Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 50
Filter
1.
Cells ; 10(10)2021 10 17.
Article in English | MEDLINE | ID: mdl-34685761

ABSTRACT

Malignant glioma is one of the most lethal cancers with rapid progression, high recurrence, and poor prognosis in the central nervous system. Fatty acid-binding protein 6 (FABP6) is a bile acid carrier protein that is overexpressed in colorectal cancer. This study aimed to assess the involvement of FABP6 expression in the progression of malignant glioma. Immunohistochemical analysis revealed that FABP6 expression was higher in glioma than in normal brain tissue. After the knockdown of FABP6, a decrease in the migration and invasion abilities of glioma cells was observed. The phosphorylation of the myosin light chain was inhibited, which may be associated with migration ability. Moreover, expression levels of invasion-related proteins, matrix metalloproteinase-2 (MMP-2) and cathepsin B, were reduced. Furthermore, tube formation was inhibited in the human umbilical vein endothelial cells with a decreased concentration of vascular endothelial growth factor (VEGF) in the conditioned medium after the knockdown of FABP6. The phosphorylation of the extracellular signal-regulated kinase (ERK), c-Jun NH2-terminal kinase (JNK), and p65 were also decreased after FABP6 reduction. Finally, the bioluminescent images and immunostaining of MMP-2, cluster of differentiation 31 (CD31), and the VEGF receptor 1 (VEGFR1) revealed attenuated tumor progression in the combination of the FABP6-knocked-down and temozolomide (TMZ)-treated group in an orthotopic xenograft mouse tumor model. This is the first study that revealed the impact of FABP6 on the invasion, angiogenesis, and progression of glioma. The results of this study show that FABP6 may be a potential therapeutic target combined with TMZ for malignant gliomas.


Subject(s)
Fatty Acid-Binding Proteins/antagonists & inhibitors , Gastrointestinal Hormones/antagonists & inhibitors , Glioblastoma/blood supply , Glioblastoma/metabolism , Matrix Metalloproteinase 2/metabolism , Neovascularization, Pathologic/metabolism , Vascular Endothelial Growth Factor A/metabolism , Animals , Cell Line, Tumor , Cell Movement/genetics , Clone Cells , Disease Progression , Extracellular Matrix/metabolism , Fatty Acid-Binding Proteins/genetics , Fatty Acid-Binding Proteins/metabolism , Gastrointestinal Hormones/genetics , Gastrointestinal Hormones/metabolism , Gene Expression Regulation, Neoplastic , Glioblastoma/pathology , Humans , Mice, Nude , Neoplasm Invasiveness , Phosphorylation , RNA, Small Interfering/metabolism , Temozolomide/pharmacology , Xenograft Model Antitumor Assays
2.
Cell Biochem Funct ; 39(2): 308-316, 2021 Mar.
Article in English | MEDLINE | ID: mdl-32876972

ABSTRACT

Cancer cell derived exosomes play important roles in cancer progression and modulation of the tumour microenvironment. This study aims to investigate the role of prokineticin receptor 1 (PKR1) positive exosomes on angiogenesis. In the present study, PKR1 expression in tumour samples from ovarian cancer patients were examined firstly. Then, two ovarian cancer cell lines, namely A2780 and HO-8910 cells, were used to isolate and obtain the PKR1 positive exosomes from the serum free medium. The function analysis of PKR1 positive exosomes on angiogenesis was conducted by cell proliferation and migration assay, tube formation analysis, and tumour volume assay. The results showed that PKR1 expression was down regulated in tumour samples of ovarian cancer patients compared with adjacent normal tissues. The intracellular expression of PKR1 could be detected in A2780 and HO-8910 cells. And, the isolated exosomes from the serum free medium were confirmed by transmission electron microscopic and NTA analysis, as well as the co-presence of PKR1 with exosome marker CD63. The function analysis of PKR1 positive exosomes on angiogenesis demonstrated the uptake of PKR1 positive exosomes by human umbilical vein endothelial cells through immunofluorescence staining. The angiogenesis assays in vitro indicated that PKR1 positive exosomes promoted migration and tube formation of HUVECs but not proliferation. The endogenous PKR1 was also verified to help to enhance migration and promote tube formation of vascular endothelial cells, which might involved in the phosphorylation of STAT3. Additionally, The tumour volume from exosomes treated A2780 tumour-bearing mice was significantly increased compared with the control group, accompanied with the induced PKR1 expression and phosphorylation of STAT3 level. SIGNIFICANCE OF THE STUDY: This study proved the important role of PKR1 positive exosomes released from ovarian cancer cells on promoting angiogenesis. The data indicated that PKR1 derived from ovarian cancer cells could act as an important tumour associated antigen and biomolecular factor for cellular communication in tumour microenvironment.


Subject(s)
Exosomes/metabolism , Gastrointestinal Hormones/metabolism , Neovascularization, Physiologic , Vascular Endothelial Growth Factor, Endocrine-Gland-Derived/metabolism , Animals , Cell Line, Tumor , Cell Movement , Exosomes/transplantation , Female , Gastrointestinal Hormones/antagonists & inhibitors , Gastrointestinal Hormones/genetics , Human Umbilical Vein Endothelial Cells , Humans , Mice , Mice, Nude , Ovarian Neoplasms/metabolism , Ovarian Neoplasms/pathology , Ovarian Neoplasms/therapy , Phosphorylation , RNA Interference , RNA, Small Interfering/metabolism , STAT3 Transcription Factor/metabolism , Signal Transduction , Transplantation, Heterologous , Vascular Endothelial Growth Factor, Endocrine-Gland-Derived/antagonists & inhibitors , Vascular Endothelial Growth Factor, Endocrine-Gland-Derived/genetics
3.
Front Immunol ; 11: 2119, 2020.
Article in English | MEDLINE | ID: mdl-33072073

