Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 274
Filtrar
1.
Sci Rep ; 13(1): 927, 2023 01 17.
Artigo em Inglês | MEDLINE | ID: mdl-36650220

RESUMO

Macrophages are a major population of immune cells in solid cancers, especially colorectal cancers. Tumor-associated macrophages (TAMs) are commonly divided into M1-like (tumor suppression) and M2-like (tumor promotion) phenotypes. Vasoactive intestinal peptide (VIP) is an immunoregulatory neuropeptide with a potent anti-inflammatory function. Inhibition of VIP signaling has been shown to increase CD8+ T cell proliferation and function in viral infection and lymphoma. However, the role of VIP in macrophage polarization and function in solid tumors remains unknown. Here, we demonstrated that conditioned medium from CT26 (CT26-CM) cells enhanced M2-related marker and VIP receptor (VPAC) gene expression in RAW264.7 macrophages. VIP hybrid, a VIP antagonist, enhanced M1-related genes but reduced Mrc1 gene expression and increased phagocytic ability in CT26-CM-treated RAW264.7 cells. In immunodeficient SCID mice, VIP antagonist alone or in combination with anti-PD-1 antibody attenuated CT26 tumor growth compared with the control. Analysis of tumor-infiltrating leukocytes found that VIP antagonist increased M1/M2 ratios and macrophage phagocytosis of CT26-GFP cells. Furthermore, Vipr2 gene silencing or VPAC2 activation affected the polarization of CT26-CM-treated RAW264.7 cells. In conclusion, the inhibition of VIP signaling enhanced M1 macrophage polarization and macrophage phagocytic function, resulting in tumor regression in a CT26 colon cancer model.


Assuntos
Neoplasias do Colo , Macrófagos , Peptídeo Intestinal Vasoativo , Animais , Camundongos , Neoplasias do Colo/patologia , Macrófagos/metabolismo , Camundongos SCID , Receptores de Peptídeo Intestinal Vasoativo/metabolismo , Receptores Tipo II de Peptídeo Intestinal Vasoativo/metabolismo , Transdução de Sinais , Peptídeo Intestinal Vasoativo/antagonistas & inibidores , Peptídeo Intestinal Vasoativo/metabolismo , Células RAW 264.7
2.
Life Sci ; 265: 118792, 2021 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-33220286

RESUMO

AIMS: In the cyclic rat in estrus, the vasoactive intestinal peptide (VIP) has an impact on ovarian function, which depends on the endocrine status of the animal. In this work, we aimed to clarify the participation of VIP in the pathophysiological condition of polycystic ovary syndrome (PCOS) using a model of PCOS induced by estradiol valerate (EV-PCOS) in rats. MAIN METHODS: In the cyclic rat in estrus and in the EV-PCOS model, we analyzed the acute effects of blocking VIP receptors with the use of an antagonist (Ant-VIP) injected into the left or right ovarian bursa on the steroidogenic response and ovarian catecholamine levels. KEY FINDINGS: In the cyclic animal in estrus, the treatment with Ant-VIP in the left ovarian bursa resulted in a reduction in testosterone serum levels and in ovarian levels of dopamine and 3,4-dihydroxyphenylacetic acid (DOPAC), without changes in 4-hydroxy-3-methoxyphenyl (MHPG) and norepinephrine (NE). When the treatment was applied on the right side, only MHPG levels increased. In the EV-PCOS model, the treatment with Ant-VIP in the left ovarian bursa increased testosterone, estradiol, MHPG, and NE levels. When the treatment was performed on the right side, progesterone levels decreased and estradiol increased, without changes in ovarian catecholamines. SIGNIFICANCE: The binding of VIP to its receptors differentially regulates steroidogenesis in the cyclic animal in estrus and in the EV-PCOS model. The blocking of VIP signaling produces changes in ovarian catecholamines.


Assuntos
Modelos Animais de Doenças , Ovário/metabolismo , Síndrome do Ovário Policístico/metabolismo , Receptores de Peptídeo Intestinal Vasoativo/antagonistas & inibidores , Receptores de Peptídeo Intestinal Vasoativo/metabolismo , Animais , Catecolaminas/metabolismo , Estradiol/metabolismo , Estradiol/toxicidade , Feminino , Ovário/efeitos dos fármacos , Síndrome do Ovário Policístico/induzido quimicamente , Síndrome do Ovário Policístico/tratamento farmacológico , Ratos , Testosterona/metabolismo , Peptídeo Intestinal Vasoativo/antagonistas & inibidores , Peptídeo Intestinal Vasoativo/metabolismo
3.
Nat Commun ; 11(1): 1453, 2020 03 19.
Artigo em Inglês | MEDLINE | ID: mdl-32193397

RESUMO

The suprachiasmatic nucleus (SCN) circadian clock is critical for optimising daily cycles in mammalian physiology and behaviour. The roles of the various SCN cell types in communicating timing information to downstream physiological systems remain incompletely understood, however. In particular, while vasoactive intestinal polypeptide (VIP) signalling is essential for SCN function and whole animal circadian rhythmicity, the specific contributions of VIP cell output to physiological control remains uncertain. Here we reveal a key role for SCN VIP cells in central clock output. Using multielectrode recording and optogenetic manipulations, we show that VIP neurons provide coordinated daily waves of GABAergic input to target cells across the paraventricular hypothalamus and ventral thalamus, supressing their activity during the mid to late day. Using chemogenetic manipulation, we further demonstrate specific roles for this circuitry in the daily control of heart rate and corticosterone secretion, collectively establishing SCN VIP cells as influential regulators of physiological timing.


