Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 3 de 3
Filter
Add more filters










Database
Language
Publication year range
1.
PLoS One ; 10(4): e0123407, 2015.
Article in English | MEDLINE | ID: mdl-25853862

ABSTRACT

The dormancy of tumor cells is a major problem in chemotherapy, since it limits the therapeutic efficacy of anti-tumor drugs that only target dividing cells. One potential way to overcome chemo-resistance is to "wake up" these dormant cells. Here we show that the opioid antagonist methylnaltrexone (MNTX) enhances the effect of docetaxel (Doc) by blocking a cell growth-suppressive pathway. We found that PENK, which encodes opioid growth factor (OGF) and suppresses cell growth, is predominantly expressed in diffuse-type gastric cancers (GCs). The blockade of OGF signaling by MNTX releases cells from their arrest and boosts the effect of Doc. In comparison with the use of Doc alone, the combined use of Doc and MNTX significantly prolongs survival, alleviates abdominal pain, and diminishes Doc-resistant spheroids on the peritoneal membrane in model mice. These results suggest that blockade of the pathways that suppress cell growth may enhance the effects of anti-tumor drugs.


Subject(s)
Naltrexone/analogs & derivatives , Receptors, Opioid/biosynthesis , Stomach Neoplasms/drug therapy , Taxoids/administration & dosage , Animals , Antineoplastic Combined Chemotherapy Protocols , Cell Cycle/drug effects , Cell Cycle/genetics , Cell Proliferation/drug effects , Docetaxel , Drug Synergism , Growth Inhibitors/biosynthesis , Growth Inhibitors/genetics , Humans , Mice , Naltrexone/administration & dosage , Narcotic Antagonists/administration & dosage , Quaternary Ammonium Compounds/administration & dosage , Receptors, Opioid/genetics , Stomach Neoplasms/genetics , Stomach Neoplasms/pathology
2.
Anesthesiology ; 117(4): 847-56, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22913923

ABSTRACT

BACKGROUND: Patients with peritoneal carcinomatosis often report abdominal pain, which is relatively refractory to morphine. It has been considered that a new animal model is required to investigate the mechanism of abdominal pain for the development of optimal treatments for this type of pain. METHODS: To prepare a peritoneal carcinomatosis model, highly peritoneal-seeding gastric cancer cells, 60As6, were implanted into the abdominal cavity. The nociceptive modality for pain-related behavior was assessed in terms of withdrawal behavior in response to mechanical stimuli and hunching behavior. Tissue samples from mouse dorsal root ganglia and spinal cord were subject to immunohistochemistry and real-time reverse transcription polymerase chain reaction. RESULTS: Mice with peritoneal dissemination showed significant hypersensitivity of the abdomen to mechanical stimulation and spontaneous visceral pain-related behavior. There was a significant increase in c-Fos-positive cells in the spinal cord in tumor-bearing mice. Those mice exhibited a remarkable increase in substance P-positive neurons in the dorsal root ganglia (control vs. tumor, 15.4 ± 1.1 vs. 24.2 ± 3.6, P < 0.05, n = 3). A significant decreases in µ-opioid receptor expression mainly in substance P-positive neurons was observed in tumor-bearing mice (69.3 ± 4.9 vs. 38.7 ± 0.9, P < 0.05, n = 3), and a relatively higher dose of morphine was required to significantly reverse the abdominal hypersensitivity. CONCLUSION: Both the up-regulation of substance P and down-regulation of µ-opioid receptor seen in the dorsal root ganglia may be, at least in part, responsible for the abdominal pain-like state associated with peritoneal carcinomatosis.


Subject(s)
Abdominal Pain/etiology , Abdominal Pain/metabolism , Carcinoma/complications , Carcinoma/metabolism , Receptors, Opioid, mu/biosynthesis , Spinal Cord/metabolism , Substance P/biosynthesis , Abdominal Pain/psychology , Animals , Behavior, Animal , Calcitonin Gene-Related Peptide/metabolism , Carcinoma/psychology , Cell Line , Cell Line, Tumor , Glyceraldehyde-3-Phosphate Dehydrogenases/metabolism , Humans , Immunohistochemistry , Inflammation/complications , Inflammation/psychology , Luciferases/metabolism , Mice , Mice, Inbred ICR , Mice, SCID , Neoplasm Transplantation , Pancreatitis/complications , Pancreatitis/psychology , Proto-Oncogene Proteins c-fos/metabolism , Real-Time Polymerase Chain Reaction , Receptors, Opioid, mu/genetics , Substance P/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...