ABSTRACT

Neurotoxicity is a common side effect of chemotherapeutics that often leads to the development of chemotherapy-induced peripheral neuropathy (CIPN). The peptide Prokineticin 2 (PK2) has a key role in experimental models of CIPN and can be considered an insult-inducible endangering mediator. Since primary afferent sensory neurons are highly sensitive to anticancer drugs, giving rise to dysesthesias, the aim of our study was to evaluate the alterations induced by vincristine (VCR) and bortezomib (BTZ) exposure in sensory neuron cultures and the possible preventive effect of blocking PK2 signaling. Both VCR and BTZ induced a concentration-dependent reduction of total neurite length that was prevented by the PK receptor antagonist PC1. Antagonizing the PK system also reduced the upregulation of PK2, PK-R1, TLR4, IL-6, and IL-10 expression induced by chemotherapeutic drugs. In conclusion, inhibition of PK signaling with PC1 prevented the neurotoxic effects of chemotherapeutics, suggesting a promising strategy for neuroprotective therapies against the sensory neuron damage induced by exposure to these drugs.


Subject(s)
Antineoplastic Agents/toxicity , Bortezomib/toxicity , Gastrointestinal Hormones/antagonists & inhibitors , Nerve Tissue Proteins/antagonists & inhibitors , Neuropeptides/antagonists & inhibitors , Neuroprotective Agents/pharmacology , Neurotoxicity Syndromes/prevention & control , Sensory Receptor Cells/drug effects , Triazines/pharmacology , Vincristine/toxicity , Animals , Cells, Cultured , Dose-Response Relationship, Drug , Down-Regulation , Drug Evaluation, Preclinical , Gastrointestinal Hormones/physiology , Gene Expression Regulation/drug effects , Male , Mice , Mice, Inbred C57BL , Nerve Tissue Proteins/physiology , Neurites/drug effects , Neurites/ultrastructure , Neuroimmunomodulation/drug effects , Neuropeptides/physiology , Neuroprotective Agents/therapeutic use , RNA, Messenger/biosynthesis , Sensory Receptor Cells/physiology , Sensory Receptor Cells/ultrastructure , Triazines/therapeutic use
4.
Int J Mol Sci ; 21(17)2020 Sep 02.
Article in English | MEDLINE | ID: mdl-32887509

ABSTRACT

Cervical cancer is the second most frequent type of gynecologic cancer worldwide. Prokineticin 2 (PROK2) is reported to be involved in tumor progression in some malignant tumors. However, the role of PROK2 in the development of cervical cancer remains unknown. Our results indicate that PROK2 is overexpressed in the human cervical cancer. Cervical cancer patients with high PROK2 expression have a shorter overall survival rate (OS) and disease-free survival rate (DFS). PROK2 acts as a potential biomarker for predicting OS and DFS of cervical cancer patients. We further show that PROK2 is important factor for oncogenic migration and invasion in human cervical cancer cells. Knockdown PROK2 significantly inhibited cell migration, invasion, and MMP15 protein expression in HeLa cells. High expression of MMP15 is confirmed in the human cervical cancer, is significantly associated with the shorter overall survival rate (OS) and is correlated with PROK2 expression. Overexpression of PROK2 using PROK2 plasmid significantly reverses the function of knockdown PROK2, and further upregulates MMP15 expression, migration and invasion of human cervical cancer cells. In conclusion, our findings are the first to demonstrate the role of PROK2 as a novel and potential biomarker for clinical use, and reveal the oncogenic functions of PROK2 as therapeutic target for cervical cancer.


Subject(s)
Biomarkers, Tumor/metabolism , Gastrointestinal Hormones/metabolism , Gene Expression Regulation, Neoplastic , Matrix Metalloproteinase 15/metabolism , Neuropeptides/metabolism , Uterine Cervical Neoplasms/pathology , Apoptosis , Biomarkers, Tumor/genetics , Cell Cycle , Cell Movement , Cell Proliferation , Female , Gastrointestinal Hormones/antagonists & inhibitors , Gastrointestinal Hormones/genetics , Humans , Matrix Metalloproteinase 15/chemistry , Matrix Metalloproteinase 15/genetics , Neoplasm Invasiveness , Neuropeptides/antagonists & inhibitors , Neuropeptides/genetics , Prognosis , Tumor Cells, Cultured , Uterine Cervical Neoplasms/genetics , Uterine Cervical Neoplasms/metabolism
5.
Mol Metab ; 29: 182-196, 2019 11.
Article in English | MEDLINE | ID: mdl-31668389

