227 results on '"Vasoactive intestinal peptide receptor"'
Search Results
2. Xenin-25 induces anion secretion by activating noncholinergic secretomotor neurons in the rat ileum.
- Author
-
Atsukazu Kuwahara, Yuko Kuwahara, Ikuo Kato, Kotoku Kawaguchi, Daiki Harata, Shinji Asano, Toshio Inui, and Yoshinori Marunaka
- Abstract
Xenin-25 is a neurotensin-like peptide that is secreted by enteroendocrine cells in the small intestine. Xenin-8 is reported to augment duodenal anion secretion by activating afferent neural pathways. The intrinsic neuronal circuits mediating the xenin-25-induced anion secretion were characterized using the Ussing-chambered, mucosa-submucosa preparation from the rat ileum. Serosal application of xenin-25 increased the short-circuit current in a concentration-dependent manner. The responses were abolished by the combination of Cl−-free and HCO3− -free solutions. The responses were almost completely blocked by TTX (10-6 M) but not by atropine (10-5 M) or hexamethonium (10-4 M). The selective antagonists for neurotensin receptor 1 (NTSR1), neurokinin 1 (NK1), vasoactive intestinal polypeptide (VIP) receptors 1 and 2 (VPAC1 and VPAC2, respectively), and capsaicin, but not 5-hydroxyltryptamine receptors 3 and 4 (5-HT3 and 5-HT4), NTSR2, and A803467, inhibited the responses to xenin-25. The expression of VIP receptors (Vipr) in rat ileum was examined using RT-PCR. The Vipr1 PCR products were detected in the submucosal plexus and mucosa. Immunohistochemical staining showed the colocalization of NTSR1 and NK1 with substance P (SP)- and calbindin-immunoreactive neurons in the submucosal plexus, respectively. In addition, NK1 was colocalized with noncholinergic VIP secretomotor neurons. Based on the results from the present study, xenin-25-induced Cl−/ HCO3− secretion is involved in NTSR1 activation on intrinsic and extrinsic afferent neurons, followed by the release of SP and subsequent activation of NK1 expressed on noncholinergic VIP secretomotor neurons. Finally, the secreted VIP may activate VPAC1 on epithelial cells to induce Cl−/ HCO3− secretion in the rat ileum. Activation of noncholinergic VIP secretomotor neurons by intrinsic primary afferent neurons and extrinsic afferent neurons by postprandially released xenin-25 may account for most of the neurogenic secretory response induced by xenin-25. NEW & NOTEWORTHY This study is the first to investigate the intrinsic neuronal circuit responsible for xenin-25-induced anion secretion in the rat small intestine. We have found that nutrient-stimulated xenin-25 release may activate noncholinergic vasoactive intestinal polypeptide (VIP) secretomotor neurons to promote Cl−/ HCO3− secretion through the activation of VIP receptor 1 on epithelial cells. Moreover, the xenin-25-induced secretory responses are mainly linked with intrinsic primary afferent neurons, which are involved in the activation of neurotensin receptor 1 and neurokinin 1 receptor. [ABSTRACT FROM AUTHOR]
- Published
- 2019
- Full Text
- View/download PDF
3. Role of VPAC1 and VPAC2 receptors in the etiology of pregnancy rhinitis: an experimental study in rats
- Author
-
Seda Vatansever, Burak Ulkumen, Sırrı Çam, Burcu Artunc Ulkumen, Muhammet Burak Batir, and Halil Gursoy Pala
- Subjects
Allergy ,medicine.medical_treatment ,Intraperitoneal injection ,Physiology ,Mucous membrane of nose ,03 medical and health sciences ,0302 clinical medicine ,Pregnancy ,Hypersensitivity ,medicine ,Animals ,Rats, Wistar ,030223 otorhinolaryngology ,Progesterone ,Rhinitis ,Subclinical infection ,VPAC2 ,030219 obstetrics & reproductive medicine ,VPAC1 ,Estradiol ,business.industry ,Vasoactive intestinal peptide receptor ,medicine.disease ,Pregnancy rhinitis ,Rats ,Otorhinolaryngology ,Gestation ,Female ,business ,Immunostaining - Abstract
Introduction Pregnancy rhinitis is a common sex hormone-related otorhinolaryngological disorder. There are some epidemiological and physiological studies on pregnancy rhinitis, but histopathological and biomolecular changes have not been studied thoroughly. Objectives The receptors VPAC1 and VPAC2 are known for their roles in allergic rhinitis. On the other hand, activation of subclinical allergy has been suggested in the pathophysiology of pregnancy rhinitis. Therefore, we aimed to compare the physiological and gestational pattern of VPAC1 and VPAC2 expression in rat nasal mucosa. Methods Twenty adult Wister albino female rats were enrolled into the study. Two groups constituted as 10 control (group A) and 10 pregnant (group B) rats. They were fed ad libitum and sheltered at room temperature (22°±2 °C). The rats were sacrificed at the 20th day of gestation by intraperitoneal injection of 400 mg/kg Na-pentobarbitone. Then, 10 − 15 mL of blood was taken, and samples were reserved for the detection of serum estradiol and progesterone levels by ELISA test. The nasal septum was resected and divided in half for immunohistochemical analyses and real time polymerase chain reaction testing of VPAC1 and VPAC2. Results VPAC1 and VPAC2 were found to be in all layers of septal specimens, but the immunostaining of surface epithelium was more distinct in specimens of both groups. We demonstrated higher overall staining intensity in the pregnant group. PCR revealed significant increase in expression of VPAC1 (p = 0.023) and VPAC2 (p = 0.021) in pregnant group when compared with control group. In addition, we demonstrated upregulatory effect of estradiol and progesterone on the vasoactive intestinal peptide receptor expression. Conclusions Gestational up-regulation of nasal VPAC1 and VPAC2 was shown both by PCR and immunohistochemical analysis. These findings support the hypothesis that PR is caused by the activation of subclinical allergy that is present before pregnancy.
- Published
- 2022
- Full Text
- View/download PDF
4. Vasoactive Intestinal Peptide: Immune Mediator and Potential Therapeutic Agent
- Author
-
Delgado, Mario, Ganea, Doina, and Levite, Mia, editor
- Published
- 2012
- Full Text
- View/download PDF
5. Expression of Somatostatin Receptors in Human Tissues in Health and Disease
- Author
-
Reubi, Jean Claude, Waser, Beatrice, Schaer, Jean-Claude, Melmed, Shlomo, editor, and Srikant, Coimbatore B., editor
- Published
- 2004
- Full Text
- View/download PDF
6. PACAP Receptor Signaling
- Author
-
Shioda, S., Zhou, C. JI, Ohtaki, H., Yada, T., Melmed, Shlomo, editor, Vaudry, Hubert, editor, and Arimura, Akira, editor
- Published
- 2003
- Full Text
- View/download PDF
7. Molecular Evolution of PACAP Precursor and PACAP Receptors
- Author
-
Chow, B. K-C, Pang, R. T-K, Ng, S. S-M, Melmed, Shlomo, editor, Vaudry, Hubert, editor, and Arimura, Akira, editor
- Published
- 2003
- Full Text
- View/download PDF
8. The Biological Significance of PACAP and PACAP Receptors in Human Tumors: From Cell Lines to Cancers
- Author
-
Lelièvre, V., Pineau, N., Waschek, J. A., Melmed, Shlomo, editor, Vaudry, Hubert, editor, and Arimura, Akira, editor
- Published
- 2003
- Full Text
- View/download PDF
9. Scintigraphy of Pancreatic Neuroendocrine Tumors and Carcinoids
- Author
-
Kwekkeboom, Dik J., de Herder, Wouter W., Krenning, Eric P., Melmed, Shlomo, editor, and de Herder, Wouter W., editor
- Published
- 2000
- Full Text
- View/download PDF
10. Peptide Imaging
- Author
-
Virgolini, I., Baert, A. L., editor, Heuck, F. H. W., editor, Youker, J. E., editor, and Schiepers, Christiaan, editor
- Published
- 2000
- Full Text
- View/download PDF
11. Different Bifidobacterium bifidum strains change the intestinal flora composition of mice via different mechanisms to alleviate loperamide-induced constipation
- Author
-
Jianxin Zhao, Hao Zhang, Wei Chen, Mao Chai, Xinping Li, Wang Gang, and Linlin Wang
- Subjects
0301 basic medicine ,medicine.medical_specialty ,Loperamide ,Constipation ,food.ingredient ,ved/biology.organism_classification_rank.species ,Population ,Biology ,03 medical and health sciences ,0302 clinical medicine ,food ,Internal medicine ,medicine ,education ,Receptor ,education.field_of_study ,Bifidobacterium bifidum ,ved/biology ,Vasoactive intestinal peptide receptor ,Anaerotruncus ,General Medicine ,030104 developmental biology ,Endocrinology ,030211 gastroenterology & hepatology ,medicine.symptom ,Food Science ,medicine.drug ,Ruminococcaceae - Abstract
Constipation is a condition with a high prevalence rate worldwide and may occur in men and women of any age. Bifidobacterium bifidum has been shown to have a relieving effect on constipation, but the underlying mechanism is still unknown. This study explored the effects of gavage of three strains of B. bifidum (CCFM668, FHNFQ25M12 and FXJCJ32M2) from different sources in mice with loperamide-induced constipation. After 38 days of intervention, B. bifidum CCFM668, FHNFQ25M12 and FXJCJ32M2 showed the ability to modify the levels of gastrointestinal active peptides and promote the expression of 5-hydroxytryptamine (5-HT or serotonin) receptor 4 (5-HT4R), thereby promoting small intestinal peristalsis. The strains could also effectively increase the thickness of the colonic mucosa. However, what was different from previous studies was that these results were independent of the levels of short-chain fatty acids (SCFAs) and 5-HT. Further analysis of the intestinal flora revealed that the relative abundances of the genera Faecalibaculum and Ruminococcaceae_UCG_014 in the constipated mice increased significantly, whereas that of Erysipelatoclostridium decreased. A correlation analysis between the intestinal flora and evaluated gastrointestinal indicators demonstrated that the relative abundances of the genera Anaerotruncus, Angelakisella, Erysipelatoclostridium and Ruminococcaceae_UCG_014 were negatively correlated with the levels of gastrointestinal active peptides. B. bifidum FXJCJ32M2 can increase the relative abundances of Turicibacter and Dubosiella, and this was positively correlated with the expression of aquaporin 8 and vasoactive intestinal peptide receptor 1 but could not effectively alleviate faecal dryness or promote colonic motility. These findings suggest that B. bifidum shows significant intraspecific differences in the remission mechanism and provides a theoretical basis for subsequent population experiments and personalised treatment for constipation.