Assuntos
Relógios Circadianos/fisiologia , Ritmo Circadiano/fisiologia , Neurônios GABAérgicos/metabolismo , Núcleo Supraquiasmático/fisiologia , Peptídeo Intestinal Vasoativo/metabolismo , Potenciais de Ação/efeitos dos fármacos , Potenciais de Ação/fisiologia , Animais , Bicuculina/farmacologia , Channelrhodopsins/química , Channelrhodopsins/genética , Channelrhodopsins/metabolismo , Relógios Circadianos/efeitos dos fármacos , Corticosterona/sangue , Corticosterona/metabolismo , Eletrodos Implantados , Feminino , Antagonistas de Receptores de GABA-A/farmacologia , Neurônios GABAérgicos/efeitos dos fármacos , Frequência Cardíaca/fisiologia , Masculino , Camundongos , Camundongos Transgênicos , Modelos Animais , Núcleo Supraquiasmático/citologia , Peptídeo Intestinal Vasoativo/antagonistas & inibidores , Peptídeo Intestinal Vasoativo/genética
4.
Braz J Med Biol Res ; 51(11): e7541, 2018 Oct 04.
Artigo em Inglês | MEDLINE | ID: mdl-30304131

RESUMO

We previously found that acute exercise inhibited the gastric emptying of liquid in awake rats by causing an acid-base imbalance. In the present study, we investigated the involvement of the nitric oxide-cyclic guanosine monophosphate (NO-cGMP) pathway, vasoactive intestinal peptide (VIP), and corticotropin-releasing factor (CRF) peptide in this phenomenon. Male rats were divided into exercise or sedentary group and were subjected to a 15-min swim session against a load (2.5 or 5% b.w.). The rate of gastric emptying was evaluated after 5, 10, or 20 min postprandially. Separate groups of rats were treated with vehicle (0.9% NaCl, 0.1 mL/100 g, ip) or one of the following agents: atropine (1.0 mg/kg, ip), the NO non-selective inhibitor Nω-nitro-L-arginine methyl ester hydrochloride (L-NAME; 10.0 mg/kg, ip), or the selective cGMP inhibitor 1H-(1,2,4)oxadiazole[4,3-a]quinoxalin-1-one (ODQ; 5.0 mg/kg, ip), the i-NOS non-specific inhibitor (aminoguanidine; 10.0 mg/kg, ip), the corticotropin-releasing factor receptor antagonist (astressin; 100 µg/kg, ip), or the vasoactive intestinal peptide (VIP) receptor antagonist Lys1, Pro2,5, Arg3,4, Tyr6 (100 µg/kg, ip). Compared to sedentary rats, both the 2.5 and 5% exercise groups exhibited higher (P<0.05) values of blood lactate and fractional gastric dye recovery. Corticosterone and NO levels increased (P<0.05) in the 5% exercised rats. Pretreatment with astressin, VIP antagonist, atropine, L-NAME, and ODQ prevented the increase in gastric retention caused by exercise in rats. Acute exercise increased gastric retention, a phenomenon that appears to be mediated by the NO-cGMP pathway, CRF, and VIP receptors.


Assuntos
Hormônio Liberador da Corticotropina/metabolismo , Esvaziamento Gástrico/fisiologia , Guanosina Monofosfato/metabolismo , Óxido Nítrico/metabolismo , Condicionamento Físico Animal/fisiologia , Peptídeo Intestinal Vasoativo/metabolismo , Animais , Atropina/farmacologia , Corticosterona/sangue , Hormônio Liberador da Corticotropina/antagonistas & inibidores , Hormônio Liberador da Corticotropina/farmacologia , Inibidores Enzimáticos/farmacologia , Esvaziamento Gástrico/efeitos dos fármacos , Guanosina Monofosfato/antagonistas & inibidores , Ácido Láctico/sangue , Masculino , NG-Nitroarginina Metil Éster/farmacologia , Óxido Nítrico/antagonistas & inibidores , Fragmentos de Peptídeos/farmacologia , Período Pós-Prandial/efeitos dos fármacos , Período Pós-Prandial/fisiologia , Distribuição Aleatória , Ratos Wistar , Valores de Referência , Reprodutibilidade dos Testes , Comportamento Sedentário , Fatores de Tempo , Peptídeo Intestinal Vasoativo/antagonistas & inibidores
5.
Am J Physiol Gastrointest Liver Physiol ; 314(5): G610-G622, 2018 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-29420068

RESUMO

We investigated whether vasoactive intestinal peptide (VIP) and/or prostaglandins contribute to peripheral corticotropin-releasing factor (CRF)-induced CRF1 receptor-mediated stimulation of colonic motor function and diarrhea in rats. The VIP antagonist, [4Cl-D-Phe6, Leu17]VIP injected intraperitoneally completely prevented CRF (10 µg/kg ip)-induced fecal output and diarrhea occurring within the first hour after injection, whereas pretreatment with the prostaglandins synthesis inhibitor, indomethacin, had no effect. In submucosal plexus neurons, CRF induced significant c-Fos expression most prominently in the terminal ileum compared with duodenum and jejunum, whereas no c-Fos was observed in the proximal colon. c-Fos expression in ileal submucosa was colocalized in 93.4% of VIP-positive neurons and 31.1% of non-VIP-labeled neurons. CRF1 receptor immunoreactivity was found on the VIP neurons. In myenteric neurons, CRF induced only a few c-Fos-positive neurons in the ileum and a robust expression in the proximal colon (17.5 ± 2.4 vs. 0.4 ± 0.3 cells/ganglion in vehicle). The VIP antagonist prevented intraperitoneal CRF-induced c-Fos induction in the ileal submucosal plexus and proximal colon myenteric plexus. At 60 min after injection, CRF decreased VIP levels in the terminal ileum compared with saline (0.8 ± 0.3 vs. 2.5 ± 0.7 ng/g), whereas VIP mRNA level detected by qPCR was not changed. These data indicate that intraperitoneal CRF activates intestinal submucosal VIP neurons most prominently in the ileum and myenteric neurons in the colon. It also implicates VIP signaling as part of underlying mechanisms driving the acute colonic secretomotor response to a peripheral injection of CRF, whereas prostaglandins do not play a role. NEW & NOTEWORTHY Corticotropin-releasing factor (CRF) in the gut plays a physiological role in the stimulation of lower gut secretomotor function induced by stress. We showed that vasoactive intestinal peptide (VIP)-immunoreactive neurons in the ileal submucosal plexus expressed CRF1 receptor and were prominently activated by CRF, unlike colonic submucosal neurons. VIP antagonist abrogated CRF-induced ileal submucosal and colonic myenteric activation along with functional responses (defecation and diarrhea). These data point to VIP signaling in ileum and colon as downstream effectors of CRF.