ABSTRACT

OBJECTIVE: Prokineticin 2 (PROK2) is a hypothalamic neuropeptide that plays a critical role in the rhythmicity of physiological functions and inhibits food intake. PROK2 is also expressed in the main olfactory bulb (MOB) as an essential factor for neuro-and morphogenesis. Since the MOB was shown to be strongly involved in eating behavior, we hypothesized that PROK2 could be a new target in the regulation of food intake and energy homeostasis, through its effects in the MOB. We also asked whether PROK2 could be associated with the pathophysiology of obesity, the metabolic syndrome (MetS), and type 2 diabetes (T2D) in humans. METHODS: We assessed in wild type mice whether the expression of Prok2 in the MOB is dependent on the nutritional status. We measured the effect of human recombinant PROK2 (rPROK2) acute injection in the MOB on food intake and olfactory behavior. Then, using a lentivirus expressing Prok2-shRNA, we studied the effects of Prok2 underexpression in the MOB on feeding behavior and glucose metabolism. Metabolic parameters and meal pattern were determined using calorimetric cages. In vivo 2-deoxyglucose uptake measurements were performed in mice after intraperitoneally insulin injection. Plasmatic PROK2 dosages and genetic associations studies were carried out respectively on 148 and more than 4000 participants from the D.E.S.I.R. (Data from an Epidemiologic Study on the Insulin Resistance Syndrome) cohort. RESULTS: Our findings showed that fasting in mice reduced Prok2 expression in the MOB. Acute injection of rPROK2 in the MOB significantly decreased food intake whereas Prok2-shRNA injection resulted in a higher dietary consumption characterized by increased feeding frequency and decreased meal size. Additionally, Prok2 underexpression in the MOB induced insulin resistance compared to scrambled shRNA-injected mice. In the human D.E.S.I.R. cohort, we found a significantly lower mean concentration of plasma PROK2 in people with T2D than in those with normoglycemia. Interestingly, this decrease was no longer significant when adjusted for Body Mass Index (BMI) or calorie intake, suggesting that the association between plasma PROK2 and diabetes is mediated, at least partly, by BMI and feeding behavior in humans. Moreover, common Single Nucleotide Polymorphisms (SNPs) in PROK2 gene were genotyped and associated with incident T2D or impaired fasting glycemia (IFG), MetS, and obesity. CONCLUSIONS: Our data highlight PROK2 as a new target in the MOB that links olfaction with eating behavior and energy homeostasis. In humans, plasma PROK2 is negatively correlated with T2D, BMI, and energy intake, and PROK2 genetic variants are associated with incident hyperglycemia (T2D/IFG), the MetS and obesity.


Subject(s)
Diabetes Mellitus, Type 2/pathology , Feeding Behavior , Gastrointestinal Hormones/metabolism , Insulin Resistance , Neuropeptides/metabolism , Adult , Aged , Animals , Diabetes Mellitus, Type 2/metabolism , Eating/drug effects , Energy Metabolism/drug effects , Feeding Behavior/drug effects , Female , Gastrointestinal Hormones/antagonists & inhibitors , Gastrointestinal Hormones/blood , Gastrointestinal Hormones/genetics , Humans , Male , Mice , Middle Aged , Neuropeptides/antagonists & inhibitors , Neuropeptides/blood , Neuropeptides/genetics , Olfactory Bulb/metabolism , Polymorphism, Single Nucleotide , RNA Interference , RNA, Small Interfering/metabolism , Recombinant Proteins/biosynthesis , Recombinant Proteins/pharmacology
6.
Physiol Res ; 67(Suppl 1): S69-S81, 2018 06 27.
Article in English | MEDLINE | ID: mdl-29947529

ABSTRACT

The heavy impact of obesity on the development and progression of cardiovascular disease has sparked sustained efforts to uncover the mechanisms linking excess adiposity to vascular dysfunction. Impaired vasodilator reactivity has been recognized as an early hemodynamic abnormality in obese patients, but also increased vasoconstrictor tone importantly contributes to their vascular damage. In particular, upregulation of the endothelin (ET)-1 system, consistently reported in these patients, might accelerate atherosclerosis and its complication, given the pro-inflammatory and mitogenic properties of ET-1. In recent years, a number of gut hormones, in addition to their role as modulators of food intake, energy balance, glucose and lipid metabolism, and insulin secretion and action, have demonstrated favorable vascular actions. They increase the bioavailability of vasodilator mediators like nitric oxide, but they have also been shown to inhibit the ET-1 system. These features make gut hormones promising tools for targeting both the metabolic and cardiovascular complications of obesity, a view supported by recent large-scale clinical trials indicating that novel drugs for type 2 diabetes with cardiovascular potential may translate into clinically significant advantages. Therefore, there is real hope that better understanding of the properties of gut-derived substances might provide more effective therapies for the obesity-related cardiometabolic syndrome.


Subject(s)
Endothelin-1/metabolism , Gastrointestinal Hormones/metabolism , Obesity/metabolism , Vasoconstriction/drug effects , Vasoconstrictor Agents/administration & dosage , Vasoconstrictor Agents/metabolism , Anti-Obesity Agents/administration & dosage , Anti-Obesity Agents/metabolism , Diabetes Mellitus, Type 2/drug therapy , Diabetes Mellitus, Type 2/metabolism , Endothelin-1/agonists , Endothelin-1/antagonists & inhibitors , Gastrointestinal Hormones/antagonists & inhibitors , Humans , Insulin Resistance/physiology , Obesity/drug therapy , Peptide Hormones/antagonists & inhibitors , Peptide Hormones/metabolism , Vasoconstriction/physiology
7.
Peptides ; 100: 190-201, 2018 02.
Article in English | MEDLINE | ID: mdl-29412819

ABSTRACT

Chemical derivatives of the gut-derived peptide hormone glucagon-like peptide 1 (GLP-1) are among the best-in-class pharmacotherapies to treat obesity and type 2 diabetes. However, GLP-1 analogs have modest weight lowering capacity, in the range of 5-10%, and the therapeutic window is hampered by dose-dependent side effects. Over the last few years, a new concept has emerged: combining the beneficial effects of several key metabolic hormones into a single molecular entity. Several unimolecular GLP-1-based polyagonists have shown superior metabolic action compared to GLP-1 monotherapies. In this review article, we highlight the history of polyagonists targeting the receptors for GLP-1, GIP and glucagon, and discuss recent progress in expanding of this concept to now allow targeted delivery of nuclear hormones via GLP-1 and other gut hormones, as a novel approach towards more personalized pharmacotherapies.