- Published
- 2021
- Full Text
- View/download PDF
12. Preventive Treatment of Alzheimer’s Disease : Peptide-Mediated Neuroprotection
- Author
-
Gozes, Illana, Davidson, Ariane, Bachar, Michal, Bardea, Amos, Perl, Orly, Rubinraut, Sara, Fridkin, Mati, Giladi, Eliezer, Brenneman, Douglas E., Fisher, Abraham, editor, Hanin, Israel, editor, and Yoshida, Mitsuo, editor
- Published
- 1998
- Full Text
- View/download PDF
13. Noradrenergic and VIPergic Signal Transduction in Rat Pinealocytes
- Author
-
Schomerus, C., Korf, H.-W., Korf, Horst-Werner, editor, and Usadel, Klaus-Henning, editor
- Published
- 1997
- Full Text
- View/download PDF
14. Interaction of a Sulfhydryl Analogue of Vasoactive Intestinal Peptide (VIP) with Murine Lymphocytes
- Author
-
Ottaway, C. A., Mestecky, Jiri, editor, Russell, Michael W., editor, Jackson, Susan, editor, Michalek, Suzanne M., editor, Tlaskalová-Hogenová, Helena, editor, and Šterzl, Jaroslav, editor
- Published
- 1995
- Full Text
- View/download PDF
15. Immunocytochemistry and Molecular Biology in the Identification of Peptide-Containing Nerves
- Author
-
Springall, David R., Polak, Julia M., Raeburn, David, editor, and Giembycz, Mark A., editor
- Published
- 1994
- Full Text
- View/download PDF
16. Immunoregulation by Neuropeptides Other than Corticotropin and Melanotropin
- Author
-
Harbour, D. V., Scharrer, Berta, editor, Smith, Eric M., editor, and Stefano, George B., editor
- Published
- 1994
- Full Text
- View/download PDF
17. Role of Neuropeptides in the Bidirectional Communication Between the Immune and Neuroendocrine Systems
- Author
-
Weigent, D. A., Blalock, J. E., Scharrer, Berta, editor, Smith, Eric M., editor, and Stefano, George B., editor
- Published
- 1994
- Full Text
- View/download PDF
18. Gastrointestinal Peptides as Therapeutic Agents and Targets: Past, Present and Future
- Author
-
Gaginella, T. S. and Brown, David R., editor
- Published
- 1993
- Full Text
- View/download PDF
19. 748 Targeting vasoactive intestinal peptide receptor signaling in pancreatic ductal adenocarcinoma for enhanced anti-tumor response to checkpoint blockade
- Author
-
Edmund K. Waller, Maria Cardenas, Gregory B. Lesinski, Bassel F. El-Rayes, Jingru Zhu, Mohammad Y. Zaidi, Shuhua Wang, Brian S. Robinson, Michael B. Ware, Susan N. Thomas, Yuan Liu, Tenzin Passang Fnu, Shanmuganathan Chandrakasan, Sruthi Ravindranathan, Jian-Ming Li, Alan B. Frey, Rohan K. Dhamsania, Sanjeev Gumber, Anish Majumdar, and Haydn T. Kissick
- Subjects
Pharmacology ,Antitumor activity ,Cancer Research ,Pancreatic ductal adenocarcinoma ,business.industry ,Vasoactive intestinal peptide receptor ,Immunology ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,Blockade ,Oncology ,Cancer research ,Molecular Medicine ,Immunology and Allergy ,Medicine ,business ,RC254-282 - Abstract
BackgroundPaucity of T cells in the immune privileged tumor microenvironment (TME) of pancreatic ductal adenocarcinoma (PDAC) is a major reason that PDAC is refractory to immune checkpoint blockade.1 In this study, we show that human PDAC tumors over-express vasoactive intestinal peptide (VIP), an immunosuppressive neuropeptide, that inhibits effector T cell responses and regulates chemokine receptor expression on activated T cells.2 3 We thus hypothesized that pharmacological inhibition of VIP receptor signaling could enhance anti-tumor responses in PDAC.MethodsVIP levels in plasma were determined via VIP-specific enzyme immunoassay and confirmed with immunohistochemistry (IHC) of tissue sections. VIP receptor (VIP-R) signaling in C57BL/6 immunocompetent murine models of KPC, MT5 or Panc02 pancreatic cancer was inhibited by daily sub-cutaneous treatment with ANT008 or ANT308, two novel VIP-R antagonists with predicted high binding affinities to VIP receptors.4–7 Multiplex IHC or flow cytometry detected frequencies and phenotypes of intra-tumoral T cells across treatment groups.ResultsHuman PDAC tumors expressed VIP by immunohistochemistry, and PDAC patients had significantly elevated plasma VIP levels when compared to healthy volunteers (pConclusionsVIP-R antagonists represent a novel approach to treat PDAC. VIP and VIP-R sequences are highly conserved between humans and mice,8 and human T cells are activated in vitro following treatment with VIP-R antagonists. Thus, we predict comparable anti-tumor activity of the combination of VIP-R antagonist and anti-PD-1 MoAb in human PDAC patients. Further clinical development of this novel concept will require appropriate pre-clinical pharmacokinetic and toxicology studies.AcknowledgementsThe authors thank healthy volunteers and patients for blood and/or tissue samples. The authors also thank the shared resources at Emory University, namely the Emory Integrated Genomics Core (EIGC), Emory Flow Cytometry Core (EFCC), Cancer Animal Models Shared Resource (CAMS), Cancer Tissue Pathology Core (CTP), Biostatistics Shared Resource (BSR) and Integrated Cellular Imaging Core (ICI), that provided services or instruments at subsidized cost to conduct some of the reported experiments. BioRender was used to make figure 4A and 5C. This work was supported in part by Katz Foundation funding and Emory School of Medicine Dean's Imagine, Innovate and Impact (I3) venture award to Edmund K. Waller and NIH R01 CA207619 awarded to Susan N. Thomas. Part of the cost for the immunohistochemistry staining of tissues was covered by Winship Cancer Institute Development Discovery and Therapeutic Program Pilot funding to Sruthi Ravindranathan.Abstract 748 Figure 1VIP is over-expressed by PDAC. (A) VIP mRNA expression levels in various solid malignancies, as obtained from TCGA. (B) Representative images of human PDAC tumor stained with antibodies to VIP or CK19, showing VIP co-expression in islets (black arrow) and cancer epithelial cells (red arrow). Levels of VIP in (C) culture supernatants collected from murine and human PDAC cell lines cultured for 24 hours (n=3 per cell line) were compared to culture supernatants from B16F10 and D4M melanoma cells; (D) plasma of mice bearing melanoma or PDAC tumors (n=5) compared to plasma of non-tumor-bearing mice; (E) plasma of PDAC patients (n=19) compared to that from healthy volunteers (n=26). Statistical differences in C and D were performed by ANOVA followed by Dunnett's post-test and in E were performed by student's t-test. Error bars show mean ± SEM. *pAbstract 748 Figure 2VIP-R antagonists improve responses to anti-PD-1. KPC.Luc, MT5 or Panc02 cells were subcutaneously implanted in immunocompetent C57BL/6 mice. About one week after tumor implantation, when the tumors were palpable, mice were randomized into treatment groups and treated with VIP-R antagonist and/or anti-PD-1 as described in methods. (A) KPC.Luc, MT5 and Panc02 tumor volumes as measured by Vernier calipers on day 22 after subcutaneous tumor implantation. (B) Kaplan-Meier survival plots of C57BL/6 mice with subcutaneously implanted KPC.Luc, MT5 or Panc02 tumors stratified by treatment. Kaplan-Meier survival plots of (C) C57BL/6 mice receiving monoclonal CD4 and/or CD8 monoclonal antibodies (D) CD4KO or (E) CD8KO mice compared to wild-type CD57BL/6 mice with subcutaneously implanted KPC.Luc tumors, stratified by treatment. Statistical differences in A were calculated by ANOVA followed by Dunnett's post-test. Solid line shows mean with in each treatment group. Statistical differences in B-E are calculated via Log-rank test. *pAbstract 748 Figure 3Enhanced T cell response with combination therapy. mRNA expression in T cells isolated from subcutaneous KPC.Luc tumors in C57BL/6 mice treated with ANT008 and/or anti-PD-1 (n=3 per treatment group), were analyzed via Nanostring metabolism panel. Volcano plot showing differential expression of genes in T cells from (A) ANT008+ isotype IgG (IgG) vs scrambled peptide (Scram) + isotype IgG, (B) scrambled peptide +anti-PD-1 vs scrambled peptide + isotype IgG and (C) ANT008+anti-PD-1 vs scrambled peptide + isotype IgG (n=3 mice per treatment group). Genes that are associated with TCR activation and co-stimulation and are at levels significantly higher when compared to Scram+ isotype IgG (FDRAbstract 748 Figure 4Increased T cell density with combination therapy. KPC.Luc cells were orthotopically implanted in the tail of the pancreas of C57BL/6 mice and treated with ANT008 and/or anti-PD-1 with n=9, 10, 8 and 11 in scrambled+IgG, ANT008+IgG, scrambled+anti-PD-1 and ANT008+anti-PD-1, respectively. (A) Schematic showing orthotopic implantation of KPC.Luc cells and treatment strategy with ANT008 and/or anti-PD-1. (B) Waterfall plot showing % change in tumor flux on day 22 relative to day 7 prior to start of treatment. (C) Total flux as measured by IVIS bioluminescent imaging in the different treatment groups. Cross symbol represents mice that were euthanized before day 25 due to ulceration of the tumor and circle symbol represent mouse that were imaged on day 26 via MRI imaging shown in supplementary figure S5. (D) Bar graph showing weight of pancreas on day 25 when the mice were euthanized. 'Star' shaped data points indicate tumor free mice and dotted horizontal line represents the average weight of healthy pancreas from naïve mice. (E) Representative multiplex IHC images (right) showing pancreatic tumors stained for DAPI (blue), CD4 (yellow), CD8 (red) and Ki67 (cyan) and trichrome staining (left) with black arrows showing blue collagen stain in the tissue. XY plot showing the correlation between number of (F) CD4+ or (G) CD8+ T cells/mm2; and (H) Ki67+ CD4+ or (I) Ki67+ CD8+ T cells/mm2 with weight of the pancreas with n=4 to 6 mice per group. P values in panel D were calculated using student ANOVA followed by Dunnett's post hoc test (comparing each treatment group with Scram+IgG). Error bars show mean ± SEM. *pAbstract 748 Figure 5Increased T cell homing with combination therapy. KPC.Luc tumors were subcutaneously implanted in C57BL/6 mice and treated with VIP-R antagonist and/or anti-PD-1 checkpoint therapy for 10 days after the tumors were palpable. Tumor draining lymph nodes were then analyzed for percentage of (A) CXCR4+CD69+ and (B) CXCR4+Ki67+ cells in CD4+ (left) and CD8+ (right) subsets of T cells. In a separate experiment, on day 15 after subcutaneous implantation of KPC.Luc tumors, GFP+ T cells from enhanced GFP transgenic mice (C57BL/6 background) were adoptively transferred (via tail vein injections) and treated with ANT308± aPD-1 for 3 days. (C) Schematic showing GFP+ T cell transfer and treatment strategy in mice with subcutaneous KPC.Luc tumors. (D) Representative Hoescht (blue for nucleus) stained tumor tissues from tumors of each treatment group. Two regions of interest (ROI) in ANT308+aPD-1 treated tumors are shown at higher magnification. Statistical differences in A and B were determined via repeated measures ANOVA and Dunnett's post-test with n=4–5 mice per group. *pReferencesSahin IH, et al. Immunotherapy in pancreatic ductal adenocarcinoma: an emerging entity? Ann Oncol 2017;28(12):2950–2961.Gonzalez-Rey E, Anderson P, Delgado M. Emerging roles of vasoactive intestinal peptide: a new approach for autoimmune therapy. Ann Rheum Dis 2007;66(Suppl 3):p. iii70–6.Anderson P, Gonzalez-Rey E. Vasoactive intestinal peptide induces cell cycle arrest and regulatory functions in human T cells at multiple levels. Mol Cell Biol 2010;30(10):2537–51.Li JM, et al. VIPhyb, an antagonist of vasoactive intestinal peptide receptor, enhances cellular antiviral immunity in murine cytomegalovirus infected mice. PLoS One 2013;8(5):e63381.Moody TW, et al., VIP receptor antagonists and chemotherapeutic drugs inhibit the growth of breast cancer cells. Breast Cancer Res Treat 2001;68(1):55–64.Moody TW, et al. A vasoactive-Intestinal-Peptide antagonist inhibits nonsmall cell lung-cancer growth. Proceedings of the National Academy of Sciences of the United States of America 1993;90(10):4345–4349.Zia H, et al. Breast cancer growth is inhibited by vasoactive intestinal peptide (VIP) hybrid, a synthetic VIP receptor antagonist. Cancer Res 1996;56(15):3486–9.Sena M, et al. High conservation of upstream regulatory sequences on the human and mouse vasoactive intestinal peptide (VIP) genes. DNA Seq 1994;5(1):25–9.Ethics ApprovalAll experimental procedures involving mice were approved by the Institutional Animal Care and Use Committee (IACUC) at Emory University. De-identified blood samples from consented patients with PDAC (IRB 00087397) or healthy volunteers (IRB 00046063) were obtained with approval from Institutional Review Boards.
- Published
- 2021
20. AIDS as a Neuropeptide Disorder: Does HIV Bind to a VIP Receptor?
- Author
-
Hill, J. M., Kook, A. I., Harris, A., Bunney, W. E., Jr., editor, Hippius, Hanns, editor, Laakmann, Gregor, editor, and Schmauss, Max, editor
- Published
- 1990
- Full Text
- View/download PDF
21. The neuropeptide VIP confers anticipatory mucosal immunity by regulating ILC3 activity
- Author
-
Peter Hickey, Lachlan Whitehead, Kylie Luong, Kelly L. Rogers, Julie Tellier, Rui Dong Shen, Nicolas Jacquelot, Gabrielle T. Belz, Alexandra L. Garnham, Cyril Seillet, Matthew E. Ritchie, Gordon K. Smyth, and Verena C. Wimmer
- Subjects
0301 basic medicine ,Periodicity ,Immunology ,Vasoactive intestinal peptide ,Neuropeptide ,Eating ,Mice ,03 medical and health sciences ,0302 clinical medicine ,Animals ,Immunology and Allergy ,Lymphocytes ,Receptor ,Immunity, Mucosal ,Chemistry ,Vasoactive intestinal peptide receptor ,Innate lymphoid cell ,Intestinal epithelium ,Immunity, Innate ,Lymphocyte Subsets ,Cell biology ,Mice, Inbred C57BL ,030104 developmental biology ,Mucosal immunology ,Vasoactive Intestinal Peptide ,030215 immunology ,VIPR2 - Abstract
Group 3 innate lymphoid cell (ILC3)-mediated production of the cytokine interleukin-22 (IL-22) is critical for the maintenance of immune homeostasis in the gastrointestinal tract. Here, we find that the function of ILC3s is not constant across the day, but instead oscillates between active phases and resting phases. Coordinate responsiveness of ILC3s in the intestine depended on the food-induced expression of the neuropeptide vasoactive intestinal peptide (VIP). Intestinal ILC3s had high expression of the G protein-coupled receptor vasoactive intestinal peptide receptor 2 (VIPR2), and activation by VIP markedly enhanced the production of IL-22 and the barrier function of the epithelium. Conversely, deficiency in signaling through VIPR2 led to impaired production of IL-22 by ILC3s and increased susceptibility to inflammation-induced gut injury. Thus, intrinsic cellular rhythms acted in synergy with the cyclic patterns of food intake to drive the production of IL-22 and synchronize protection of the intestinal epithelium through a VIP-VIPR2 pathway in ILC3s.