Assuntos
Hormônio Liberador da Corticotropina/metabolismo , Diarreia , Motilidade Gastrointestinal , Plexo Mientérico , Peptídeo Intestinal Vasoativo , Animais , Colo/metabolismo , Colo/fisiopatologia , Defecação/efeitos dos fármacos , Defecação/fisiologia , Diarreia/metabolismo , Diarreia/fisiopatologia , Motilidade Gastrointestinal/efeitos dos fármacos , Motilidade Gastrointestinal/fisiologia , Genes fos/fisiologia , Íleo/metabolismo , Íleo/fisiopatologia , Mucosa Intestinal/metabolismo , Masculino , Plexo Mientérico/efeitos dos fármacos , Plexo Mientérico/metabolismo , Fármacos Neuroprotetores/metabolismo , Ratos , Peptídeo Intestinal Vasoativo/antagonistas & inibidores , Peptídeo Intestinal Vasoativo/metabolismo
6.
Blood Adv ; 2(3): 210-223, 2018 02 13.
Artigo em Inglês | MEDLINE | ID: mdl-29386194

RESUMO

Adoptive therapy with ex vivo-expanded genetically modified antigen-specific T cells can induce remissions in patients with relapsed/refractory cancer. The clinical success of this therapy depends upon efficient transduction and expansion of T cells ex vivo and their homing, persistence and cytotoxicity following reinfusion. Lower rates of ex vivo expansion and clinical response using anti-CD19 chimeric antigen receptor (CAR) T cells have been seen in heavily pretreated lymphoma patients compared with B-cell acute lymphoblastic leukemia patients and motivate the development of novel strategies to enhance ex vivo T cell expansion and their persistence in vivo. We demonstrate that inhibition of phosphatidylinositol 3-kinase δ (PI3Kδ) and antagonism of vasoactive intestinal peptide (VIP) signaling partially inhibits the terminal differentiation of T cells during anti-CD3/CD28 bead-mediated expansion (mean, 54.4% CD27+CD28+ T cells vs 27.4% in control cultures; P < .05). This strategy results in a mean of 83.7% more T cells cultured from lymphoma patients in the presence of PI3Kδ and VIP antagonists, increased survival of human T cells from a lymphoma patient in a murine xenograft model, enhanced cytotoxic activity of antigen-specific human CAR T cells and murine T cells against lymphoma, and increased transduction and expansion of anti-CD5 human CAR T cells. PI3Kδ and VIP antagonist-expanded T cells from lymphoma patients show reduced terminal differentiation, enhanced polyfunctional cytokine expression, and preservation of costimulatory molecule expression. Taken together, synergistic blockade of these pathways is an attractive strategy to enhance the expansion and functional capacity of ex vivo-expanded cancer-specific T cells.


Assuntos
Técnicas de Cultura de Células/métodos , Classe I de Fosfatidilinositol 3-Quinases/antagonistas & inibidores , Imunoterapia Adotiva/métodos , Linfócitos T/citologia , Peptídeo Intestinal Vasoativo/antagonistas & inibidores , Adulto , Idoso , Animais , Senescência Celular/efeitos dos fármacos , Feminino , Xenoenxertos , Humanos , Linfoma/terapia , Linfoma Difuso de Grandes Células B/terapia , Camundongos , Pessoa de Meia-Idade , Neurotensina/farmacologia , Purinas/farmacologia , Quinazolinonas/farmacologia , Proteínas Recombinantes de Fusão/farmacologia , Linfócitos T/efeitos dos fármacos , Peptídeo Intestinal Vasoativo/farmacologia
7.
J Neurosci ; 38(6): 1326-1334, 2018 02 07.
Artigo em Inglês | MEDLINE | ID: mdl-29054877