Subject(s)
Diabetes Mellitus, Type 2/drug therapy , Gastric Inhibitory Polypeptide/therapeutic use , Glucagon-Like Peptide 1/therapeutic use , Obesity/drug therapy , Diabetes Mellitus, Type 2/metabolism , Diabetes Mellitus, Type 2/pathology , Gastric Inhibitory Polypeptide/antagonists & inhibitors , Gastrointestinal Hormones/antagonists & inhibitors , Gastrointestinal Hormones/genetics , Gastrointestinal Hormones/therapeutic use , Glucagon-Like Peptide 1/analogs & derivatives , Glucagon-Like Peptide 1/antagonists & inhibitors , Glucagon-Like Peptide-1 Receptor/antagonists & inhibitors , Glucagon-Like Peptide-1 Receptor/genetics , Humans , Incretins/metabolism , Insulin/genetics , Insulin/metabolism , Obesity/metabolism , Obesity/pathology , Receptors, Gastrointestinal Hormone/antagonists & inhibitors , Receptors, Gastrointestinal Hormone/genetics , Receptors, Glucagon/antagonists & inhibitors , Receptors, Glucagon/genetics
8.
BMC Musculoskelet Disord ; 17: 387, 2016 09 08.
Article in English | MEDLINE | ID: mdl-27609223

ABSTRACT

BACKGROUND: Prokineticin 2 (PK2) expression is upregulated in mice with collagen-induced arthritis (CIA), an animal model of rheumatoid arthritis. The purpose of our study was to investigate the effects of PK2 inhibition on CIA. METHODS: PK2, prokineticin receptor (PKR) 1, and PKR2 mRNA transcripts in the joints of CIA mice were measured by real-time PCR on Days 21, 28, and 35 (n = 15/day). Localization of PKR1 and PKR2 proteins was examined immunohistochemically. PKRA7, a PK2 antagonist, was administered intraperitoneally for 2 weeks to CIA mice, and the severity of arthritis was compared between treated (n = 12) and untreated (n = 12) mice. The gene expression levels of inflammatory cytokines IL-1ß, IL-6, TNF-α, and VEGF were also measured by real-time PCR and compared between treated (n = 6) and untreated (n = 6) CIA mice. The data was statistically analyzed, and P values of less than 0.05 were considered significant. RESULTS: In the thickened synovial membrane, PKR1 protein was expressed in infiltrating neutrophils, while PKR2 expression was found in macrophage-like mononuclear cells. PK2 gene expression was significantly more pronounced on Days 28 and 35 than on Day 21 (2.15 and 2.03 versus 1.00, P = 0.0311 and 0.0247; Dunn's multiple comparison). PKR2 gene expression levels were significantly higher on Days 28 and 35 compared to Day 21 (25.4 and 39.3 versus 1.0, P = 0.002 and < 0.0001; Dunn's multiple comparison). Administration of PKRA7 suppressed the severity of arthritis (P < 0.001; two-way analysis of variance). A gene expression analysis of inflammatory cytokines revealed significantly reduced IL-1ß and lL-6 expression in the joints of PKRA7-treated mice compared to untreated mice (0.1 versus 1.0, P = 0.0043 and 0.04 versus 1.0, P = 0.0022, respectively; Mann-Whitney test). CONCLUSIONS: PK2 inhibition suppressed arthritis in mice with CIA.


Subject(s)
Angiogenesis Inhibitors/therapeutic use , Arthritis, Experimental/drug therapy , Arthritis, Rheumatoid/drug therapy , Gastrointestinal Hormones/antagonists & inhibitors , Neuropeptides/antagonists & inhibitors , Oxepins/therapeutic use , Pyrrolidines/therapeutic use , Angiogenesis Inhibitors/administration & dosage , Animals , Arthritis, Experimental/immunology , Collagen Type II/immunology , Cytokines/metabolism , Gastrointestinal Hormones/metabolism , Gene Expression Profiling , Humans , Injections, Intraperitoneal , Mice , Mice, Inbred DBA , Neuropeptides/metabolism , Oxepins/administration & dosage , Pyrrolidines/administration & dosage , Real-Time Polymerase Chain Reaction , Synovial Membrane/metabolism
9.
J Med Chem ; 59(17): 8094-102, 2016 09 08.
Article in English | MEDLINE | ID: mdl-27500412

ABSTRACT

Fatty acid binding protein 6 (FABP6) is a potential drug discovery target, which, if inhibited, may have a therapeutic benefit for the treatment of diabetes. Currently, there are no published inhibitors of FABP6, and with the target believed to be amenable to fragment-based drug discovery, a structurally enabled program was initiated. This program successfully identified fragment hits using the surface plasmon resonance (SPR) platform. Several hits were validated with SAR and were found to be displaced by the natural ligand taurocholate. We report the first crystal structure of human FABP6 in the unbound form, in complex with cholate, and with one of the key fragments.