- Published
- 2019
- Full Text
- View/download PDF
22. Generation of KS-133 as a Novel Bicyclic Peptide with a Potent and Selective VIPR2 Antagonist Activity that Counteracts Cognitive Decline in a Mouse Model of Psychiatric Disorders
- Author
-
Kotaro Sakamoto, Lu Chen, Tatsunori Miyaoka, Mei Yamada, Teruaki Masutani, Kenji Ishimoto, Nobumasa Hino, Shinsaku Nakagawa, Satoshi Asano, and Yukio Ago
- Subjects
medicine.medical_specialty ,Peptide ,RM1-950 ,KS-133 ,cyclic peptide ,In vivo ,Medicine ,Pharmacology (medical) ,bicyclization ,Cognitive decline ,Receptor ,Psychiatry ,Original Research ,chemistry.chemical_classification ,Pharmacology ,business.industry ,Vasoactive intestinal peptide receptor ,Antagonist ,antagonist ,VIPR2/VPAC2 ,schizophrenia ,chemistry ,Therapeutics. Pharmacology ,VIPR1 ,business ,VIPR2 - Abstract
Worldwide, more than 20 million people suffer from schizophrenia, but effective and definitive new therapeutic drugs/treatments have not been established. Vasoactive intestinal peptide receptor 2 (VIPR2) might be an attractive drug target for the treatment of schizophrenia because both preclinical and clinical studies have demonstrated a strong link between high expression/overactivation of VIPR2 and schizophrenia. Nevertheless, VIPR2-targeting drugs are not yet available. VIPR2 is a class-B G protein-coupled receptor that possesses high structural homology to its subtypes, vasoactive intestinal peptide receptor 1 (VIPR1) and pituitary adenylate cyclase-activating polypeptide type-1 receptor (PAC1). These biological and structural properties have made it difficult to discover small molecule drugs against VIPR2. In 2018, cyclic peptide VIpep-3, a VIPR2-selective antagonist, was reported. The aim of this study was to generate a VIpep-3 derivative for in vivo experiments. After amino acid substitution and structure optimization, we successfully generated KS-133 with 1) a VIPR2-selective and potent antagonistic activity, 2) at least 24 h of stability in plasma, and 3) in vivo pharmacological efficacies in a mouse model of psychiatric disorders through early postnatal activation of VIPR2. To the best of our knowledge, this is the first report of a VIPR2-selective antagonistic peptide that counteracts cognitive decline, a central feature of schizophrenia. KS-133 may contribute to studies and development of novel schizophrenia therapeutic drugs that target VIPR2.
- Published
- 2021
23. Protocatechuic acid influences immune-metabolic changes in the adipose tissue of pregnant women with gestational diabetes mellitus
- Author
-
Rosaria Varì, Tiziana Filardi, Paola Galoppi, Susanna Morano, Carmela Santangelo, Roberto Brunelli, Beatrice Scazzocchio, and Roberta Masella
- Subjects
0301 basic medicine ,Adult ,medicine.medical_specialty ,endocrine system diseases ,Metabolite ,Glucose uptake ,Adipose tissue ,030209 endocrinology & metabolism ,Inflammation ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,Immune system ,Pregnancy ,Internal medicine ,medicine ,Hydroxybenzoates ,Anticarcinogenic Agents ,Humans ,Adiponectin ,business.industry ,Vasoactive intestinal peptide receptor ,nutritional and metabolic diseases ,General Medicine ,medicine.disease ,female genital diseases and pregnancy complications ,Gestational diabetes ,Diabetes, Gestational ,030104 developmental biology ,Endocrinology ,chemistry ,Adipose Tissue ,Female ,medicine.symptom ,business ,gestational diabetes mellitus ,protocatechuic acid ,visceral adipose tissue ,Food Science - Abstract
Gestational diabetes mellitus (GDM) is associated with immune metabolic changes that increase women's risk of developing metabolic disorders later in life. Nutritional intervention is a crucial component in reducing the burden of these pathological features. We examined whether protocatechuic acid (PCA), a major metabolite of anthocyanins abundant in plant food, is able to exert insulin-mimetic activity and modulate inflammation in the visceral adipose tissue (VAT) obtained at delivery, from pregnant women with GDM or normal glucose tolerance (NGT). PCA stimulated glucose uptake in the VAT from both GDM and NGT women. This capability was associated with increased phosphorylation of p38 mitogen-activated protein kinase (p38MAPK), as further demonstrated by the inhibitory effect of SB203580, a p38MAPK inhibitor, on PCA-induced glucose uptake. The GDM-VAT expressed lower adiponectin levels and PCA stimulated adiponectin release in the NGT-VAT and, albeit to a lower extent, in the GDM-VAT. Higher levels of IL6 and TNFα were secreted by the GDM-VAT compared with the NGT one, and PCA had no effects on them. PCA reduced the overexpression of vasoactive intestinal peptide receptor 2 (VPAC2) in the GDM-VAT. Further studies are needed to establish whether and how anthocyanins and food rich in these compounds may contribute to prevent or delay metabolic disorders in women with GDM.
- Published
- 2021
24. Overexpressing miR-122-5p Inhibits the Relaxation of Vaginal Smooth Muscle in Female Sexual Arousal Disorder by Targeting Vasoactive Intestinal Peptide Receptor 1
- Author
-
Lianjun Pan, Aixia Zhang, Qinchuan Shi, Tao Gui, Jingjing Zhang, Jingyi Feng, Jiehua Ma, and Shengnan Cong
- Subjects
medicine.medical_specialty ,Urology ,Endocrinology, Diabetes and Metabolism ,Dermatology ,Cell morphology ,Other systems of medicine ,Behavioral Neuroscience ,chemistry.chemical_compound ,Endocrinology ,Basic Science ,Western blot ,Internal medicine ,miR-122-5p ,medicine ,Cyclic adenosine monophosphate ,Female sexual arousal disorder ,Protein kinase A ,Original Research ,medicine.diagnostic_test ,Cell growth ,Chemistry ,Vasoactive intestinal peptide receptor ,Vasoactive intestinal peptide receptor 1 ,Psychiatry and Mental health ,Reproductive Medicine ,Medicine ,Vaginal smooth muscle cell ,Erratum ,VIPR1 ,RZ201-999 ,Intracellular - Abstract
Introduction Female sexual arousal disorder (FSAD) is a common issue causing physical and psychological pain, but it has no standard diagnostic criteria or treatment. So its pathogenesis desiderates to be explored. Aim To investigate the specific function of miR-122-5p in FSAD. Methods 18 subjects were grouped into FSAD and normal control groups according to the Chinese version of the Female Sexual Function Index, and the expression levels of miR-122-5p and vasoactive intestinal peptide receptor 1 (VIPR1) protein in their tissue were verified through real-time quantitative polymerase chain reaction (qRT-PCR) and western blot (WB) analysis. Then in vitro experiment, miR-122-5p was overexpressed or inhibited in rat vaginal smooth muscle cells (SMCs). The relaxation of rat vaginal SMCs was reflected by the cell morphology, intracellular free cytosolic calcium ion (Ca2+) levels, cell proliferation and apoptosis, together with the cyclic adenosine monophosphate (cAMP) concentration and protein kinase A (PKA) activities. Additionally, the expression levels of relaxation-related proteins, including VIPR1, stimulatory G protein (Gs), adenylate cyclase (AC), and PKA, were detected based on WB analysis. Furthermore, a rescue experiment that simultaneously overexpressed or silenced miR-122-5p and VIPR1 was conducted, and all the indicators were evaluated. Main Outcomes Measure The expression level of VIPR1 and downstream proteins, cell morphology, cell proliferation and apoptosis, and intracellular free Ca2+ levels were examined. Results We verified that women with FSAD had higher miR-122-5p and lower VIPR1 protein. Then overexpressing miR-122-5p decreased relaxation of rat vaginal SMCs, which was manifested as a contractile morphology of cells, an increased intracellular free Ca2+ concentration, and lower cAMP concentration and PKA activity. Moreover, by rescue experiments, we inferred that VIPR1 was the target of miR-122-5p and affected the relaxation function of vaginal SMCs. Conclusion miR-122-5p regulates the relaxation of vaginal SMCs in FSAD by targeting VIPR1, ulteriorly providing an underlying diagnostic and therapeutic target for FSAD. Cong S, Gui T, Shi Q, et al. Overexpressing miR-122-5p Inhibits the Relaxation of Vaginal Smooth Muscle in Female Sexual Arousal Disorder by Targeting Vasoactive Intestinal Peptide Receptor 1. Sex Med 2021;9:100390.
- Published
- 2021
25. 819 Targeting vasoactive intestinal peptide receptor signaling: a novel approach to enhance anti-tumor response in pancreatic ductal adenocarcinoma
- Author
-
Shuhua Wang, Gregory B. Lesinski, Bassel F. El-Rayes, Rohan K. Dhamsania, Susan N. Thomas, Sanjay Chandrasekaran, Brandon Ware, Mohammad Y. Zaidi, Edmund K. Waller, Sruthi Ravindranathan, Passang Tenzin, Anish Majumdar, and Jingru Zhu
- Subjects
Tumor microenvironment ,business.industry ,medicine.medical_treatment ,Vasoactive intestinal peptide receptor ,Receptor expression ,T cell ,Vasoactive intestinal peptide ,Immunotherapy ,lcsh:Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,lcsh:RC254-282 ,medicine.anatomical_structure ,Cancer research ,medicine ,Cytotoxic T cell ,business ,CD8 - Abstract
Background Pancreatic ductal adenocarcinoma (PDAC), the fourth leading cause of cancer related death in the U.S, has a 5-year survival rate of only 10%.1 The paucity of T cells in the immune privileged tumor microenvironment (TME) is a major limitation in developing an effective immunotherapy against PDAC.2The cancer genome atlas (TCGA) shows that human PDAC tumors express high levels of vasoactive intestinal peptide (VIP), an immunosuppressive neuropeptide (figure 1A), that inhibits effector T cell responses.3 4 We hypothesized that paracrine secretion of VIP in the TME is a targetable mediator of immune paralysis in PDAC, and that pharmacological inhibition of VIP receptor signaling could enhance anti-tumor responses in PDAC. Methods VIP levels in plasma or cell culture supernatant was determined via VIP-specific enzyme immunoassay. Luciferase transfected KPC (KPC.luc) cells were injected subcutaneously or orthotopically into the pancreas of C57BL/6, CD4KO, or CD8KO mice from Jackson Laboratories. C57BL/6 mice T cell subsets in were depleted post tumor implantation with anti-CD4 and/or anti-CD8 antibodies. Tumor-bearing mice were treated daily with ANT008, a novel VIP receptor antagonist peptide, and/or anti-PD1 monoclonal antibody (MoAb) for 10 days, starting 7-10 days after implantation. T cells isolated from peripheral blood of PDAC patients were expanded 9 days ex vivo in anti-CD3 MoAb coated plates with 30U/ml IL-2 and either control peptide (scrambled VIP sequence) or ANT008. Survival by VIP and VIP receptor expression from the TCGA was clinically correlated. Results Increased human and mouse PDAC expression correlates with elevated blood levels (figure 1). While the PDAC cancer cell lines express VIP receptors, ANT008 does not have direct cytotoxic effect on cell growth in vitro (figure 2). However, in orthotopic KPC model, treatment with ANT008 & anti-PD-1 significantly decreased tumor growth rate and burden (figure 3) while increasing the intratumoral levels of CD4 and CD8 proliferating T cells (figure 4) via a T cell dependent mechanism (figure 5). Additionally, in ex vivo cultures of T cells isolated from PDAC patients, ANT008 improved the effector properties of T cells via decreasing expression levels of co-inhibitory molecules and decreasing frequency of regulatory T cells (figure 6). Clinically, VIPR1 receptor expression, but not VIP, provides a survival benefit (figure 7). Conclusions VIP is a targetable mechanism of immune escape in PDAC. Inhibiting VIP receptor signaling improves effector properties of T cells and synergistically improves the anti-tumor response to checkpoint inhibitors in mouse PDAC models. References Siegel RL, Miller KD, Jemal A. Cancer statistics, 2020. CA Cancer J Clin2020;70(1):7-30. Sahin IH, et al. Immunotherapy in pancreatic ductal adenocarcinoma: an emerging entity?Ann Oncol 2017;28(12):2950-2961. Gonzalez-Rey E, Anderson P, Delgado M. Emerging roles of vasoactive intestinal peptide: a new approach for autoimmune therapy. Ann Rheum Dis 2007;66(Suppl 3):iii70-6. Anderson P, Gonzalez-Rey E. Vasoactive intestinal peptide induces cell cycle arrest and regulatory functions in human T cells at multiple levels. Mol Cell Biol 2010;30(10):2537-51.
