE10A(C57)) genetic background. Consistent with our previous results, PDE10A(C57) knockout mice showed a decrease in exploratory locomotor activity and a delay in the acquisition of conditioned avoidance responding. Also consistent with previous studies, the elimination of PDE10A did not alter basal levels of striatal cGMP or cAMP or affect behavior in several other well-characterized behavioral assays. PDE10A(C57) knockout mice showed a blunted response to MK-801, although to a lesser degree than previously observed in the PDE10A(DBA) knockout mice, and no differences were observed following a PCP challenge. PDE10A(C57) knockout mice showed a significant change in striatal dopamine turnover, which was accompanied by an enhanced locomotor response to AMPH, These studies demonstrate that while many of the behavioral effects of the PDE10A gene deletion appear to be independent of genetic background, the impact of the deletion on behavior can vary in magnitude. Furthermore, the effects on the dopaminergic system appear to be background-dependent, with significant effects observed only in knockout mice on the C57BL6N genetic background.


Subject(s)
Behavior, Animal/physiology , Phosphoric Diester Hydrolases/genetics , Phosphoric Diester Hydrolases/physiology , Amphetamine/pharmacology , Animals , Anxiety/psychology , Avoidance Learning/drug effects , Avoidance Learning/physiology , Behavior, Animal/drug effects , Biogenic Monoamines/metabolism , Brain Chemistry/drug effects , Brain Chemistry/genetics , Chromatography, High Pressure Liquid , Depression/psychology , Dizocilpine Maleate/pharmacology , Dopamine/metabolism , Dopamine Uptake Inhibitors/pharmacology , Excitatory Amino Acid Antagonists/pharmacology , Hot Temperature , Methamphetamine/pharmacology , Mice , Mice, Inbred C57BL , Mice, Knockout , Motor Activity/drug effects , Nucleotides, Cyclic/metabolism , Pain Measurement/drug effects , Phencyclidine/pharmacology , Phosphoproteins/metabolism , Serotonin/metabolism , Swimming/psychology
18.
Neuropharmacology ; 51(2): 386-96, 2006 Aug.
Article in English | MEDLINE | ID: mdl-16780899

ABSTRACT

Phosphodiesterase 10A (PDE10A) is a recently identified cyclic nucleotide phosphodiesterase expressed primarily in dopaminoreceptive medium spiny neurons of the striatum. We report that papaverine is a potent, specific inhibitor of PDE10A and use this compound to explore the role of PDE10A in regulating striatal function. Papaverine administration produces an increase in striatal tissue levels of cGMP and an increase in extracellular cAMP measured by microdialysis. These cyclic nucleotide changes are accompanied by increases in the phosphorylation of CREB and ERK, downstream markers of neuronal activation. In rats, papaverine potentiates haloperidol-induced catalepsy, consistent with the hypothesis that inhibition of PDE10A can increase striatal output and prompting a further evaluation of papaverine in models predictive of antipsychotic activity. Papaverine is found to inhibit conditioned avoidance responding in rats and mice and to inhibit PCP- and amphetamine-stimulated locomotor activity in rats. The effects of papaverine on striatal cGMP and CREB and ERK phosphorylation, as well as on conditioned avoidance responding, were absent in PDE10A knockout mice, indicating that the effects of the compound are the result of PDE10A inhibition. These results indicate that PDE10A regulates the activation of striatal medium spiny neurons through effects on cAMP- and cGMP-dependent signaling cascades. Furthermore, the present results demonstrate that papaverine has efficacy in behavioral models predictive of antipsychotic activity. Thus, inhibition of PDE10A may represent a novel approach to the treatment of psychosis.


Subject(s)
Antipsychotic Agents/pharmacology , Corpus Striatum/enzymology , Papaverine/pharmacology , Phosphodiesterase Inhibitors/pharmacology , Phosphoric Diester Hydrolases/physiology , Animals , Avoidance Learning/drug effects , Catalepsy/chemically induced , Central Nervous System Stimulants/pharmacology , Corpus Striatum/metabolism , Cyclic AMP/metabolism , Cyclic AMP Response Element-Binding Protein/metabolism , Cyclic GMP/metabolism , Dendritic Spines/drug effects , Dendritic Spines/physiology , Dextroamphetamine/pharmacology , Drug Synergism , Excitatory Amino Acid Antagonists/pharmacology , Extracellular Signal-Regulated MAP Kinases/metabolism , Haloperidol/pharmacology , Mice , Mice, Knockout , Motor Activity/drug effects , Phencyclidine/pharmacology , Phosphorylation , Rats
19.
Clin Cancer Res ; 12(5): 1431-40, 2006 Mar 01.
Article in English | MEDLINE | ID: mdl-16533765