RESUMO

In mammals, the suprachiasmatic nucleus (SCN) of the hypothalamus coordinates daily rhythms including sleep-wake, hormone release, and gene expression. The cells of the SCN must synchronize to each other to drive these circadian rhythms in the rest of the body. The ontogeny of circadian cycling and intercellular coupling in the SCN remains poorly understood. Recent in vitro studies have recorded circadian rhythms from the whole embryonic SCN. Here, we tracked the onset and precision of rhythms in PERIOD2 (PER2), a clock protein, within the SCN isolated from embryonic and postnatal mice of undetermined sex. We found that a few SCN cells developed circadian periodicity in PER2 by 14.5 d after mating (E14.5) with no evidence for daily cycling on E13.5. On E15.5, the fraction of competent oscillators increased dramatically corresponding with stabilization of their circadian periods. The cells of the SCN harvested at E15.5 expressed sustained, synchronous daily rhythms. By postnatal day 2 (P2), SCN oscillators displayed the daily, dorsal-ventral phase wave in clock gene expression typical of the adult SCN. Strikingly, vasoactive intestinal polypeptide (VIP), a neuropeptide critical for synchrony in the adult SCN, and its receptor, VPAC2R, reached detectable levels after birth and after the onset of circadian synchrony. Antagonists of GABA or VIP signaling or action potentials did not disrupt circadian synchrony in the E15.5 SCN. We conclude that endogenous daily rhythms in the fetal SCN begin with few noisy oscillators on E14.5, followed by widespread oscillations that rapidly synchronize on E15.5 by an unknown mechanism.SIGNIFICANCE STATEMENT We recorded the onset of PER2 circadian oscillations during embryonic development in the mouse SCN. When isolated at E13.5, the anlagen of the SCN expresses high, arrhythmic PER2. In contrast, a few cells show noisy circadian rhythms in the isolated E14.5 SCN and most show reliable, self-sustained, synchronized rhythms in the E15.5 SCN. Strikingly, this synchrony at E15.5 appears before expression of VIP or its receptor and persists in the presence of blockers of VIP, GABA or neuronal firing. Finally, the dorsal-ventral phase wave of PER2 typical of the adult SCN appears ∼P2, indicating that multiple signals may mediate circadian synchrony during the ontogeny of the SCN.


Assuntos
Ritmo Circadiano/fisiologia , Núcleo Supraquiasmático/fisiologia , Envelhecimento/genética , Envelhecimento/fisiologia , Animais , Feminino , Antagonistas GABAérgicos/farmacologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Neurônios/fisiologia , Proteínas Circadianas Period/genética , Proteínas Circadianas Period/fisiologia , Gravidez , Receptores Tipo II de Peptídeo Intestinal Vasoativo/biossíntese , Receptores Tipo II de Peptídeo Intestinal Vasoativo/genética , Núcleo Supraquiasmático/citologia , Núcleo Supraquiasmático/crescimento & desenvolvimento , Peptídeo Intestinal Vasoativo/antagonistas & inibidores , Peptídeo Intestinal Vasoativo/metabolismo , Peptídeo Intestinal Vasoativo/fisiologia
8.
Artigo em Chinês | MEDLINE | ID: mdl-27197456

RESUMO

OBJECTIVE: To explore the expression and significance of vasoactive intestinal peptide and Pituitary adenylate cyclase activiting polypeptide (VIP/PACAP) of nasal mucosa in rats with allergic rhinitis (AR), and the function of botulinum toxin-A(BTX-A) to inhibit the expression of VIP/PACAP in AR. METHOD: Thirty Sprague-Dawley rats were randomly divided into 3 groups, which were the AR group, the intervention group, and the control group. In the AR group, ovalbumin was used to sensitize healthy rats. In the intervention group, BTX-A was dripped into the nasal cavity of AR rats 7 times. In the control group, only physiological saline was used to drip into the nasal cavity of AR rats. Changes of the rats' behavior were observed. ELISA were used to detected the concentration variation of serum IFN-γ and IL-4. Histopathology and immunohistochemistry were employed to observe morphology in the rats' nasal mucosal and the expression of VIP/PACAP. Statistical analysis was also made. RESULT: (1)The typical symptoms marks of nasal scratching, sneezing, nasal blockage and rhinorrhea of AR group (7.5 ± 0.50) were higher than intervention group (1 ± 0.27) and control group (0.8 ± 0.31). (2) Comparing to intervention group and control group, the serm IFN-γ of the AR group obvious reduced (P < 0.05), the serm IL-4 of the AR group obvious rose (P < 0.01), and the serm Th1/Th2 (IFN-γ/IL-4) of the AR group obvious reduced (P < 0.01). (3) Comparing to intervention group and control group, the cilium loss, inflammatory cells infiltration, and inflammatory cells exudation of nasal mucosa in AR group were more obviously (P < 0.01), and the intervention group of the 3 indexes was obviously than control group. (4) The expression of VIP in the rats' nasal mucosa of the AR group (13.27 ± 2.74) were more intense than intervention group (5.21 ± 2.18) and control group (3.56 ± 5.30) (P < 0.01), and the expression of PACAP in the rats' nasal mucosa of the AR group (20.97 ± 2.14) were more intense than intervention group (6.33 ± 3.04) and control group (4.63 ± 1.25) (P < 0.01). (5) In all the 3 groups, there was positive correlation between expression of negative in VIP/PACAP and Thl/Th2 cell infiltration(r were respectively -0.340 and -0.223, P < 0.05). CONCLUSION: The VIP/PACAP in the rats' nasal mucosa may play an important role in pathogenesis of AR, and BTX-A could improve the symptoms of AR through inhibition of the expression of VIP/ PACAP.


Assuntos
Toxinas Botulínicas Tipo A/farmacologia , Mucosa Nasal/efeitos dos fármacos , Polipeptídeo Hipofisário Ativador de Adenilato Ciclase/antagonistas & inibidores , Rinite Alérgica/tratamento farmacológico , Peptídeo Intestinal Vasoativo/antagonistas & inibidores , Animais , Modelos Animais de Doenças , Interferon gama/sangue , Interleucina-4/sangue , Mucosa Nasal/metabolismo , Ovalbumina , Seios Paranasais , Polipeptídeo Hipofisário Ativador de Adenilato Ciclase/metabolismo , Ratos , Ratos Sprague-Dawley , Peptídeo Intestinal Vasoativo/metabolismo
9.
J Comp Neurol ; 524(12): 2516-28, 2016 08 15.
Artigo em Inglês | MEDLINE | ID: mdl-26801210

RESUMO

Neurogenesis and neuronal recruitment occur in adult brains of many vertebrates, and the hypothesis is that these phenomena contribute to the brain plasticity that enables organisms to adjust to environmental changes. In mammals, vasoactive intestinal polypeptide (VIP) is known to have many neuroprotective properties, but in the avian brain, although widely distributed, its role in neuronal recruitment is not yet understood. In the present study we actively immunized adult zebra finches against VIP conjugated to KLH and compared neuronal recruitment in their brains, with brains of control birds, which were immunized against KLH. We looked at two forebrain regions: the nidopallium caudale (NC), which plays a role in vocal communication, and the hippocampus (HC), which is involved in the processing of spatial information. Our data demonstrate that active immunization against VIP reduces neuronal recruitment, inhibits reproduction, and induces molting, with no change in plasma prolactin levels. Thus, our observations suggest that VIP has a direct positive role in neuronal recruitment and reproduction in birds. J. Comp. Neurol. 524:2516-2528, 2016. © 2016 Wiley Periodicals, Inc.