Subject(s)
Bile Acids and Salts/chemistry , Fatty Acid-Binding Proteins/chemistry , Gastrointestinal Hormones/chemistry , Binding Sites , Crystallography, X-Ray , Fatty Acid-Binding Proteins/antagonists & inhibitors , Gastrointestinal Hormones/antagonists & inhibitors , Humans , Models, Molecular , Protein Conformation , Structure-Activity Relationship , Surface Plasmon Resonance , Taurocholic Acid/chemistry
10.
Ann Surg ; 262(5): 824-29; discussion 829-30, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26583672

ABSTRACT

OBJECTIVES: To characterize the gut hormone profile and determine the effect of satiety gut hormone blockade on food intake in disease-free postesophagectomy patients. BACKGROUND: Improved oncologic outcomes for esophageal cancer have resulted in increased survivorship and a focus on health-related quality of life. Anorexia and early satiety are common, but putative causative factors, in particular the gut-brain hormonal axis, have not been systematically studied. METHODS: In a double-blind, placebo-controlled, randomized crossover study, disease-free patients at least 1 year postresection and gastric conduit reconstruction received either 1 mL 0.9% saline or 1 mL (100 µg) octreotide acetate subcutaneously followed by a standardized ad libitum meal on each of two assessments. Fasting and postprandial plasma glucagon-like peptide 1 (GLP-1), peptide YY (PYY), and ghrelin immunoreactivity were measured. Gut hormone responses and calorie intake postsaline versus octreotide were compared between experimental and control groups. RESULTS: Eighteen subjects [esophagectomy (ES), n = 10, 2.4 ±â€Š0.75 years postresection; and unoperated control subjects, n = 8] were studied. ES demonstrated significant weight loss at 3, 6, 12, and 24 months postoperatively (all P < 0.05). Ghrelin levels were similar (P = 0.58) for both groups, but postprandial GLP-1 and PYY responses were significantly (P < 0.001) greater among ES as compared with controls. After octreotide, ad libitum calorie intake increased among ES (1.5 ±â€Š0.2 fold-change, P = 0.02) but not controls (1.1 ±â€Š0.1 fold-change, P = 0.30). CONCLUSIONS: ES demonstrated an exaggerated postprandial satiety gut hormone response that was attenuated by octreotide, thus identifying a potential therapeutic target to modulate in the ES patient with early satiety.


Subject(s)
Eating/drug effects , Esophageal Neoplasms/surgery , Esophagectomy , Gastrointestinal Hormones/antagonists & inhibitors , Gastroplasty , Octreotide/administration & dosage , Quality of Life , Body Mass Index , Cross-Over Studies , Double-Blind Method , Esophageal Neoplasms/metabolism , Female , Gastrointestinal Agents/administration & dosage , Humans , Injections, Subcutaneous , Male , Middle Aged
11.
Eur J Pharmacol ; 767: 24-9, 2015 Nov 15.
Article in English | MEDLINE | ID: mdl-26435025

ABSTRACT

Prokineticin 2 (PK2), a new chemokine, causes mechanical hypersensitivity in the rat hind paw, but little is known about the molecular mechanism. Here, we have found that ionotropic P2X receptor is essential to mechanical allodynia induced by PK2. First, intraplantar injection of high dose (3 or 10 pmol) of PK2 significantly increased paw withdrawal response frequency (%) to innocuous mechanical stimuli (mechanical allodynia). And the mechanical allodynia induced by PK2 was prevented by co-administration of TNP-ATP, a selective P2X receptor antagonist. Second, although low dose (0.3 or 1 pmol) of PK2 itself did not produce an allodynic response, it significantly facilitated the mechanical allodynia evoked by intraplantar injection of α,ß-methylene ATP (α,ß-meATP). Third, PK2 concentration-dependently potentiated α,ß-meATP-activated currents in rat dorsal root ganglion (DRG) neurons. Finally, PK2 receptors and intracellular signal transduction were involved in PK2 potentiation of α,ß-meATP-induced mechanical allodynia and α,ß-meATP-activated currents, since the potentiation were blocked by PK2 receptor antagonist PKRA and selective PKC inhibitor GF 109203X. These results suggested that PK2 facilitated mechanical allodynia induced by α,ß-meATP through a mechanism involved in sensitization of cutaneous P2X receptors expressed by nociceptive nerve endings.


Subject(s)
Adenosine Triphosphate/analogs & derivatives , Gastrointestinal Hormones/pharmacology , Hyperalgesia/chemically induced , Neuropeptides/pharmacology , Adenosine Triphosphate/adverse effects , Adenosine Triphosphate/pharmacology , Animals , Drug Synergism , Ganglia, Spinal/drug effects , Ganglia, Spinal/physiology , Gastrointestinal Hormones/antagonists & inhibitors , Hyperalgesia/physiopathology , Indoles/pharmacology , Male , Maleimides/pharmacology , Membrane Potentials/drug effects , Membrane Potentials/physiology , Neuropeptides/antagonists & inhibitors , Protein Kinase C/antagonists & inhibitors , Purinergic P2X Receptor Antagonists/pharmacology , Rats , Receptors, G-Protein-Coupled/antagonists & inhibitors , Receptors, Peptide/antagonists & inhibitors , Receptors, Purinergic P2X3/drug effects , Receptors, Purinergic P2X3/physiology
12.
Neoplasia ; 16(6): 501-10, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24957319

ABSTRACT

Weekly gemcitabine therapy is the major treatment offered for patients with pancreatic adenocarcinoma cancer; however, relative resistance of tumor cells to chemotherapy, rapid regrowth, and metastasis are the main causes of death within a year. Recently, the daily continuous administration of chemotherapy in low doses--called metronomic chemotherapy (MC)--has been shown to inhibit primary tumor growth and delay metastases in several tumor types; however, its use as a single therapy is still in question due to its moderate therapeutic benefit. Here, we show that the combination of weekly gemcitabine with MC of the same drug delays tumor regrowth and inhibits metastasis in mice implanted orthotopically with pancreatic tumors. We further demonstrate that weekly gemcitabine, but not continuous MC gemcitabine or the combination of the two drug regimens, promotes rebound myeloid-derived suppressor cell (MDSC) mobilization and increases angiogenesis in this tumor model. Furthermore, Bv8 is highly expressed in MDSCs colonizing pancreatic tumors in mice treated with weekly gemcitabine compared to MC gemcitabine or the combination of the two regimens. Blocking Bv8 with antibodies in weekly gemcitabine-treated mice results in a significant reduction in tumor regrowth, angiogenesis, and metastasis. Overall, our results suggest that pro-tumorigenic effects induced by weekly gemcitabine are mediated in part by MDSCs expressing Bv8. Therefore, both Bv8 inhibition and MC can be used as legitimate 'add-on' treatments for preventing post-chemotherapy pancreatic cancer recurrence, progression, and metastasis following weekly gemcitabine therapy.