- Published
- 2020
- Full Text
- View/download PDF
26. Tamoxifen-inducible cardiac-specific Cre transgenic mouse using VIPR2 intron
- Author
-
Soyoung Lee, Hyun Jung Chin, and Daekee Lee
- Subjects
0301 basic medicine ,Genetically modified mouse ,lcsh:R5-920 ,ERT2CreERT2 ,Vasoactive intestinal peptide receptor ,Research ,Tamoxifen-inducible Cre transgenic mouse ,Mutant ,Intron ,VIPR2 intron ,Cre recombinase ,Heart ,Biology ,Cell biology ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,lcsh:Biology (General) ,Genetically Engineered Mouse ,lcsh:Medicine (General) ,lcsh:QH301-705.5 ,Gene ,030217 neurology & neurosurgery ,VIPR2 - Abstract
Genetically engineered mouse models through gene deletion are useful tools for analyzing gene function. To delete a gene in a certain tissue temporally, tissue-specific and tamoxifen-inducible Cre transgenic mice are generally used. Here, we generated transgenic mouse with cardiac-specific expression of Cre recombinase fused to a mutant estrogen ligand-binding domain (ERT2) on both N-terminal and C-terminal under the regulatory region of human vasoactive intestinal peptide receptor 2 (VIPR2) intron and Hsp68 promoter (VIPR2-ERT2CreERT2). In VIPR2-ERT2CreERT2 transgenic mice, mRNA for Cre gene was highly expressed in the heart. To further reveal heart-specific Cre expression, VIPR2-ERT2CreERT2 mice mated with ROSA26-lacZ reporter mice were examined by X-gal staining. Results of X-gal staining revealed that Cre-dependent recombination occurred only in the heart after treatment with tamoxifen. Taken together, these results demonstrate that VIPR2-ERT2CreERT2 transgenic mouse is a useful model to unveil a specific gene function in the heart.
- Published
- 2020
27. Synbiotic yogurt containing konjac mannan oligosaccharides and Bifidobacterium animalis ssp. lactis BB12 alleviates constipation in mice by modulating the stem cell factor (SCF)/c-Kit pathway and gut microbiota
- Author
-
Jun Liu, Li Tao, Jian Sun, Zhengqiang Jiang, Qiaojuan Yan, and Wen Yongping
- Subjects
Vasoactive intestinal peptide ,Oligosaccharides ,Substance P ,Stem cell factor ,Synbiotics ,Pharmacology ,Gut flora ,Motilin ,Mannans ,03 medical and health sciences ,chemistry.chemical_compound ,Mice ,Bifidobacterium animalis ,Genetics ,Animals ,030304 developmental biology ,0303 health sciences ,Stem Cell Factor ,biology ,Chemistry ,Vasoactive intestinal peptide receptor ,Probiotics ,0402 animal and dairy science ,food and beverages ,04 agricultural and veterinary sciences ,biology.organism_classification ,Yogurt ,040201 dairy & animal science ,Gastrointestinal Microbiome ,Animal Science and Zoology ,VIPR1 ,Constipation ,Food Science - Abstract
Synbiotic dietary supplements, as an effective means of regulating the gut microbiota, may have a beneficial effect on constipation. This study evaluated the effects of synbiotic yogurt containing konjac mannan oligosaccharides (KMOS) and Bifidobacterium animalis ssp. lactis BB12 (BB12) on constipated Kunming mice (the model group). Following administration of yogurt containing 2.0% KMOS and BB12 (YBK2.0), black fecal weight and number and gastrointestinal transit rate increased by 97.5, 106.3, and 55.7%, respectively, compared with the model group. Serum levels of excitability neurotransmitters (motilin, substance P, and acetylcholine) in the YBK2.0 group were increased by 139.7, 120.4, and 91.8%, respectively, and serum levels of inhibitory neurotransmitters (vasoactive intestinal peptide, nitric oxide, and acetylcholine) were decreased. Moreover, synbiotic yogurt supplementation significantly downregulated the expression of vasoactive intestinal peptide receptor 1 (VIPR1) and upregulated the expression of serotonin receptor 4 (5-HT4) in the colon, and enhanced the expression of the stem cell factor (SCF)/c-Kit pathway. Additionally, YBK2.0 treatment significantly regulated the community composition and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathways of gut microbiota, which were positively correlated with physiological parameters of constipation. Thus, supplementation with synbiotic yogurt composed of KMOS and BB12 could facilitate fecal excretion by regulating related pathways and the gut microbiota. These findings demonstrated that the synbiotic yogurt can be considered a functional food for alleviating constipation.
- Published
- 2020
28. Pharmacologic Characterization of ALD1910, a Potent Humanized Monoclonal Antibody against the Pituitary Adenylate Cyclase-Activating Peptide
- Author
-
Brian Baker, Dan Scott Allison, Lee Hendrix, David Jurchen, Susan Pederson, Heidi Boshaw, Sam Marzolf, Jens J. Billgren, John A. Latham, Vanessa Lisbeth Rubin, Gayle Kwon, Cristina Moldovan Loomis, Leon F. Garcia-Martinez, Pei Fan, Roger Cady, Erica Stewart, Ethan W. Ojala, Charlie Karasek, Jenny Mulligan, Michelle Scalley-Kim, Lisa Perrino McCulloch, and Benjamin H. Dutzar
- Subjects
Male ,0301 basic medicine ,medicine.drug_class ,Migraine Disorders ,Population ,Dose-Response Relationship, Immunologic ,Pharmacology ,Calcitonin gene-related peptide ,Antibodies, Monoclonal, Humanized ,Monoclonal antibody ,PC12 Cells ,Rats, Sprague-Dawley ,Epitopes ,03 medical and health sciences ,0302 clinical medicine ,Chronic Migraine ,Antibody Specificity ,Animals ,Humans ,Medicine ,education ,education.field_of_study ,business.industry ,Vasoactive intestinal peptide receptor ,medicine.disease ,Rats ,Kinetics ,Pituitary adenylate cyclase-activating peptide ,030104 developmental biology ,Migraine ,Calcitonin ,Pituitary Adenylate Cyclase-Activating Polypeptide ,Molecular Medicine ,business ,030217 neurology & neurosurgery - Abstract
Migraine is a debilitating disease that affects almost 15% of the population worldwide and is the first cause of disability in people under 50 years of age, yet its etiology and pathophysiology remain incompletely understood. Recently, small molecules and therapeutic antibodies that block the calcitonin gene-related peptide (CGRP) signaling pathway have reduced migraine occurrence and aborted acute attacks of migraine in clinical trials and provided prevention in patients with episodic and chronic migraine. Heterogeneity is present within each diagnosis and patient's response to treatment, suggesting migraine as a final common pathway potentially activated by multiple mechanisms, e.g., not all migraine attacks respond to or are prevented by anti-CGRP pharmacological interventions. Consequently, other unique mechanisms central to migraine pathogenesis may present new targets for drug development. Pituitary adenylate cyclase-activating peptide (PACAP) is an attractive novel target for treatment of migraines. We generated a specific, high-affinity, neutralizing monoclonal antibody (ALD1910) with reactivity to both PACAP38 and PACAP27. In vitro, ALD1910 effectively antagonizes PACAP38 signaling through the pituitary adenylate cyclase-activating peptide type I receptor, vasoactive intestinal peptide receptor 1, and vasoactive intestinal peptide receptor 2. ALD1910 recognizes a nonlinear epitope within PACAP and blocks its binding to the cell surface. To test ALD1910 antagonistic properties directed against endogenous PACAP, we developed an umbellulone-induced rat model of neurogenic vasodilation and parasympathetic lacrimation. In vivo, this model demonstrates that the antagonistic activity of ALD1910 is dose-dependent, retaining efficacy at doses as low as 0.3 mg/kg. These results indicate that ALD1910 represents a potential therapeutic antibody to address PACAP-mediated migraine.
- Published
- 2019
- Full Text
- View/download PDF
29. Design, synthesis and in vitro evaluation of heterobivalent peptidic radioligands targeting both GRP- and VPAC1-Receptors concomitantly overexpressed on various malignancies – Is the concept feasible?
- Author
-
Björn Wängler, Peter Bartenstein, Ralf Schirrmacher, Simon Lindner, Luise Fiedler, and Carmen Wängler
- Subjects
0301 basic medicine ,Pharmacology ,chemistry.chemical_classification ,010405 organic chemistry ,Chemistry ,Vasoactive intestinal peptide receptor ,Organic Chemistry ,Peptide ,General Medicine ,Prostate carcinoma ,01 natural sciences ,In vitro ,0104 chemical sciences ,03 medical and health sciences ,030104 developmental biology ,medicine.anatomical_structure ,Biochemistry ,Cell culture ,Prostate ,Drug Discovery ,Radioligand ,medicine ,Receptor - Abstract
Radiolabeled heterobivalent peptidic ligands (HBPLs), being able to address different receptors, are highly interesting tumor imaging agents as they can offer multiple advantages over monovalent peptide receptor ligands. However, few examples of radiolabeled HBPLs have been described so far. One promising approach is the combination of gastrin-releasing peptide receptor (GRPR)- and vasoactive intestinal peptide receptor subtype 1 (VPAC1R)-targeting peptides into one single radioligand since gastrinomas, prostate and breast cancer have been shown to concomitantly or complementarily overexpress both receptors. Here we report the design and synthesis of different HBPLs, comprising a GRPR-binding (BBN7-14) and a VPAC1R-targeting (PACAP-27) peptide. The heterodimers were varied with regard to the distance between the peptide binders and the steric rigidity of the systems. We radiolabeled the HBPLs 19–23 as well as their monomeric reference standards 26 and 27 with 68Ga, achieving radiochemical yields and purities of 95–99% and non-optimized molar activities of 25–61 GBq/μmol. We tested the stability of the radioligands and further evaluated them in vitro regarding their uptake in different prostate carcinoma cell lines (PC-3, DU-145 and VCaP cells). We found that the heterobivalent substances [68Ga]19 − [68Ga]23 showed comparable uptakes into the tumor cells to those of the respective monomers [68Ga]26 and [68Ga]27, indicating that both peptides are still able to address their target receptors. Furthermore, the obtained results indicate that in case of overall low receptor densities, heterobivalent peptides surpass peptide monomers in tumor cell uptake. Most importantly, it could be shown by blocking studies that both peptide parts of the HBPL [68Ga]19 contributed to tumor cell uptake in VCaP cells, expressing both receptor types. Thus, we describe here the first examples of HBPLs being able to address the GRPR as well as the VPAC1R and have the potential to − by several mechanisms − improve tumor targeting for several malignancies compared to monospecific peptides.
- Published
- 2018
- Full Text
- View/download PDF
30. PACAP causes PAC1/VPAC2 receptor mediated hypertension and sympathoexcitation in normal and hypertensive rats.
- Author
-
Farnham, M. M. J., Lung, M. S. Y., Tallapragada, V. J., and Pilowsky, P. M.