Assuntos
Neurogênese/fisiologia , Neurônios/efeitos dos fármacos , Reprodução/efeitos dos fármacos , Vacinação/métodos , Peptídeo Intestinal Vasoativo/antagonistas & inibidores , Sequência de Aminoácidos/fisiologia , Animais , Galinhas , Feminino , Tentilhões , Neurogênese/efeitos dos fármacos , Neurônios/fisiologia , Reprodução/fisiologia , Peptídeo Intestinal Vasoativo/fisiologia
10.
Heart Rhythm ; 12(6): 1352-61, 2015 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-25748673

RESUMO

BACKGROUND: Vagal hyperactivity promotes atrial fibrillation (AF), which has been almost exclusively attributed to acetylcholine. Vasoactive intestinal polypeptide (VIP) and acetylcholine are neurotransmitters co-released during vagal stimulation. Exogenous VIP has been shown to promote AF by shortening action potential duration (APD), increasing APD spatial heterogeneity, and causing intra-atrial conduction block. OBJECTIVE: The purpose of this study was to investigate the effects of neuronally released VIP on atrial electrophysiologic properties during vagal stimulation. METHODS: We used a specific VIP antagonist (H9935) to uncover the effects of endogenous VIP released during vagal stimulation in canine hearts. RESULTS: H9935 significantly attenuated (1) the vagally induced shortening of atrial effective refractory period and widening of atrial vulnerability window during stimulation of cervical vagosympathetic trunks (VCNS) and (2) vagal effects on APD during stimulation through fat-pad ganglion plexus (VGPS). Atropine completely abolished these vagal effects during VCNS and VGPS. In contrast, VGPS-induced slowing of local conduction velocity was completely abolished by either VIP antagonist or atropine. In pacing-induced AF during VGPS, maximal dominant frequencies and their spatial gradients were reduced significantly by H9935 and, more pronouncedly, by atropine. Furthermore, VIP release in the atria during vagal stimulation was inhibited by atropine, which may account for the concealment of VIP effects with muscarinic blockade. CONCLUSION: Neuronally released VIP contributes to vagal effects on atrial electrophysiologic properties and affects the pathophysiology of vagally induced AF. Neuronal release of VIP in the atria is inhibited by muscarinic blockade, a novel mechanism by which VIP effects are concealed by atropine during vagal stimulation.


Assuntos
Fibrilação Atrial/etiologia , Função Atrial/fisiologia , Estimulação do Nervo Vago , Peptídeo Intestinal Vasoativo/metabolismo , Peptídeo Intestinal Vasoativo/fisiologia , Potenciais de Ação , Animais , Atropina/farmacologia , Cães , Muscarina/farmacologia , Período Refratário Eletrofisiológico/efeitos dos fármacos , Peptídeo Intestinal Vasoativo/antagonistas & inibidores
11.
ScientificWorldJournal ; 2013: 429545, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-24228003

RESUMO

OBJECTIVE: To investigate VIP effect on the cytotoxicity of NK cell to gastric cancer cells in vitro and the relation between the effect with the NKG2D signal molecules in NK cells. MATERIAL AND METHODS: NK cells were purified from peripheral blood mononuclear cells (PBMC). Before and after NK cells were incubated with VIP or its antagonist (D-p-Cl-Phe6,Leu17)-VIP, we detected the cytotoxicity of NK cells to MKN45 gastric cancer cells by MTT and detected the expressions of NKG2D, DAP10, and NF- κ B proteins and mRNAs in NK cells by immunocytochemistry and RT-PCR in those conditions. Then we analyzed the effect of VIP and its antagonist on the cytotocicity of NK cell to gastric cancer cells and on expressions of NKG2D, DAP10, and NF- κ B signal molecules in NK cells. RESULTS: VIP could inhibit the cytotoxicity of NK cells to MKN45 cells and could inhibit the expressions of NKG2D, DAP10, and NF- κ B in NK cells. However, (D-p-Cl-Phe6, Leu17)-VIP could reverse those effects. CONCLUSIONS: The VIP inhibited the cytotoxicity of NK cell to MKN45 cells which might get through inhibiting the expressions of NKG2D signal molecules in NK cells. This may be one mechanism of gastric cancer cells escaping organism immune clearance.