Subject(s)
Adenocarcinoma/drug therapy , Antibodies, Monoclonal/administration & dosage , Antimetabolites, Antineoplastic/administration & dosage , Deoxycytidine/analogs & derivatives , Gastrointestinal Hormones/antagonists & inhibitors , Neuropeptides/antagonists & inhibitors , Pancreatic Neoplasms/drug therapy , Adenocarcinoma/mortality , Adenocarcinoma/pathology , Administration, Metronomic , Animals , Cell Line, Tumor , Cell Movement/drug effects , Deoxycytidine/administration & dosage , Disease Models, Animal , Female , Gastrointestinal Hormones/metabolism , Humans , Mice , Neoplasm Metastasis , Neuropeptides/metabolism , Pancreatic Neoplasms/mortality , Pancreatic Neoplasms/pathology , Treatment Outcome , Tumor Burden/drug effects , Xenograft Model Antitumor Assays , Gemcitabine
13.
PLoS One ; 9(3): e90860, 2014.
Article in English | MEDLINE | ID: mdl-24633064

ABSTRACT

The possible signaling role of prokineticin 2 (PK2) and its receptor, prokineticin receptor 2 (PKR2), on female reproduction was investigated. First, the expression of PKR2 and its co-localization with estrogen receptor (ERα) in the hypothalamus was examined. Sexually dimorphic expression of PKR2 in the preoptic area of the hypothalamus was observed. Compared to the male mice, there was more widespread PKR2 expression in the preoptic area of the hypothalamus in the female mice. The likely co-expression of PKR2 and ERα in the preoptic area of the hypothalamus was observed. The estrous cycles in female PK2-null, and PKR2-null heterozygous mice, as well as in PK2-null and PKR2-null compound heterozygous mice were examined. Loss of one copy of PK2 or PKR2 gene caused elongated and irregular estrous cycle in the female mice. The alterations in the estrous cycle were more pronounced in PK2-null and PKR2-null compound heterozygous mice. Consistent with these observations, administration of a small molecule PK2 receptor antagonist led to temporary blocking of estrous cycle at the proestrous phase in female mice. The administration of PKR2 antagonist was found to blunt the circulating LH levels. Taken together, these studies indicate PK2 signaling is required for the maintenance of normal female estrous cycles.


Subject(s)
Estrous Cycle/physiology , Gastrointestinal Hormones/metabolism , Neuropeptides/metabolism , Animals , Estrogen Receptor alpha/metabolism , Estrous Cycle/drug effects , Female , Gastrointestinal Hormones/antagonists & inhibitors , Gastrointestinal Hormones/genetics , Hypothalamus/metabolism , Mice , Mice, Knockout , Neuropeptides/antagonists & inhibitors , Neuropeptides/genetics
14.
Drug Discov Today ; 19(7): 845-58, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24291217

ABSTRACT

Obesity is a leading cause of preventable mortality worldwide, with current strategies for treatment including life-style changes, pharmacological intervention and bariatric surgery. With pharmacological intervention showing at best modest patient benefits, new treatments are required. Modulation of anorectic gut hormones could offer the potential to elicit the required life-changing level of efficacy only currently seen with bariatric surgery, and without the cardiovascular risk associated with a number of the current marketed therapies. This review will discuss the gut hormones glucagon-like peptide-1 (GLP-1), Ghrelin and cholecystokinin (CCK)--for which more advanced non-peptide chemical matter has been discovered acting through these hormone pathways and/or their receptors.


Subject(s)
Gastrointestinal Hormones/antagonists & inhibitors , Obesity/drug therapy , Obesity/surgery , Animals , Anti-Obesity Agents/pharmacology , Anti-Obesity Agents/therapeutic use , Bariatric Surgery/methods , Gastrointestinal Hormones/metabolism , Gastrointestinal Tract/drug effects , Gastrointestinal Tract/metabolism , Gastrointestinal Tract/surgery , Humans , Obesity/metabolism , Risk Factors
15.
PLoS One ; 8(1): e54916, 2013.
Article in English | MEDLINE | ID: mdl-23372791