- Abstract
Pituitary adenylate cyclase- activating polypeptide (PACAP) is an excitatory neuropeptide that plays an important role in hypertension and stress responses. PACAP acts at three G protein-coupled receptors [PACAP type 1 receptor (PAC1) and vasoactive intestinal peptide receptor types 1 and 2 (VPAC1 and VPAC2)] and is localized to sites involved in cardiovascular control, most significantly the rostral ventrolateral medulla (RVLM). The RVLM is crucial for the tonic and reflex control of efferent sympathetic activity. Increases in sympathetic activity are observed in most types of hypertension and heart failure. PACAP delivered intrathecally also causes massive sympathoexcitation. We aimed to determine the presence and abundance of the three PACAP receptors in the RVLM, the role, in vivo, of PACAP in the RVLM on tonic and reflex cardiovascular control, and the contribution of PACAP to hypertension in the spontaneously hypertensive rat (SHR). Data were obtained using quantitative PCR and microinjection of PACAP and its antagonist, PACAP(6–38), into the RVLM of anesthetized artificially ventilated normotensive rats or SHRs. All three receptors were present in the RVLM. PACAP microinjection into the RVLM caused sustained sympathoexcitation and tachycardia with a transient hypertension but did not affect homeostatic reflexes. The responses were partially mediated through PAC1/VPAC2 receptors since the effect of PACAP was attenuated (~50%) by PACAP(6–38). PACAP was not tonically active in the RVLM in this preparation because PACAP(6–38) on its own had no inhibitory effect. PACAP has long-lasting cardiovascular effects, but altered PACAP signaling within the RVLM is not a cause of hypertension in the SHR. [ABSTRACT FROM AUTHOR]
- Published
- 2012
- Full Text
- View/download PDF
31. Abstract PO-072: Inhibiting vasoactive intestinal peptide receptor signaling elicits T cell dependent anti-tumor response of pancreatic ductal adenocarcinoma to immune checkpoint therapy
- Author
-
Susan N. Thomas, Haydn T. Kissick, Brian S. Robinson, Mohammad Y. Zaidi, Rohan K. Dhamsania, Gregory B. Lesinski, Alan B. Frey, Shuhua Wang, Sanjeev Gumber, Michael B. Ware, Jingru Zhu, Yuan Liu, Maria Cardenas, Passang Tenzin, Gaurav N. Joshi, Anish Sen-Majumdar, Shanmuganathan Chandrakasan, Sruthi Ravindranathan, Bassel F. El-Rayes, Jian-Ming Li, and Edmund K. Waller
- Subjects
Cancer Research ,Tumor microenvironment ,business.industry ,Vasoactive intestinal peptide receptor ,T cell ,Vasoactive intestinal peptide ,CXCR4 ,Immune checkpoint ,medicine.anatomical_structure ,Oncology ,Cancer research ,Medicine ,business ,Receptor ,CD8 - Abstract
Pancreatic ductal adenocarcinoma (PDAC) is unresponsive to immune checkpoint therapy largely due to a paucity of T cells within the tumor microenvironment (TME) and abundant immunosuppressive signaling pathways. In this study we show that human PDAC tumors over-express vasoactive intestinal peptide (VIP), an immunosuppressive neuropeptide that suppresses T cell effector properties and promotes the generation of regulatory T cells (Tregs). Therefore, we treated tumor-bearing mice with VIP receptor peptide antagonists and measured T cell homing, activation, and anti-tumor responses in preclinical murine models of PDAC. Pharmacological inhibition of VIP receptor (VIP-R) signaling using daily subcutaneous injections of peptide antagonists had no discernable toxicity in healthy and tumor-bearing mice. Treatment with VIP-R antagonists in combination with anti-PD-1 checkpoint blockade significantly decreased tumor burden, and improved survival in subcutaneous and orthotopic murine PDAC models. Combination therapy significantly enhanced T cell activation and proliferation and decreased frequencies of Tregs within the TME. Anti-tumor responses were T cell dependent, as the combination therapy failed to improve survival in CD4 or CD8 deficient mice using knock-out strains and antibody depletion. Furthermore, combination therapy significantly increased frequencies of tumor specific T cells (measured with a tetramer reagent) and provided protective immunity against tumor rechallenge. Combination therapy led to significant increases in the infiltration of adoptively transferred GFP+ T cells into PDAC tumors and decreased CXCR4 expression levels on T cells. Encouragingly, peptide-based VIP-R antagonists enhanced the in vitro activation of human T cells isolated from peripheral blood of PDAC patients. Human T cells cultured with VIP-R antagonists had increased proliferation, activation, and decreased proportions of T regs and exhausted T cells co-expressing PD-1, Tim-3 and Lag-3. Taken together, our findings show that VIP is a targetable mechanism of immune escape in PDAC. Inhibiting VIP receptor signaling improves T cell effector properties and synergistically improves anti-tumor responses to checkpoint inhibitors in mouse PDAC models. Additionally, as the VIP sequence is identical between human and mice, and since VIP-R antagonists have similar effects on human and murine T cells in culture, clinical translation is highly feasible. Citation Format: Sruthi Ravindranathan, Passang Tenzin, Jian Ming Li, Rohan Dhamsania, Michael Ware, Mohammad Zaidi, Shuhua Wang, Jingru Zhu, Maria Cardenas, Yuan Liu, Gaurav Joshi, Sanjeev Gumber, Brian Robinson, Anish Sen-Majumdar, Shanmuganathan Chandrakasan, Haydn Kissick, Alan Frey, Susan Thomas, Bassel El-Rayes, Gregory Lesinski, Edmund K. Waller. Inhibiting vasoactive intestinal peptide receptor signaling elicits T cell dependent anti-tumor response of pancreatic ductal adenocarcinoma to immune checkpoint therapy [abstract]. In: Proceedings of the AACR Virtual Special Conference on Pancreatic Cancer; 2021 Sep 29-30. Philadelphia (PA): AACR; Cancer Res 2021;81(22 Suppl):Abstract nr PO-072.
- Published
- 2021
- Full Text
- View/download PDF
32. Are heterobivalent GRPR- and VPAC1R-bispecific radiopeptides suitable for efficient in vivo tumor imaging of prostate carcinomas?
- Author
-
Peter Bartenstein, Ralph Hübner, Henning Rudolf, Carmen Wängler, Ralf Schirrmacher, Melissa Antons, Rosel Oos, Björn Wängler, Giovanna Palumbo, and Simon Lindner
- Subjects
Tumor imaging ,medicine.diagnostic_test ,Peptide receptor ,Chemistry ,Vasoactive intestinal peptide receptor ,Organic Chemistry ,Clinical Biochemistry ,Pharmaceutical Science ,Receptor type ,Biochemistry ,medicine.anatomical_structure ,In vivo ,Positron emission tomography ,Prostate ,Drug Discovery ,medicine ,Cancer research ,Molecular Medicine ,Clinical imaging ,Molecular Biology - Abstract
Receptor-specific peptides labeled with positron emitters play an important role in the clinical imaging of several malignancies by positron emission tomography (PET). Radiolabeled heterobivalent bispecific peptidic ligands (HBPLs) can target more than one receptor type and by this - besides exhibiting other advantages - increase tumor imaging sensitivity. In the present study, we show the initial in vivo evaluation of the most potent heterobivalent gastrin-releasing peptide receptor (GRPR)- and vasoactive intestinal peptide receptor subtype 1 (VPAC1R)-bispecific radiotracer and determined its tumor visualization potential via PET/CT imaging. For this purpose, the most potent described HBPL was synthesized together with its partly scrambled heterobivalent monospecific homologs and its monovalent counterparts. The agents were efficiently labeled with 68Ga3+ and evaluated in an initial PET/CT tumor imaging study in a human prostate carcinoma (PCa) xenograft rat tumor model established for this purpose. None of the three 68Ga-HBPLs enabled a clear tumor visualization and a considerably higher involvement in receptor-mediated uptake was found for the GRPR-binding part of the molecule than for the VPAC1R-binding one. Of the monovalent radiotracers, only [68Ga]Ga-NODA-GA-PESIN could efficiently delineate the tumor, confirming the results. Thus, this work sets the direction for future developments in the field of GRPR- and VPAC1R-bispecific radioligands, which should be based on other VPAC1R-specific peptides than PACAP-27.
- Published
- 2021
- Full Text
- View/download PDF
33. Inhibitory action of oxytocin on spontaneous contraction of rat distal colon by nitrergic mechanism: involvement of cyclic GMP and apamin-sensitive K+channels
- Author
-
C. Y. Liu, M. T. Han, Y. A. Yu, X. L. Lv, C. M. Qu, S. Q. Chai, and R. Wang
- Subjects
Male ,0301 basic medicine ,medicine.medical_specialty ,Potassium Channels ,Colon ,Physiology ,Nitric Oxide Synthase Type I ,Nitric Oxide ,Oxytocin ,Apamin ,03 medical and health sciences ,chemistry.chemical_compound ,Adenosine A1 receptor ,0302 clinical medicine ,Internal medicine ,medicine ,Animals ,PPADS ,Cyclic GMP ,Prostacyclin receptor ,Vasoactive intestinal peptide receptor ,Antagonist ,Muscle, Smooth ,Rats ,030104 developmental biology ,Endocrinology ,Gene Expression Regulation ,chemistry ,Receptors, Oxytocin ,Hexamethonium ,Soluble guanylyl cyclase ,030217 neurology & neurosurgery ,Muscle Contraction - Abstract
Aim The mechanisms underlying the inhibitory effects of oxytocin (OT) on colon tone are not totally understood. We explore the mechanisms of OT on spontaneous contractility in rat distal colon, and identify the mediators involved in this action. Methods In rat distal colon strips, mechanical activity was analyzed and the production of nitric oxide (NO) in tissue loaded with the fluorochrome DAF-FM was visualized by confocal microscopy. OT receptor (OTR) expression was determined by western blotting and immunofluorescence. Results In rat distal colon, OT produced a concentration-dependent reduction of the spontaneous contraction, which was abolished by the OTR antagonist atosiban, the neural blocker tetrodotoxin and the inhibitor of neuronal nitric oxide synthase (nNOS) NPLA. The inhibitory effects of OT were not affected by propranolol, atropine, the nicotinic cholinoceptor blocker hexamethonium, the vasoactive intestinal peptide receptor antagonist VIPHyb, the P2 purinoceptor antagonist PPADS, the adenosine A1 receptors antagonist DPCPX and the prostacyclin receptor antagonist Ro1138452. The soluble guanylyl cyclase (sGC) inhibitor ODQ and the small conductance Ca2+-activated K+ (CaK+) channels blocker apamin significantly reduced the relaxation induced by OT, nicotine, sodium nitroprusside and the sGC activator BAY 41-2272. The neural release of NO elicited by OT was prevented by NPLA, tetrodotoxin, and atosiban. The presence of the OTR and its co-localization with nNOS was detected by immunohistochemistry and western blotting experiments. Conclusion These results demonstrate the NO release from enteric neurons induced by activation of OTR mediates distal colon relaxation. sGC and small conductance CaK+ channels are involved in this relaxation. This article is protected by copyright. All rights reserved.
- Published
- 2017
- Full Text
- View/download PDF
34. The palmitoylation of the N-terminal extracellular Cys37 mediates the nuclear translocation of VPAC1 contributing to its anti-apoptotic activity
- Author
-
Yue Cui, An Hong, Rongjie Yu, Huahua Zhang, Hongyu Liu, Suqin Song, Like Wang, Xinhe Peng, and Tianhong Zhou
- Subjects
0301 basic medicine ,Lipoylation ,nuclear translocation ,Vasoactive intestinal peptide ,Apoptosis ,CHO Cells ,Transfection ,03 medical and health sciences ,Cricetulus ,Palmitoylation ,Extracellular ,Animals ,palmitoylation ,Cysteine ,Receptor ,Cells, Cultured ,G protein-coupled receptor ,vasoactive intestinal peptide receptor 1 (VPAC1) ,Chemistry ,Vasoactive intestinal peptide receptor ,Chinese hamster ovary cell ,cysteine (Cys) ,Molecular biology ,Fusion protein ,030104 developmental biology ,Oncology ,Biochemistry ,anti-apoptotic activity ,Vasoactive Intestinal Peptide ,Research Paper - Abstract
// Rongjie Yu 1, 2 , Hongyu Liu 1, 2 , Xinhe Peng 1, 2 , Yue Cui 1, 2 , Suqin Song 1, 2 , Like Wang 1, 2 , Huahua Zhang 3 , An Hong 1, 2 and Tianhong Zhou 4 1 Institute of Biomedicine, School of Life Science and Technology, Jinan University, Guangzhou, Guangdong, China 2 National Engineering Research Center of Genetic Medicine, Jinan University, Guangzhou, Guangdong, China 3 Department of Medical Genetics, Guangdong Medical University, Dongguan, Guangdong, China 4 Department of Bioengineering, School of Life Science and Technology, Jinan University, Guangzhou, Guangdong, China Correspondence to: Rongjie Yu, email: rongjie_yu1123@163.com Keywords: vasoactive intestinal peptide receptor 1 (VPAC1), cysteine (Cys), palmitoylation, nuclear translocation, anti-apoptotic activity Received: March 06, 2017 Accepted: April 14, 2017 Published: April 27, 2017 ABSTRACT VPAC1 is class B G protein-coupled receptors (GPCR) shared by pituitary adenylate cyclase activating polypeptide (PACAP) and vasoactive intestinal peptide (VIP). The first cysteine (Cys37) in the N-terminal extracellular domain of mature VPAC1 is a free Cys not involved in the formation of conserved intramolecular disulfide bonds. In order to investigate the biological role of this Cys37 in VPAC1, the wild-type VPAC1 and Cys37/Ala mutant (VPAC1-C37/A) were expressed stably as fusion proteins with enhanced yellow fluorescent protein (EYFP) respectively in Chinese hamster ovary (CHO) cells. Both VPAC1-EYFP and VPAC1-C37/A-EYFP trafficked to the plasma membrane normally, and CHO cells expressing VPAC1-EYFP displayed higher anti-apoptotic activity against camptothecin (CPT) induced apoptosis than the cells expressing VPAC1-C37/A-EYFP, while VPAC1-C37/A-CHO cells showed higher proliferative activity than VPAC1-CHO cells. Confocal microscopic analysis, western blotting and fluorescence quantification assay showed VPAC1-EYFP displayed significant nuclear translocation while VPAC1-C37/A-EYFP did not transfer into nucleus under the stimulation of VIP (0.1 nM). Acyl-biotin exchange assay and click chemistry-based palmitoylation assay confirmed for the first time the palmitoylation of Cys37, which has been predicted by bioinformatics analysis. And the palmitoylation inhibitor 2-bromopalmitate significantly inhibited the nuclear translocation of VPAC1-EYFP and its anti-apoptotic activity synchronously. These results indicated the palmitoylation of the Cys37 in the N-terminal extracellular domain of VPAC1 mediates the nuclear translocation of VPAC1 contributing to its anti-apoptotic activity. These findings reveal for the first time the lipidation-mediating nuclear translocation of VPAC1 produces a novel anti-apoptotic signal pathway, which may help to promote new drug development strategy targeting VPAC1.