Assuntos
Células Matadoras Naturais/imunologia , Subfamília K de Receptores Semelhantes a Lectina de Células NK/metabolismo , Transdução de Sinais/efeitos dos fármacos , Evasão Tumoral/efeitos dos fármacos , Peptídeo Intestinal Vasoativo/farmacologia , Análise de Variância , Linhagem Celular Tumoral , Testes Imunológicos de Citotoxicidade , Primers do DNA , Humanos , Imuno-Histoquímica , NF-kappa B/metabolismo , Receptores Imunológicos/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Estatísticas não Paramétricas , Sais de Tetrazólio , Tiazóis , Peptídeo Intestinal Vasoativo/antagonistas & inibidores
12.
Horm Behav ; 64(3): 511-8, 2013 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-23899763

RESUMO

In both mammals and birds, vasoactive intestinal polypeptide (VIP) neurons and fibers are present in virtually every brain area that is important for social behavior. VIP influences aggression in birds, social recognition in rodents, and prolactin secretion in both taxa, but other possible functions in social modulation remain little explored. VIP effects are mediated by VPAC receptors, which bind both VIP and pituitary adenylate cyclase activating peptide. Within the lateral septum and medial bed nucleus of the stria terminalis, VPAC receptors are found at higher densities in gregarious finch species relative to territorial species, suggesting that VPAC receptor activation promotes social contact and/or preference for larger groups. Here we here test this hypothesis in zebra finches (Taeniopygia guttata), and also examine the relevance of VPAC receptors to anxiety-like processes. Intraventricular infusions of the VPAC receptor antagonist, neurotensin6-11 mouseVIP7-28, strongly reduce social contact when animals are tested in a novel environment, and exert sex-specific effects on grouping behavior. Specifically, VPAC receptor antagonism reduces gregariousness in females but increases gregariousness in males. Interestingly, VPAC antagonism in the medial pallium (putative prefrontal cortex homologue) significantly reduces gregariousness in both sexes, suggesting site-specific effects of VIP signaling. However, VPAC antagonism does not modulate novel-familiar social preferences in a familiar environment or general anxiety-like behaviors. The current results suggest that endogenous activation of VPAC receptors promotes social contact under novel environmental conditions, a function that may be accentuated in gregarious species. Moreover, endogenous VIP modulates gregariousness in both males and females.


Assuntos
Comportamento Animal/fisiologia , Comportamento Exploratório/fisiologia , Tentilhões/fisiologia , Neurotensina/farmacologia , Córtex Pré-Frontal/efeitos dos fármacos , Receptores de Peptídeo Intestinal Vasoativo/fisiologia , Comportamento Social , Peptídeo Intestinal Vasoativo/farmacologia , Animais , Ansiedade/fisiopatologia , Comportamento Animal/efeitos dos fármacos , Meio Ambiente , Comportamento Exploratório/efeitos dos fármacos , Feminino , Masculino , Córtex Pré-Frontal/fisiologia , Receptores de Peptídeo Intestinal Vasoativo/antagonistas & inibidores , Proteínas Recombinantes de Fusão/farmacologia , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/fisiologia , Peptídeo Intestinal Vasoativo/análogos & derivados , Peptídeo Intestinal Vasoativo/antagonistas & inibidores
13.
Blood ; 121(12): 2347-51, 2013 Mar 21.
Artigo em Inglês | MEDLINE | ID: mdl-23325838

RESUMO

Cytomegalovirus (CMV) infection following allogeneic bone marrow transplant (allo-BMT) is controlled by donor-derived cellular immunity. Vasoactive intestinal peptide (VIP) suppresses Th1 immunity. We hypothesized that blocking VIP-signaling would enhance anti-CMV immunity in murine recipients of allo-BMT. Recipients were transplanted with bone marrow (BM) and T-cells from major histocompatibility complex (MHC)-mismatched VIP-knockout (KO) or wild-type donors, and treated with 7 daily subcutaneous injections of VIPhyb (peptidic VIP-antagonist) or phosphate-buffered saline (PBS). Genetic and pharmacological blockade of VIP-signaling protected allo-BMT recipients from lethal murine CMV (mCMV) infection, improving survival without increasing graft-versus-host disease. Mice treated with VIPhyb or transplanted with VIP-KO allografts had significantly lower viral loads, increased numbers of mCMV-M45-peptide-MHC-tetramer(+) CD8(+) T-cells, with lower PD-1 expression, and enhanced primary and secondary cellular immune responses after mCMV infection than did PBS-treated mice. These results demonstrate that administration of a VIP antagonist after allo-BMT is a promising safely therapeutic approach to enhance antiviral cellular immunity.


Assuntos
Imunidade Adaptativa/efeitos dos fármacos , Transplante de Medula Óssea/mortalidade , Infecções por Herpesviridae/imunologia , Muromegalovirus/imunologia , Neurotensina/farmacologia , Peptídeo Intestinal Vasoativo/antagonistas & inibidores , Imunidade Adaptativa/genética , Animais , Transplante de Medula Óssea/imunologia , Células Cultivadas , Doença Enxerto-Hospedeiro/complicações , Doença Enxerto-Hospedeiro/tratamento farmacológico , Doença Enxerto-Hospedeiro/genética , Doença Enxerto-Hospedeiro/imunologia , Infecções por Herpesviridae/tratamento farmacológico , Infecções por Herpesviridae/genética , Infecções por Herpesviridae/mortalidade , Hospedeiro Imunocomprometido/efeitos dos fármacos , Hospedeiro Imunocomprometido/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Muromegalovirus/efeitos dos fármacos , Neurotensina/uso terapêutico , Proteínas Recombinantes de Fusão/farmacologia , Proteínas Recombinantes de Fusão/uso terapêutico , Transdução de Sinais/efeitos dos fármacos , Análise de Sobrevida , Transplante Homólogo , Regulação para Cima/efeitos dos fármacos , Regulação para Cima/imunologia , Peptídeo Intestinal Vasoativo/genética , Peptídeo Intestinal Vasoativo/farmacologia , Peptídeo Intestinal Vasoativo/uso terapêutico , Carga Viral/efeitos dos fármacos
14.
J Clin Invest ; 121(10): 4180-91, 2011 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-21926462