ABSTRACT

Infiltration of myeloid cells in the tumor microenvironment is often associated with enhanced angiogenesis and tumor progression, resulting in poor prognosis in many types of cancer. The polypeptide chemokine PK2 (Bv8, PROK2) has been shown to regulate myeloid cell mobilization from the bone marrow, leading to activation of the angiogenic process, as well as accumulation of macrophages and neutrophils in the tumor site. Neutralizing antibodies against PK2 were shown to display potent anti-tumor efficacy, illustrating the potential of PK2-antagonists as therapeutic agents for the treatment of cancer. In this study we demonstrate the anti-tumor activity of a small molecule PK2 antagonist, PKRA7, in the context of glioblastoma and pancreatic cancer xenograft tumor models. For the highly vascularized glioblastoma, PKRA7 was associated with decreased blood vessel density and increased necrotic areas in the tumor mass. Consistent with the anti-angiogenic activity of PKRA7 in vivo, this compound effectively reduced PK2-induced microvascular endothelial cell branching in vitro. For the poorly vascularized pancreatic cancer, the primary anti-tumor effect of PKRA7 appears to be mediated by the blockage of myeloid cell migration/infiltration. At the molecular level, PKRA7 inhibits PK2-induced expression of certain pro-migratory chemokines and chemokine receptors in macrophages. Combining PKRA7 treatment with standard chemotherapeutic agents resulted in enhanced effects in xenograft models for both types of tumor. Taken together, our results indicate that the anti-tumor activity of PKRA7 can be mediated by two distinct mechanisms that are relevant to the pathological features of the specific type of cancer. This small molecule PK2 antagonist holds the promise to be further developed as an effective agent for combinational cancer therapy.


Subject(s)
Antineoplastic Agents/pharmacology , Carrier Proteins/metabolism , Cell Transformation, Neoplastic/drug effects , Gastrointestinal Hormones/antagonists & inhibitors , Glioma/pathology , Myeloid Cells/pathology , Neovascularization, Pathologic , Nerve Tissue Proteins/metabolism , Neuropeptides/antagonists & inhibitors , Pancreatic Neoplasms/pathology , Animals , Antineoplastic Agents/administration & dosage , Cell Line, Tumor , Cell Movement/drug effects , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Glioma/drug therapy , Glioma/mortality , Humans , Macrophages/drug effects , Macrophages/pathology , Mice , Myeloid Cells/drug effects , Neovascularization, Pathologic/drug therapy , Pancreatic Neoplasms/drug therapy , Pancreatic Neoplasms/mortality , Receptors, N-Methyl-D-Aspartate , Tumor Burden/drug effects , Tumor Microenvironment , Xenograft Model Antitumor Assays
16.
Trends Pharmacol Sci ; 32(1): 8-15, 2011 Jan.
Article in English | MEDLINE | ID: mdl-21130506

ABSTRACT

Bariatric surgery has proven to be an effective means of treating 'diabesity': the combination of type 2 diabetes mellitus (T2DM) and obesity. The effects of surgery go beyond weight loss but reflect a complex alteration in secretion of gut hormones. Finding a pharmaceutical alternative that mimics the benefits of surgery without surgical complications has become the 'holy grail' of the twenty-first century. As knowledge of the multifaceted functions of gut hormones increases, a multitude of drugs that exploit these actions has emerged. In this review, we examine the current understanding of the mechanisms by which bariatric surgery improves diabesity. We also discuss the rapidly emerging role of glucagon-like peptide-1-based treatments as well as the potential for new therapeutics based on other gut hormones (e.g. oxyntomodulin, peptide YY, gastric inhibitory peptide, ghrelin).


Subject(s)
Diabetes Mellitus, Type 2/complications , Diabetes Mellitus, Type 2/drug therapy , Gastrointestinal Hormones/metabolism , Obesity/complications , Obesity/drug therapy , Receptors, Gastrointestinal Hormone/metabolism , Animals , Bariatric Surgery/adverse effects , Diabetes Mellitus, Type 2/metabolism , Diabetes Mellitus, Type 2/surgery , Gastrointestinal Hormones/agonists , Gastrointestinal Hormones/antagonists & inhibitors , Ghrelin/antagonists & inhibitors , Ghrelin/metabolism , Humans , Molecular Targeted Therapy , Obesity/metabolism , Obesity/physiopathology , Obesity/surgery , Receptors, Gastrointestinal Hormone/agonists , Receptors, Gastrointestinal Hormone/antagonists & inhibitors , Receptors, Ghrelin/antagonists & inhibitors , Receptors, Ghrelin/metabolism
17.
Pflugers Arch ; 457(3): 609-22, 2009 Jan.
Article in English | MEDLINE | ID: mdl-18536934

ABSTRACT

The gastric H,K-ATPase, a member of the P(2)-type ATPase family, is the integral membrane protein responsible for gastric acid secretion. It is an alpha,beta-heterodimeric enzyme that exchanges cytoplasmic hydronium with extracellular potassium. The catalytic alpha subunit has ten transmembrane segments with a cluster of intramembranal carboxylic amino acids located in the middle of the transmembrane segments TM4, TM5,TM6, and TM8. Comparison to the known structure of the SERCA pump, mutagenesis, and molecular modeling has identified these as constituents of the ion binding domain. The beta subunit has one transmembrane segment with N terminus in cytoplasmic region. The extracellular domain of the beta subunit contains six or seven N-linked glycosylation sites. N-glycosylation is important for the enzyme assembly, maturation, and sorting. The enzyme pumps acid by a series of conformational changes from an E(1) (ion site in) to an E(2) (ion site out) configuration following binding of MgATP and phosphorylation. Several experimental observations support the hypothesis that expulsion of the proton at 160 mM (pH 0.8) results from movement of lysine 791 into the ion binding site in the E(2)P configuration. Potassium access from the lumen depends on activation of a K and Cl conductance via a KCNQ1/KCNE2 complex and Clic6. K movement through the luminal channel in E(2)P is proposed to displace the lysine along with dephosphorylation to return the enzyme to the E(1) configuration. This enzyme is inhibited by the unique proton pump inhibitor class of drug, allowing therapy of acid-related diseases.