- Published
- 2017
- Full Text
- View/download PDF
35. Activation of the vasoactive intestinal peptide 2 receptor modulates normal and atrophying skeletal muscle mass and force.
- Author
-
Hinkle, Richard T., Donnelly, Elizabeth, Cody, David B., Sheldon, Russell J., and Isfort, Robert J.
- Subjects
VASOACTIVE intestinal peptide ,HYPERTROPHY ,GASTROINTESTINAL hormones ,NEUROTRANSMITTERS ,PEPTIDES ,PHYSIOLOGY - Abstract
Of the two known vasoactive intestinal peptide receptors (VPAC1R and VPAC2R), the VPAC2R is expressed in skeletal muscle. To evaluate the function of the VPAC2R in the physiological control of skeletal muscle mass, we utilized the VPAC1R selective agonist [K
15 ,R16 ,L27 ] VIP(1–7) GRF(8–27)-NH2 and the VPAC2R selective agonist Ro-25–1553 to treat mice and rats undergoing either nerve damage-, corticosteroid-, or disuse-induced skeletal muscle atrophy. These analyses demonstrated that activation of VPAC2R, but not VPAC I R, reduced the loss of skeletal muscle mass and force during conditions of skeletal muscle atrophy resulting from corticosteroid administration, denervation, casting-induced disuse, increased skeletal muscle mass, and force of nonatrophying muscles. These studies indicate that VPAC2R agonists may have utility for the treatment of skeletal muscle-wasting diseases. [ABSTRACT FROM AUTHOR]- Published
- 2005
- Full Text
- View/download PDF
36. Identification of G-Proteins Coupling to the Vasoactive Intestinal Peptide Receptor VPAC1 Using Immunoaffinity Chromatography: Evidence for Precoupling
- Author
-
Shreeve, S. Martin
- Subjects
- *
G proteins , *PROTEIN crosslinking - Abstract
VPAC1 receptor subtype-specific G-protein interactions were identified using a strategy that exploits an essential initial signaling event, namely the functional and physical association of the receptor with G-protein. An immunoaffinity purification column was constructed using a previously characterized antibody that had been raised against the first extracellular loop of the VPAC1 receptor. VPAC1/G-protein complexes were solubilized from membranes and copurified. Receptor and Gα-proteins were detected in eluates using 125I-VIP labeling and immunoblotting, respectively. Human VPAC1 transfected in HEK293 cells couples to Gs but not Gi3, Gi1/2, or Gq. Rat VPAC1 in brain membranes is coupled to Gs and Gi3. Rat VPAC1 in lung membranes couples to Gs, Gi3, and Gq. Pretreatment of membranes with VIP increased the level of all G-proteins copurifying with VPAC1. Immunoaffinity chromatography also revealed VPAC1 receptor precoupling to G-protein in the absence of VIP pretreatment. This was confirmed using a cross-linking procedure to capture VIP receptor/G-protein complexes in the native membrane milieu prior to solubilization. Precoupling suggests that there is a significant basal level of VPAC1 receptor activity especially in cells, such as some human malignant tumor cells, that express high levels of receptor. [Copyright &y& Elsevier]
- Published
- 2002
- Full Text
- View/download PDF
37. Vasoactive intestinal peptide regulates ileal goblet cell production in mice
- Author
-
Luke A. Schwerdtfeger and Stuart A. Tobet
- Subjects
Male ,Physiology ,Vasoactive intestinal peptide ,Peripherins ,Enteroendocrine cell ,Tetrodotoxin ,030204 cardiovascular system & hematology ,digestive system ,lcsh:Physiology ,Signalling Pathways ,03 medical and health sciences ,Mice ,0302 clinical medicine ,Intestinal mucosa ,Physiology (medical) ,medicine ,Neural Circuits and Systems ,Animals ,intestine ,mucosa ,Cell Proliferation ,organotypic ,Neurons ,Goblet cell ,lcsh:QP1-981 ,Cell growth ,Chemistry ,goblet cell ,Vasoactive intestinal peptide receptor ,Peripherin ,Original Articles ,respiratory system ,Mucus ,3. Good health ,Cell biology ,Mice, Inbred C57BL ,VIP ,medicine.anatomical_structure ,Gastrointestinal, Hepatic and Pancreatic Physiology ,Receptors, Vasoactive Intestinal Peptide ,Female ,Original Article ,Goblet Cells ,030217 neurology & neurosurgery ,Vasoactive Intestinal Peptide - Abstract
Innervation of the intestinal mucosa has gained more attention with demonstrations of tuft and enteroendocrine cell innervation. However, the role(s) these fibers play in maintaining the epithelial and mucus barriers are still poorly understood. This study therefore examines the proximity of mouse ileal goblet cells to neuronal fibers, and the regulation of goblet cell production by vasoactive intestinal peptide (VIP). An organotypic intestinal slice model that maintains the cellular diversity of the intestinal wall ex vivo was used. An ex vivo copper‐free click‐reaction to label glycosaminoglycans was used to identify goblet cells. Pharmacological treatment of slices was used to assess the influence of VIP receptor antagonism on goblet cell production and neuronal fiber proximity. Goblet cells were counted and shown to have at least one peripherin immunoreactive fiber within 3 µm of the cell, 51% of the time. Treatment with a VIP receptor type I and II antagonist (VPACa) resulted in an increase in the percentage of goblet cells with peripherin fibers. Pharmacological treatments altered goblet cell counts in intestinal crypts and villi, with tetrodotoxin and VPACa substantially decreasing goblet cell counts. When cultured with 5‐Ethynyl‐2’‐deoxyuridine (EdU) as an indicator of cell proliferation, colocalization of labeled goblet cells and EdU in ileal crypts was decreased by 77% when treated with VPACa. This study demonstrates a close relationship of intestinal goblet cells to neuronal fibers. By using organotypic slices from mouse ileum, vasoactive intestinal peptide receptor regulation of gut wall goblet cell production was revealed., A close relationship between neuronal fibers and enteric goblet cells was demonstrated, with more neuronal fibers being in close proximity to goblet cells in vasoactive intestinal peptide (VIP) receptor antagonized ileum slices. Further, antagonizing VIP receptors ex vivo was shown to regulate goblet cell production in the ileal crypt.
- Published
- 2019
38. A Synthetic Agonist to Vasoactive Intestinal Peptide Receptor-2 Induces Regulatory T Cell Neuroprotective Activities in Models of Parkinson’s Disease
- Author
-
Krista L. Namminga, Scott Shandler, R. Lee Mosley, Jenell R. Smith, Jatin Machhi, Yaman Lu, Wenhui Yan, Howard E. Gendelman, and Katherine E. Olson
- Subjects
0301 basic medicine ,Agonist ,Regulatory T cell ,medicine.drug_class ,alpha-synuclein ,Vasoactive intestinal peptide ,microglia ,6-hydroxydopamine ,Pharmacology ,Neuroprotection ,regulatory T cells ,lcsh:RC321-571 ,03 medical and health sciences ,Cellular and Molecular Neuroscience ,0302 clinical medicine ,medicine ,lcsh:Neurosciences. Biological psychiatry. Neuropsychiatry ,agonist ,Neuroinflammation ,Original Research ,vasoactive intestinal peptide ,business.industry ,Vasoactive intestinal peptide receptor ,Dopaminergic ,Neurodegeneration ,neurodegeneration ,medicine.disease ,030104 developmental biology ,medicine.anatomical_structure ,Parkinson’s disease ,business ,030217 neurology & neurosurgery ,Neuroscience - Abstract
A paradigm shift has emerged in Parkinson’s disease (PD) highlighting the prominent role of CD4+ Tregs in pathogenesis and treatment. Bench to bedside research, conducted by others and our own laboratories, advanced a neuroprotective role for Tregs making pharmacologic transformation of immediate need. Herein, a vasoactive intestinal peptide receptor-2 (VIPR2) peptide agonist, LBT-3627, was developed as a neuroprotectant for PD-associated dopaminergic neurodegeneration. Employing both 6-hydroxydopamine (6-OHDA) and α-synuclein (α-Syn) overexpression models in rats, the sequential administration of LBT-3627 increased Treg activity without altering cell numbers both in naïve animals and during progressive nigrostriatal degeneration. LBT-3627 administration was linked to reductions of inflammatory microglia, increased survival of dopaminergic neurons, and improved striatal densities. While α-Syn overexpression resulted in reduced Treg activity, LBT-3627 rescued these functional deficits. This occurred in a dose-dependent manner closely mimicking neuroprotection. Taken together, these data provide the basis for the use of VIPR2 agonists as potent therapeutic immune modulating agents to restore Treg activity, attenuate neuroinflammation, and interdict dopaminergic neurodegeneration in PD. The data underscore an important role of immunity in PD pathogenesis.
- Published
- 2019
39. A Molecular Dynamics Study of Vasoactive Intestinal Peptide Receptor 1 and the Basis of Its Therapeutic Antagonism
- Author
-
Lukasz Charzewski, Dorota Latek, Krystiana A. Krzysko, and Ingrid Langer
- Subjects
Protein Conformation ,homology modeling ,Quantitative Structure-Activity Relationship ,Ligands ,PACAP ,lcsh:Chemistry ,G protein-coupled receptors ,Receptor ,lcsh:QH301-705.5 ,agonist ,Spectroscopy ,Molecular Structure ,VPAC1 ,Chemistry ,Drug discovery ,Vasoactive intestinal peptide receptor ,Biological activity ,General Medicine ,Computer Science Applications ,Cell biology ,Molecular Docking Simulation ,GPCRM ,medicine.symptom ,VIPR1 ,Protein Binding ,Receptors, Vasoactive Intestinal Polypeptide, Type I ,Molecular Dynamics Simulation ,Catalysis ,Article ,Inorganic Chemistry ,Pharmacologie moléculaire et cellulaire ,medicine ,Humans ,Homology modeling ,Physical and Theoretical Chemistry ,Molecular Biology ,G protein-coupled receptor ,Binding Sites ,Organic Chemistry ,gut hormone receptors ,antagonist ,molecular dynamics ,VIP ,Mechanism of action ,lcsh:Biology (General) ,lcsh:QD1-999 ,vasoactive intestinal peptide receptor 1 ,Drug Design ,GPCR activation - Abstract
Vasoactive intestinal peptide receptor 1 (VPAC1) is a member of a secretin-like subfamily of G protein-coupled receptors. Its endogenous neuropeptide (VIP), secreted by neurons and immune cells, modulates various physiological functions such as exocrine and endocrine secretions, immune response, smooth muscles relaxation, vasodilation, and fetal development. As a drug target, VPAC1 has been selected for therapy of inflammatory diseases but drug discovery is still hampered by lack of its crystal structure. In this study we presented the homology model of this receptor constructed with the well-known web service GPCRM. The VPAC1 model is composed of extracellular and transmembrane domains that form a complex with an endogenous hormone VIP. Using the homology model of VPAC1 the mechanism of action of potential drug candidates for VPAC1 was described. Only two series of small-molecule antagonists of confirmed biological activity for VPAC1 have been described thus far. Molecular docking and a series of molecular dynamics simulations were performed to elucidate their binding to VPAC1 and resulting antagonist effect. The presented work provides the basis for the possible binding mode of VPAC1 antagonists and determinants of their molecular recognition in the context of other class B GPCRs. Until the crystal structure of VPAC1 will be released, the presented homology model of VPAC1 can serve as a scaffold for drug discovery studies and is available from the author upon request., info:eu-repo/semantics/published
- Published
- 2019
40. Xenin-25 induces anion secretion by activating noncholinergic secretomotor neurons in the rat ileum
- Author
-
Yoshinori Marunaka, Toshio Inui, Daiki Harata, Ikuo Kato, Shinji Asano, Atsukazu Kuwahara, Kotoku Kawaguchi, and Yuko Kuwahara
- Subjects
0301 basic medicine ,Anions ,Physiology ,Receptors, Vasoactive Intestinal Polypeptide, Type I ,Ileum ,Secretomotor ,Xenin ,Rat Small Intestine ,Enteric Nervous System ,Gastrointestinal Hormones ,03 medical and health sciences ,0302 clinical medicine ,Physiology (medical) ,Neural Pathways ,medicine ,Animals ,Receptors, Neurotensin ,Secretion ,Intestinal Mucosa ,Neurotensin ,Hepatology ,Chemistry ,Vasoactive intestinal peptide receptor ,Gastroenterology ,Cell biology ,Rats ,030104 developmental biology ,medicine.anatomical_structure ,Enteric nervous system ,030217 neurology & neurosurgery - Abstract
Xenin-25 is a neurotensin-like peptide that is secreted by enteroendocrine cells in the small intestine. Xenin-8 is reported to augment duodenal anion secretion by activating afferent neural pathways. The intrinsic neuronal circuits mediating the xenin-25-induced anion secretion were characterized using the Ussing-chambered, mucosa-submucosa preparation from the rat ileum. Serosal application of xenin-25 increased the short-circuit current in a concentration-dependent manner. The responses were abolished by the combination of Cl−-free and [Formula: see text]-free solutions. The responses were almost completely blocked by TTX (10−6M) but not by atropine (10−5M) or hexamethonium (10−4M). The selective antagonists for neurotensin receptor 1 (NTSR1), neurokinin 1 (NK1), vasoactive intestinal polypeptide (VIP) receptors 1 and 2 (VPAC1 and VPAC2, respectively), and capsaicin, but not 5-hydroxyltryptamine receptors 3 and 4 (5-HT3and 5-HT4), NTSR2, and A803467, inhibited the responses to xenin-25. The expression of VIP receptors ( Vipr) in rat ileum was examined using RT-PCR. The Vipr1 PCR products were detected in the submucosal plexus and mucosa. Immunohistochemical staining showed the colocalization of NTSR1 and NK1 with substance P (SP)- and calbindin-immunoreactive neurons in the submucosal plexus, respectively. In addition, NK1 was colocalized with noncholinergic VIP secretomotor neurons. Based on the results from the present study, xenin-25-induced Cl−/[Formula: see text] secretion is involved in NTSR1 activation on intrinsic and extrinsic afferent neurons, followed by the release of SP and subsequent activation of NK1 expressed on noncholinergic VIP secretomotor neurons. Finally, the secreted VIP may activate VPAC1 on epithelial cells to induce Cl−/[Formula: see text] secretion in the rat ileum. Activation of noncholinergic VIP secretomotor neurons by intrinsic primary afferent neurons and extrinsic afferent neurons by postprandially released xenin-25 may account for most of the neurogenic secretory response induced by xenin-25.NEW & NOTEWORTHY This study is the first to investigate the intrinsic neuronal circuit responsible for xenin-25-induced anion secretion in the rat small intestine. We have found that nutrient-stimulated xenin-25 release may activate noncholinergic vasoactive intestinal polypeptide (VIP) secretomotor neurons to promote Cl−/[Formula: see text] secretion through the activation of VIP receptor 1 on epithelial cells. Moreover, the xenin-25-induced secretory responses are mainly linked with intrinsic primary afferent neurons, which are involved in the activation of neurotensin receptor 1 and neurokinin 1 receptor.