RESUMO

Mast cell degranulation is important in the pathogenesis of anaphylaxis and allergic disorders. Many animal venoms contain components that can induce mast cell degranulation, and this has been thought to contribute to the pathology and mortality caused by envenomation. However, we recently reported evidence that mast cells can enhance the resistance of mice to the venoms of certain snakes and that mouse mast cell-derived carboxypeptidase A3 (CPA3) can contribute to this effect. Here, we investigated whether mast cells can enhance resistance to the venom of the Gila monster, a toxic component of that venom (helodermin), and the structurally similar mammalian peptide, vasoactive intestinal polypeptide (VIP). Using 2 types of mast cell-deficient mice, as well as mice selectively lacking CPA3 activity or the chymase mouse mast cell protease-4 (MCPT4), we found that mast cells and MCPT4, which can degrade helodermin, can enhance host resistance to the toxicity of Gila monster venom. Mast cells and MCPT4 also can limit the toxicity associated with high concentrations of VIP and can reduce the morbidity and mortality induced by venoms from 2 species of scorpions. Our findings support the notion that mast cells can enhance innate defense by degradation of diverse animal toxins and that release of MCPT4, in addition to CPA3, can contribute to this mast cell function.


Assuntos
Carboxipeptidases A/fisiologia , Mastócitos/enzimologia , Venenos de Escorpião/toxicidade , Serina Endopeptidases/fisiologia , Peptídeo Intestinal Vasoativo/toxicidade , Peçonhas/toxicidade , Sequência de Aminoácidos , Animais , Carboxipeptidases A/deficiência , Carboxipeptidases A/genética , Peptídeos e Proteínas de Sinalização Intercelular , Lagartos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Mutantes , Dados de Sequência Molecular , Peptídeos/antagonistas & inibidores , Peptídeos/genética , Peptídeos/toxicidade , Venenos de Escorpião/antagonistas & inibidores , Serina Endopeptidases/deficiência , Serina Endopeptidases/genética , Peptídeo Intestinal Vasoativo/antagonistas & inibidores , Peptídeo Intestinal Vasoativo/genética
15.
Pacing Clin Electrophysiol ; 34(9): 1092-9, 2011 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-21793861

RESUMO

BACKGROUND: Atrial electrical remodeling (AER) is one of the mechanisms by which atrial fibrillation (AF) begets AF. It is known that vagal activity increases the propensity for AF. However, vagal effects on AER have not been fully investigated. METHODS: Adult mongrel dogs were divided in four groups: group I, rapid atria pacing (RAP); group II, RAP plus vagal nerve stimulation (VNS); group III, RAP and VNS with atropine (0.2 mg/kg/h, intravenous), and group IV, group III plus vasoactive intestinal polypeptide (VIP) antagonist ([D-p-Cl-Phe(6), Leu(17)]-VIP, 0.125 µg/kg/h). VNS was performed bilaterally through vagosympathetic trunks to achieve second-degree AV block or sinus rate slowing of >30 beats per minute. Atrial effective refractory periods (AERPs) were determined in the coronary sinus and right atrial appendage every hour at drive cycle lengths (DCLs) 350 ms, 300 ms, and 250 ms. RESULTS: During 5 hours RAP with or without VNS, AERP shortened progressively from baseline at both pacing sites and at all DCLs (P < 0.01). Furthermore, RAP-induced AERP shortening was more pronounced with VNS (P < 0.01). With atropine, the AERP shortening during VNS was blunted (P < 0.01), but was still significantly more pronounced than that in group I (P < 0.05). However, VNS effect on AERP shortening was eliminated completely with the combination of atropine and VIP antagonist (P = 0.15 vs group I). CONCLUSION: Increased vagal activity promotes RAP-induced AER, which could not be totally accounted for by cholinergic effect but could be blocked by the combination of atropine and VIP antagonist. Vagally released VIP may have important role in the vagal promotion of AER.


Assuntos
Função Atrial/fisiologia , Estimulação Cardíaca Artificial , Estimulação do Nervo Vago , Animais , Antiarrítmicos/farmacologia , Função Atrial/efeitos dos fármacos , Atropina/farmacologia , Cães , Quimioterapia Combinada , Masculino , Período Refratário Eletrofisiológico/efeitos dos fármacos , Período Refratário Eletrofisiológico/fisiologia , Peptídeo Intestinal Vasoativo/antagonistas & inibidores
16.
Regul Pept ; 166(1-3): 68-75, 2011 Jan 17.
Artigo em Inglês | MEDLINE | ID: mdl-20801165

RESUMO

UNLABELLED: Aims of the present study were to describe the distribution of corticotropin releasing factor (CRF) immunoreactivity in rat small and large intestines, to quantify the percentage of CRF-immunoreactive (CRF-IR) enteric neurons, to reveal possible CRF immunoreactivity in cultured myenteric neurons from rat ileum and to examine if additions of CRF, urocortin 1 (Ucn1), CRF antagonist or vasoactive intestinal peptide (VIP) affect neuronal survival in vitro. Co-localization of CRF- and VIP-immunoreactivity was examined, as well as a possible interplay between CRF and VIP in neuroprotection. Further we wanted to elucidate if mast cells affect neuronal survival via CRF signaling. Networks of CRF-containing nerve cell bodies and fibers were detected in rat intestine. CRF-IR neurons contained to a high degree also VIP. A low number of cultured myenteric neurons was CRF-IR. CRF, Ucn1 or CRF-antagonist did not promote neuronal survival of cultured myenteric neurons, while VIP significantly enhanced neuronal survival. Simultaneous presence of CRF attenuated the VIP mediated increase in neuronal survival. Co-culturing neurons and mast cells resulted in a marked reduction in neuronal survival, not executed via CRF signaling pathways. CONCLUSION: CRF is present in enteric neurons and counteracts the neuroprotective effect of VIP in vitro.