Subject(s)
Gastrointestinal Hormones , Stomach/enzymology , Binding Sites , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/metabolism , Gastrointestinal Hormones/antagonists & inhibitors , Gastrointestinal Hormones/chemistry , Gastrointestinal Hormones/metabolism , Humans , Models, Molecular , Molecular Structure , Potassium/metabolism , Protein Conformation , Stomach/cytology
18.
FASEB J ; 22(5): 1306-16, 2008 May.
Article in English | MEDLINE | ID: mdl-18096816

ABSTRACT

The heat-stable enterotoxin of Escherichia coli (STa) is a potent stimulant of intestinal chloride and bicarbonate secretion. Guanylyl cyclase C (GC-C) has been shown to be the primary receptor involved in mediating this response. However, numerous studies have suggested the existence of an alternative STa-binding receptor. The aims of this study were to determine whether a non-GC-C receptor exists for STa and what is the functional relevance of this for intestinal bicarbonate secretion in mice. (125)I-STa-binding experiments were performed with intestinal mucosae from GC-C knockout (KO) and wild type (WT) mice. Subsequently, the functional relevance of an alternative STa-binding receptor was explored by examining STa-, uroguanylin-, and guanylin-stimulated duodenal bicarbonate secretion (DBS) in GC-C KO mice in vitro and in vivo. Significant (125)I-STa-binding occurred in the proximal small intestines of GC-C KO and WT mice. Analysis of binding coefficients and pH dependence showed that (125)I-STa-binding in GC-C KO mice involved a receptor distinct from that of WT mice. Functionally, STa, uroguanylin, and guanylin all stimulated a significant increase in DBS in GC-C KO mice. Uroguanylin- and guanylin-stimulated DBS were significantly inhibited by glibenclamide, but not by 4,4'-diisothiocyanato-stilbene-2,2'-disulfonic acid (DIDS). However, STa-stimulated DBS was unaffected by glibenclamide but inhibited by DIDS. Taken together, our results suggest that alternative, non-GC-C, receptors likely exist for STa, uroguanylin, and guanylin in the intestines of mice. While uroguanylin- and guanylin-stimulated DBS are cystic fibrosis transmembrane conductance regulator (CFTR) dependent, STa-stimulated DBS is CFTR independent. Further understanding of this alternative receptor and its signaling pathway may provide important insights into rectification of intestinal bicarbonate secretion in cystic fibrosis.


Subject(s)
Bacterial Toxins/pharmacology , Bicarbonates/metabolism , Cystic Fibrosis Transmembrane Conductance Regulator/physiology , Duodenum/drug effects , Duodenum/metabolism , Enterotoxins/pharmacology , Guanylate Cyclase/physiology , Receptors, Peptide/physiology , 4,4'-Diisothiocyanostilbene-2,2'-Disulfonic Acid/pharmacology , Animals , Duodenum/ultrastructure , Escherichia coli Proteins , Gastrointestinal Hormones/antagonists & inhibitors , Gastrointestinal Hormones/pharmacology , Glyburide/pharmacology , In Vitro Techniques , Mice , Mice, Knockout , Microvilli/ultrastructure , Natriuretic Peptides/pharmacology , Receptors, Enterotoxin , Receptors, Guanylate Cyclase-Coupled , Sodium-Hydrogen Exchangers/metabolism
19.
Nature ; 450(7171): 825-31, 2007 Dec 06.
Article in English | MEDLINE | ID: mdl-18064003

ABSTRACT

Bone-marrow-derived cells facilitate tumour angiogenesis, but the molecular mechanisms of this facilitation are incompletely understood. We have previously shown that the related EG-VEGF and Bv8 proteins, also known as prokineticin 1 (Prok1) and prokineticin 2 (Prok2), promote both tissue-specific angiogenesis and haematopoietic cell mobilization. Unlike EG-VEGF, Bv8 is expressed in the bone marrow. Here we show that implantation of tumour cells in mice resulted in upregulation of Bv8 in CD11b+Gr1+ myeloid cells. We identified granulocyte colony-stimulating factor as a major positive regulator of Bv8 expression. Anti-Bv8 antibodies reduced CD11b+Gr1+ cell mobilization elicited by granulocyte colony-stimulating factor. Adenoviral delivery of Bv8 into tumours was shown to promote angiogenesis. Anti-Bv8 antibodies inhibited growth of several tumours in mice and suppressed angiogenesis. Anti-Bv8 treatment also reduced CD11b+Gr1+ cells, both in peripheral blood and in tumours. The effects of anti-Bv8 antibodies were additive to those of anti-Vegf antibodies or cytotoxic chemotherapy. Thus, Bv8 modulates mobilization of CD11b+Gr1+ cells from the bone marrow during tumour development and also promotes angiogenesis locally.


Subject(s)
Gastrointestinal Hormones/metabolism , Myeloid Cells/metabolism , Neoplasms/blood supply , Neovascularization, Pathologic , Neuropeptides/metabolism , Animals , Antibodies/immunology , Antibodies/pharmacology , Antineoplastic Agents/pharmacology , Cell Division/drug effects , Cell Line, Tumor , Gastrointestinal Hormones/antagonists & inhibitors , Gastrointestinal Hormones/immunology , Gene Expression Regulation/drug effects , Granulocyte Colony-Stimulating Factor/pharmacology , Humans , Mice , Mice, Nude , Myeloid Cells/drug effects , Neoplasm Transplantation , Neoplasms/drug therapy , Neoplasms/metabolism , Neoplasms/pathology , Neovascularization, Pathologic/drug therapy , Neuropeptides/antagonists & inhibitors , Neuropeptides/immunology , Vascular Endothelial Growth Factor A/antagonists & inhibitors
SELECTION OF CITATIONS
SEARCH DETAIL