- Published
- 2019
41. T
- Author
-
Geenen, Vincent, Robert, Françoise, Ericsson, Anders, Persson, Hakan, Smith, Barry, editor, and Adelman, George, editor
- Published
- 1992
- Full Text
- View/download PDF
42. Role of pituitary adenylyl cyclase-activating polypeptide in intracellular calcium dynamics of neurons and satellite cells in rat superior cervical ganglia
- Author
-
Kanako Isobe, Tomoyuki Saino, Miho Kumagai, Takuya Yokoyama, Akiyoshi Kuji, Yoh-ichi Satoh, and Kasumi Moriguchi-Mori
- Subjects
0301 basic medicine ,Nervous system ,medicine.medical_specialty ,Vasoactive intestinal peptide receptor ,Stimulation ,General Medicine ,General Biochemistry, Genetics and Molecular Biology ,Calcium in biology ,Cell biology ,Adenylyl cyclase ,03 medical and health sciences ,chemistry.chemical_compound ,030104 developmental biology ,0302 clinical medicine ,medicine.anatomical_structure ,Endocrinology ,chemistry ,Internal medicine ,medicine ,Extracellular ,Receptor ,030217 neurology & neurosurgery ,Intracellular - Abstract
Pituitary adenylyl cyclase-activating polypeptide (PACAP) is a bioactive peptide with diverse effects in the nervous system. The present study investigated whether stimulation of PACAP receptors (PACAPRs) induces responses in neurons and satellite cells of the superior cervical ganglia (SCG), with special reference to intracellular Ca2+ ([Ca2+]i) changes. The expression of PACAPRs in SCG was detected by reverse transcription-PCR. PACAP type 1 receptor (PAC1R), vasoactive intestinal peptide receptor type (VPAC)1R, and VPAC2R transcripts were expressed in SCG, with PAC1R showing the highest levels. Confocal microscopy analysis revealed that PACAP38 and PACAP27 induced an increase in [Ca2+]i in SCG, first in satellite cells and subsequently in neurons. Neither extracellular Ca2+ removal nor Ca2+ channel blockade affected the PACAP38-induced increase in [Ca2+]i in satellite cells; however, this was partly inhibited in neurons. U73122 or xestospongin C treatment completely and partly abrogated [Ca2+]i changes in satellite cells and in neurons, respectively, whereas VPAC1R and VPAC2R agonists increased [Ca2+]i in satellite cells only. This is the first report demonstrating the expression of PACAPRs specifically, VPAC1 and VPAC2 in SCG and providing evidence for PACAP38-induced [Ca2+]i changes in both satellite cells and neurons via Ca2+ mobilization.
- Published
- 2017
- Full Text
- View/download PDF
43. Manganese-Enhanced Magnetic Resonance Imaging for Detection of Vasoactive Intestinal Peptide Receptor 2 Agonist Therapy in a Model of Parkinson’s Disease
- Author
-
Yutong Liu, Charles R. Schutt, Jingdong Dong, Howard E. Gendelman, Michael D. Boska, Aditya N. Bade, Katherine E. Olson, Scott Shandler, and R. Lee Mosley
- Subjects
Male ,0301 basic medicine ,Agonist ,Parkinson's disease ,medicine.drug_class ,Biology ,Neuroprotection ,Mice ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,Parkinsonian Disorders ,medicine ,Animals ,Pharmacology (medical) ,Pharmacology ,Manganese ,Tyrosine hydroxylase ,Dopaminergic Neurons ,Vasoactive intestinal peptide receptor ,MPTP ,Brain ,Parkinson Disease ,Protein-Tyrosine Kinases ,Image Enhancement ,medicine.disease ,Magnetic Resonance Imaging ,Astrogliosis ,Biomarker (cell) ,Mice, Inbred C57BL ,Neuroprotective Agents ,030104 developmental biology ,chemistry ,Receptors, Vasoactive Intestinal Peptide, Type II ,Original Article ,Microglia ,Neurology (clinical) ,Oligopeptides ,Neuroscience ,030217 neurology & neurosurgery - Abstract
Neuroprotective immunity is defined by transformation of T-cell polarity for therapeutic gain. For neurodegenerative disorders and specifically for Parkinson's disease (PD), granulocyte-macrophage colony stimulating factor or vasoactive intestinal peptide receptor 2 (VIPR2) agonists elicit robust anti-inflammatory microglial responses leading to neuronal sparing in 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-intoxicated mice. While neurotherapeutic potential was demonstrated for PD, there remain inherent limitations in translating these inventions from the laboratory to patients. One obstacle in translating such novel neurotherapeutics centers on the availability of suitable noninvasive methods to track disease progression and therapeutic efficacy. To this end, we developed manganese-enhanced magnetic resonance imaging (MEMRI) assays as a way to track a linkage between glial activation and VIPR2 agonist (LBT-3627)-induced neuroprotective immunity for MPTP-induced nigrostriatal degeneration. Notably, LBT-3627-treated, MPTP-intoxicated mice show reduced MEMRI brain signal intensities. These changes paralleled reduced astrogliosis and resulted in sparing of nigral tyrosine hydroxylase neurons. Most importantly, the data suggest that MEMRI can be developed as a biomarker tool to monitor neurotherapeutic responses that are relevant to common neurodegenerative disorders used to improve disease outcomes.
- Published
- 2016
- Full Text
- View/download PDF
44. Regulation of vasoactive intestinal peptide receptor (VPAC) signaling in Undifferentiated hen granulosa cells
- Author
-
Alan L. Johnson and Dongwon Kim
- Subjects
0301 basic medicine ,Granulosa Cells ,Vasoactive intestinal peptide receptor ,Cell Biology ,Biology ,Cell biology ,Avian Proteins ,03 medical and health sciences ,030104 developmental biology ,Genetics ,Animals ,Receptors, Vasoactive Intestinal Peptide ,Female ,Signal transduction ,Receptor ,Chickens ,Cells, Cultured ,Developmental Biology ,Signal Transduction ,Vasoactive Intestinal Peptide - Published
- 2018
45. Discovery of artificial VIPR2-antagonist peptides possessing receptor- and ligand-selectivity
- Author
-
Yusuke Kamada, Masanori Miwa, Kotaro Sakamoto, Ryokichi Koyama, and Akiyoshi Tani
- Subjects
0301 basic medicine ,Vasoactive intestinal peptide ,Biophysics ,Peptide ,Biosensing Techniques ,CHO Cells ,Ligands ,Biochemistry ,03 medical and health sciences ,Mice ,Cricetulus ,Peptide Library ,Drug Discovery ,Animals ,Humans ,Receptor ,Molecular Biology ,G protein-coupled receptor ,chemistry.chemical_classification ,Vasoactive intestinal peptide receptor ,Cell Biology ,Surface Plasmon Resonance ,Cyclic peptide ,Recombinant Proteins ,Rats ,030104 developmental biology ,chemistry ,Pituitary Adenylate Cyclase-Activating Polypeptide ,Receptors, Vasoactive Intestinal Peptide, Type II ,VIPR1 ,VIPR2 ,Vasoactive Intestinal Peptide - Abstract
Vasoactive intestinal peptide receptor 2 (VIPR2, also known as VPAC2) is a class B G-protein coupled receptor (GPCR) and plays important roles in the physiology of central nervous system (CNS) by interaction with natural ligands; vasoactive intestinal peptide (VIP) and pituitary adenylate cyclase-activating polypeptide (PACAP). Because it has been reported that high-expression and/or overactivation of VIPR2 link to schizophrenic symptoms, VIPR2 antagonists could be good drug candidates for schizophrenia therapeutics. In this study, we discovered several artificial peptides that antagonize both human and rodent VIPR2 with selectivities against receptor subtypes VIPR1 (also known as VPAC1) and pituitary adenylate cyclase-activating polypeptide type-1 receptor (PAC1). Of them, the representative 16-mer cyclic peptide VIpep-3 (Ac-CPPYLPRRLCTLLLRS-OH) exhibited strong binding affinity with KD value of 41 nM to extracellular domain of human VIPR2 in SPR analysis and showed potent antagonist activity with IC50 values of 47 nM (human), 180 nM (mouse), and 44 nM (rat) against VIP–VIPR2 signal in cell-based Ca influx assay. This is not only the first report on artificial VIPR2-selective antagonist peptides but also good example of the effective approach to discover novel antagonist against class B GPCR. Our peptides will contribute to study and development of the novel CNS drugs targeting to VIPR2.