ABSTRACT

PURPOSE: In vivo studies have focused on the latter stages of the bone metastatic process (osteolysis), whereas little is known about earlier events, e.g., arrival, localization, and initial colonization. Defining these initial steps may potentially identify the critical points susceptible to therapeutic intervention. EXPERIMENTAL DESIGN: MDA-MB-435 human breast cancer cells engineered with green fluorescent protein were injected into the cardiac left ventricle of athymic mice. Femurs were analyzed by fluorescence microscopy, immunohistochemistry, real-time PCR, flow cytometry, and histomorphometry at times ranging from 1 hour to 6 weeks. RESULTS: Single cells were found in distal metaphyses at 1 hour postinjection and remained as single cells up to 72 hours. Diaphyseal arrest occurred rarely and few cells remained there after 24 hours. At 1 week, numerous foci (2-10 cells) were observed, mostly adjacent to osteoblast-like cells. By 2 weeks, fewer but larger foci (> or =50 cells) were seen. Most bones had a single large mass at 4 weeks (originating from a colony or coalescing foci) which extended into the diaphysis by 4 to 6 weeks. Little change (<20%) in osteoblast or osteoclast numbers was observed at 2 weeks, but at 4 to 6 weeks, osteoblasts were dramatically reduced (8% of control), whereas osteoclasts were reduced modestly (to approximately 60% of control). CONCLUSIONS: Early arrest in metaphysis and minimal retention in diaphysis highlight the importance of the local milieu in determining metastatic potential. These results extend the Seed and Soil hypothesis by demonstrating both intertissue and intratissue differences governing metastatic location. Ours is the first in vivo evidence that tumor cells influence not only osteoclasts, as widely believed, but also eliminate functional osteoblasts, thereby restructuring the bone microenvironment to favor osteolysis. The data may also explain why patients receiving bisphosphonates fail to heal bone despite inhibiting resorption, implying that concurrent strategies that restore osteoblast function are needed to effectively treat osteolytic bone metastases.


Subject(s)
Bone Neoplasms/secondary , Breast Neoplasms/pathology , Animals , Cell Line, Tumor/transplantation , Female , Femur/pathology , Flow Cytometry , Green Fluorescent Proteins/genetics , Humans , Immunohistochemistry , Kinetics , Mice , Mice, Nude , Microscopy, Fluorescence , Osteoblasts/pathology , Reverse Transcriptase Polymerase Chain Reaction
20.
Cancer Res ; 65(16): 7370-7, 2005 Aug 15.
Article in English | MEDLINE | ID: mdl-16103089

ABSTRACT

Skeletal metastases occur with high incidence in patients with breast cancer and cause long-term skeletal morbidity. Osteonectin (SPARC, BM-40) is a bone matrix factor that is an in vitro chemoattractant for breast and prostate cancer cells. Increased expression of osteonectin is found in malignant breast tumors. We infected MDA-231 breast cancer cells with an adenovirus expressing osteonectin to examine the role of osteonectin expression in breast cancer cells and its effect on metastasis, in particular to bone. Expression of osteonectin did not affect MDA-231 cell proliferation, apoptosis, migration, cell aggregation, or protease cleavage of collagen IV. However, in vitro invasion of these osteonectin-infected cells through Matrigel and colony formation on Matrigel was decreased. Interestingly, high osteonectin expression in MDA-231 cells inhibited metastasis in a dose-dependent manner to many different organs including bone. The reduction in metastasis may be due to decreased platelet-tumor cell aggregation, because exogenous osteonectin inhibited platelet aggregation in vitro and the high osteonectin expression in MDA-231 cells reduced tumor cell-induced thrombocytopenia in vivo compared with control-infected cells. These studies suggest that high endogenous expression of osteonectin in breast cancer cells may reduce metastasis via reduced invasive activity and reduced tumor cell-platelet aggregation.


Subject(s)
Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Osteonectin/biosynthesis , Adenoviridae/genetics , Animals , Blood Platelets/cytology , Breast Neoplasms/blood , Breast Neoplasms/virology , Cell Growth Processes/physiology , Cell Line, Tumor , Female , Genetic Vectors/genetics , Humans , Mice , Mice, Nude , Neoplasm Invasiveness , Neoplasm Metastasis , Osteonectin/genetics , Thrombocytopenia/therapy
SELECTION OF CITATIONS
SEARCH DETAIL
...