- Published
- 2018
46. Development by Genetic Immunization of Monovalent Antibodies Against Human Vasoactive Intestinal Peptide Receptor 1 (VPAC1), New Innovative, and Versatile Tools to Study VPAC1 Receptor Function
- Author
-
Xavier Peyrassol, Toon Laeremans, Vannessa Lahura, Maja Debulpaep, Hassan El Hassan, Jan Steyaert, Marc Parmentier, Ingrid Langer, Structural Biology Brussels, and Department of Bio-engineering Sciences
- Subjects
0301 basic medicine ,DNA immunization ,Phage display ,Vasoactive intestinal peptide ,Endocrinology, Diabetes and Metabolism ,Allosteric regulation ,Biopanning ,Monovalent antibodies ,lcsh:Diseases of the endocrine glands. Clinical endocrinology ,Epitope ,03 medical and health sciences ,Endocrinology ,0302 clinical medicine ,G protein-coupled receptor ,Receptor ,llama ,Original Research ,Vasoactive intestinal polypeptide receptor ,lcsh:RC648-665 ,Llama ,Chemistry ,Vasoactive intestinal peptide receptor ,Généralités ,Vasoactive intestinal peptide receptor 1 ,030104 developmental biology ,Biochemistry ,030217 neurology & neurosurgery - Abstract
Multi-membrane spanning proteins, such as G protein-coupled receptors (GPCRs) and ion channels, are extremely difficult to purify as native proteins. Consequently, the generation of antibodies that recognize the native conformation can be challenging. By combining genetic immunization, phage display, and biopanning, we identified a panel of monovalent antibodies (nanobodies) targeting the vasoactive intestinal peptide receptor 1 (VPAC1) receptor. The nine unique nanobodies that were classified into four different families based on their CDR3 amino acid sequence and length, were highly specific for the human receptor and bind VPAC1 with moderate affinity. They all recognize a similar epitope localized in the extracellular N-terminal domain of the receptor and distinct from the orthosteric binding site. In agreement with binding studies, which showed that the nanobodies did not interfere with VIP binding, all nanobodies were devoid of any functional properties. However, we observed that the binding of two nanobodies was slightly increased in the presence of VPAC1 agonists [vasoactive intestinal polypeptide (VIP) and pituitary adenylate cyclase-activating polypeptide-27 (PACAP-27)], but decreased in the presence of VPAC1 antagonist. As no evidence of allosteric activity was seen in VIP binding studies nor in functional assays, it is, therefore, possible that the two nanobodies may behave as very weak allosteric modulators of VPAC1, detectable only in some sensitive settings, but not in others. We demonstrated that the fluorescently labeled nanobodies detect VPAC1 on the surface of human leukocytes as efficiently as a reference mouse monoclonal antibody. We also developed a protocol allowing efficient detection of VPAC1 by immunohistochemistry in paraffin-embedded human gastrointestinal tissue sections. Thus, these nanobodies constitute new original tools to further investigate the role of VPAC1 in physiological and pathological conditions., SCOPUS: ar.j, info:eu-repo/semantics/published
- Published
- 2018
- Full Text
- View/download PDF
47. PACAP and its receptors in cranial arteries and mast cells
- Author
-
Inger Jansen-Olesen and Sara Hougaard Pedersen
- Subjects
0301 basic medicine ,medicine.medical_specialty ,Receptors, Vasoactive Intestinal Polypeptide, Type I ,Middle meningeal artery ,Migraine Disorders ,Vasoactive intestinal peptide ,Cerebral arteries ,Receptors, Pituitary Adenylate Cyclase-Activating Polypeptide ,lcsh:Medicine ,Review Article ,PACAP ,Cyclase ,Cell Degranulation ,03 medical and health sciences ,0302 clinical medicine ,Internal medicine ,medicine.artery ,Medicine ,Animals ,Humans ,Receptor ,Migraine ,business.industry ,Vasoactive intestinal peptide receptor ,lcsh:R ,Degranulation ,General Medicine ,Meningeal Arteries ,VIP ,Pituitary adenylate cyclase-activating peptide ,030104 developmental biology ,Anesthesiology and Pain Medicine ,Endocrinology ,Cerebral artery ,Pituitary Gland ,Mast cells ,Pituitary Adenylate Cyclase-Activating Polypeptide ,Receptors, Vasoactive Intestinal Peptide, Type II ,Neurology (clinical) ,business ,030217 neurology & neurosurgery ,Receptors, Pituitary Adenylate Cyclase-Activating Polypeptide, Type I ,Signal Transduction ,Vasoactive Intestinal Peptide - Abstract
Background In migraineurs pituitary adenylate cyclase activating peptide1–38 (PACAP1–38) is a potent migraine provoking substance and the accompanying long lasting flushing suggests degranulation of mast cells. Infusion of the closely related vasoactive intestinal peptide (VIP) either induces headache or flushing. This implicates the pituitary adenylate cyclase activating peptide type I receptor (PAC1) to be involved in the pathophysiology of PACAP1–38 provoked headaches. Here we review studies characterizing the effects of mainly PACAP but also of VIP on cerebral and meningeal arteries and mast cells. Discussion PACAP1–38, PACAP1–27 and VIP dilate cerebral and meningeal arteries from several species including man. In rat cerebral and meningeal arteries the dilation seems to be mediated preferably via vasoactive intestinal peptide receptor type 1 (VPAC1) receptors while, in human, middle meningeal artery dilation induced via vasoactive intestinal peptide receptor type 2 (VPAC2) receptors cannot be ruled out. PACAP1–38 is a strong degranulator of peritoneal and dural mast cells while PACAP1–27 and VIP only have weak effects. More detailed characterization studies suggest that mast cell degranulation is not mediated via the known receptors for PACAP1–38 but rather via a still unknown receptor coupled to phospholipase C. Conclusion It is suggested that PACAP1–38 might induce migraine via degranulation of dural mast cells via a yet unknown receptor.
- Published
- 2018
48. Drug delivery targets and systems for targeted treatment of rheumatoid arthritis
- Author
-
Xun Feng and Yang Chen
- Subjects
0301 basic medicine ,Side effect ,Angiogenesis ,Polymers ,Pharmaceutical Science ,02 engineering and technology ,Arthritis, Rheumatoid ,03 medical and health sciences ,Drug Delivery Systems ,Medicine ,Animals ,Humans ,Prodrugs ,Micelles ,business.industry ,Vasoactive intestinal peptide receptor ,021001 nanoscience & nanotechnology ,medicine.disease ,030104 developmental biology ,Targeted drug delivery ,Folate receptor ,Rheumatoid arthritis ,Antirheumatic Agents ,Drug delivery ,Cancer research ,0210 nano-technology ,business ,Selectin - Abstract
Rheumatoid arthritis (RA) is an immune-mediated inflammatory disease that selectively attacks human joints. The common non-targeted treatment approaches lead to obvious side effect and systemtic complication for RA patients. Therefore, targeted drug delivery for treatment of RA has gained much attetntion in the past few years. In this paper, we reviewed the potential targets (folate receptor, angiogenesis, matrix metalloproteases, selectins, vasoactive intestinal peptide receptor andFc-γ receptor) that could be utilised to facilitate the specific delivery of drugs to the inflammed synovium and also presented different drug delivery systems for targeting RA, including the liposomes, various types of nanoparticles, polymeric micelles and the macromolecular prodrugs. The strategies combining nanotechnologies and ligand mediated active targeting for RA would be emphatically illustrated, which was expected to be helpful for identifying technologies and drug delivery methods for targeted treatment of RA.
- Published
- 2018
49. Monocytes from Sjögren's syndrome patients display increased vasoactive intestinal peptide receptor 2 expression and impaired apoptotic cell phagocytosis
- Author
-
Laura Fraccaroli, Esteban Grasso, Vanesa Hauk, A. Eimon, Rosanna Ramhorst, C. Pérez Leirós, and Osvaldo Hübscher
- Subjects
Adult ,Lipopolysaccharides ,CIENCIAS MÉDICAS Y DE LA SALUD ,Lipopolysaccharide ,Immunology ,Vasoactive intestinal peptide ,Inmunología ,Apoptosis ,VPAC RECEPTORS ,Biology ,Monocytes ,Apoptotic cell clearance ,Young Adult ,chemistry.chemical_compound ,medicine ,Humans ,APOPTOTIC CELL CLEARANCE ,Immunology and Allergy ,Macrophage ,Receptor ,Aged ,Autoimmune disease ,Vasoactive intestinal peptide receptor ,Monocyte ,Original Articles ,Middle Aged ,medicine.disease ,Medicina Básica ,Sjogren's Syndrome ,medicine.anatomical_structure ,Gene Expression Regulation ,chemistry ,Cytophagocytosis ,MONOCYTES ,Case-Control Studies ,Receptors, Vasoactive Intestinal Peptide ,Receptors, Vasoactive Intestinal Peptide, Type II ,SJÖGREN'S SYNDROME PATIENTS ,Vasoactive Intestinal Peptide - Abstract
Sjögren's syndrome (SS) is a chronic autoimmune disease characterized by salivary and lacrimal gland dysfunction. Clinical observations and results from animal models of SS support the role of aberrant epithelial cell apoptosis and immune homeostasis loss in the glands as triggering factors for the autoimmune response. Vasoactive intestinal peptide (VIP) promotes potent anti-inflammatory effects in several inflammatory and autoimmune disease models, including the non-obese diabetic (NOD) mouse model of SS. With the knowledge that VIP modulates monocyte function through vasoactive intestinal peptide receptors (VPAC) and that immune homeostasis maintenance depends strongly upon a rapid and immunosuppressant apoptotic cell clearance by monocytes/macrophages, in this study we explored VPAC expression on monocytes from primary SS (pSS) patients and the ability of VIP to modulate apoptotic cell phagocytic function and cytokine profile. Monocytes isolated from individual pSS patients showed an increased expression of VPAC2 subtype of VIP receptors, absent in monocytes from control subjects, with no changes in VPAC1 expression. VPAC2 receptor expression could be induced further with lipopolysaccharide (LPS) in pSS monocytes and VIP inhibited the effect. Moreover, monocytes from pSS patients showed an impaired phagocytosis of apoptotic epithelial cells, as evidenced by reduced engulfment ability and the failure to promote an immunosuppressant cytokine profile. However, VIP neither modulated monocyte/macrophage phagocytic function nor did it reverse their inflammatory profile. We conclude that monocytes from pSS patients express high levels of VPAC2 and display a deficient clearance of apoptotic cells that is not modulated by VIP. Fil: Hauk, Vanesa Cintia. Universidad de Buenos Aires. Facultad de Ciencias Exactas y Naturales. Departamento de Química Biológica. Laboratorio de Inmunofarmacología; Argentina. Consejo Nacional de Investigaciones Científicas y Técnicas. Oficina de Coordinación Administrativa Ciudad Universitaria. Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales. Universidad de Buenos Aires. Facultad de Ciencias Exactas y Naturales. Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales; Argentina Fil: Fraccaroli, Laura Virginia. Universidad de Buenos Aires. Facultad de Ciencias Exactas y Naturales. Departamento de Química Biológica. Laboratorio de Inmunofarmacología; Argentina. Consejo Nacional de Investigaciones Científicas y Técnicas. Oficina de Coordinación Administrativa Ciudad Universitaria. Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales. Universidad de Buenos Aires. Facultad de Ciencias Exactas y Naturales. Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales; Argentina Fil: Grasso, Esteban Nicolas. Universidad de Buenos Aires. Facultad de Ciencias Exactas y Naturales. Departamento de Química Biológica. Laboratorio de Inmunofarmacología; Argentina. Consejo Nacional de Investigaciones Científicas y Técnicas. Oficina de Coordinación Administrativa Ciudad Universitaria. Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales. Universidad de Buenos Aires. Facultad de Ciencias Exactas y Naturales. Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales; Argentina Fil: Eimon, Alicia. Centro de Educación Médica e Investigaciones Clínicas “Norberto Quirno”; Argentina Fil: Ramhorst, Rosanna Elizabeth. Universidad de Buenos Aires. Facultad de Ciencias Exactas y Naturales. Departamento de Química Biológica. Laboratorio de Inmunofarmacología; Argentina. Consejo Nacional de Investigaciones Científicas y Técnicas. Oficina de Coordinación Administrativa Ciudad Universitaria. Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales. Universidad de Buenos Aires. Facultad de Ciencias Exactas y Naturales. Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales; Argentina Fil: Hubscher, Osvaldo. Centro de Educación Médica e Investigaciones Clínicas “Norberto Quirno”; Argentina Fil: Perez Leiros, Claudia. Universidad de Buenos Aires. Facultad de Ciencias Exactas y Naturales. Departamento de Química Biológica. Laboratorio de Inmunofarmacología; Argentina. Consejo Nacional de Investigaciones Científicas y Técnicas. Oficina de Coordinación Administrativa Ciudad Universitaria. Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales. Universidad de Buenos Aires. Facultad de Ciencias Exactas y Naturales. Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales; Argentina
- Published
- 2014
- Full Text
- View/download PDF
50. Stapled Vasoactive Intestinal Peptide (VIP) Derivatives Improve VPAC2 Agonism and Glucose-Dependent Insulin Secretion
- Author
-
Claire Priest, Laurent Knerr, Adrian Liam Gill, Fabrizio Giordanetto, Annika Janefeldt, Jefferson D. Revell, Amalia Paunovic, and Marie Hostettler
- Subjects
Agonist ,chemistry.chemical_classification ,medicine.medical_specialty ,Protease ,medicine.drug_class ,business.industry ,medicine.medical_treatment ,Vasoactive intestinal peptide receptor ,Organic Chemistry ,Vasoactive intestinal peptide ,Endogeny ,Peptide ,Peptide hormone ,Biochemistry ,Endocrinology ,chemistry ,In vivo ,Internal medicine ,Drug Discovery ,medicine ,business - Abstract
Agonists of vasoactive intestinal peptide receptor 2 (VPAC2) stimulate glucose-dependent insulin secretion, making them attractive candidates for the treatment of hyperglycaemia and type-II diabetes. Vasoactive intestinal peptide (VIP) is an endogenous peptide hormone that potently agonizes VPAC2. However, VIP has a short serum half-life and poor pharmacokinetics in vivo and is susceptible to proteolytic degradation, making its development as a therapeutic agent challenging. Here, we investigated two peptide cyclization strategies, lactamisation and olefin-metathesis stapling, and their effects on VPAC2 agonism, peptide secondary structure, protease stability, and cell membrane permeability. VIP analogues showing significantly enhanced VPAC2 agonist potency, glucose-dependent insulin secretion activity, and increased helical content were discovered; however, neither cyclization strategy appeared to effect proteolytic stability or cell permeability of the resulting peptides.
- Published
- 2013
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.