Assuntos
Hormônio Liberador da Corticotropina/metabolismo , Mucosa Intestinal/metabolismo , Neurônios/metabolismo , Animais , Apoptose , Técnicas de Cocultura , Hormônio Liberador da Corticotropina/antagonistas & inibidores , Hormônio Liberador da Corticotropina/farmacologia , Feminino , Íleo/citologia , Íleo/inervação , Intestinos/inervação , Mastócitos/fisiologia , Plexo Mientérico/efeitos dos fármacos , Plexo Mientérico/metabolismo , Neurônios/efeitos dos fármacos , Fármacos Neuroprotetores/antagonistas & inibidores , Fármacos Neuroprotetores/metabolismo , Fragmentos de Peptídeos/farmacologia , Ratos , Ratos Sprague-Dawley , Urocortinas/farmacologia , Peptídeo Intestinal Vasoativo/antagonistas & inibidores , Peptídeo Intestinal Vasoativo/farmacologia
18.
Gastroenterol Clin North Am ; 39(3): 495-507, 2010 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-20951914

RESUMO

According to the World Health Organization, there are approximately 2 billion annual cases of diarrhea worldwide. Diarrhea is the leading cause of death in children younger than 5 years and kills 1.5 million children each year. It is especially prevalent in the developing world, where mortality is related to dehydration, electrolyte disturbance, and the resultant acidosis, and in 2001, it accounted for 1.78 million deaths (3.7% of total deaths) in low- and middle-income countries. However, diarrhea is also a common problem in the developed world, with 211 million to 375 million episodes of infectious diarrheal illnesses in the United States annually, resulting in 73 million physician consultations, 1.8 million hospitalizations, and 3100 deaths. Furthermore, 4% to 5% of the Western population suffers from chronic diarrhea. Given the high prevalence of diarrhea, research has been directed at learning more about the cellular mechanisms underlying diarrheal illnesses in order to develop new medications directed at novel cellular targets. These cellular mechanisms and targets are discussed in this article.


Assuntos
Antidiarreicos/uso terapêutico , Diarreia/tratamento farmacológico , Anti-Infecciosos/uso terapêutico , Bismuto/uso terapêutico , Bloqueadores dos Canais de Cálcio/uso terapêutico , Calmodulina/antagonistas & inibidores , Canais de Cloreto/antagonistas & inibidores , Resina de Colestiramina/uso terapêutico , Diarreia/fisiopatologia , Diarreia/prevenção & controle , Hidratação , Humanos , Entorpecentes/uso terapêutico , Neprilisina/antagonistas & inibidores , Receptores sigma/antagonistas & inibidores , Antagonistas da Serotonina/uso terapêutico , Somatostatina/uso terapêutico , Substância P/antagonistas & inibidores , Peptídeo Intestinal Vasoativo/antagonistas & inibidores
19.
Diabetes ; 59(7): 1591-600, 2010 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-20357362

RESUMO

OBJECTIVE: The unraveling of the elaborate brain networks that control glucose metabolism presents one of the current challenges in diabetes research. Within the central nervous system, the hypothalamus is regarded as the key brain area to regulate energy homeostasis. The aim of the present study was to investigate the hypothalamic mechanism involved in the hyperglycemic effects of the neuropeptide pituitary adenylyl cyclase-activating polypeptide (PACAP). RESEARCH DESIGN AND METHODS: Endogenous glucose production (EGP) was determined during intracerebroventricular infusions of PACAP-38, vasoactive intestinal peptide (VIP), or their receptor agonists. The specificity of their receptors was examined by coinfusions of receptor antagonists. The possible neuronal pathway involved was investigated by 1) local injections in hypothalamic nuclei, 2) retrograde neuronal tracing from the thoracic spinal cord to hypothalamic preautonomic neurons together with Fos immunoreactivity, and 3) specific hepatic sympathetic or parasympathetic denervation to block the autonomic neuronal input to liver. RESULTS: Intracerebroventricular infusion of PACAP-38 increased EGP to a similar extent as a VIP/PACAP-2 (VPAC2) receptor agonist, and intracerebroventricular administration of VIP had significantly less influence on EGP. The PACAP-38 induced increase of EGP was significantly suppressed by preinfusion of a VPAC2 but not a PAC1 receptor antagonist, as well as by hepatic sympathetic but not parasympathetic denervation. In the hypothalamus, Fos immunoreactivity induced by PACAP-38 was colocalized within autonomic neurons in paraventricular nuclei projecting to preganglionic sympathetic neurons in the spinal cord. Local infusion of PACAP-38 directly into the PVN induced a significant increase of EGP. CONCLUSIONS: This study demonstrates that PACAP-38 signaling via sympathetic preautonomic neurons located in the paraventricular nucleus is an important component in the hypothalamic control of hepatic glucose production.


Assuntos
Glucose/biossíntese , Fígado/metabolismo , Núcleo Hipotalâmico Paraventricular/metabolismo , Polipeptídeo Hipofisário Ativador de Adenilato Ciclase/metabolismo , Sistema Nervoso Simpático/metabolismo , Análise de Variância , Animais , Homeostase , Injeções Intraventriculares , Fígado/efeitos dos fármacos , Fígado/inervação , Masculino , Marcadores do Trato Nervoso , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Núcleo Hipotalâmico Paraventricular/efeitos dos fármacos , Polipeptídeo Hipofisário Ativador de Adenilato Ciclase/antagonistas & inibidores , Polipeptídeo Hipofisário Ativador de Adenilato Ciclase/farmacologia , Proteínas Proto-Oncogênicas c-fos/metabolismo , Ratos , Ratos Wistar , Peptídeo Intestinal Vasoativo/antagonistas & inibidores , Peptídeo Intestinal Vasoativo/metabolismo , Peptídeo Intestinal Vasoativo/farmacologia
20.
Mol Cell Biol ; 30(10): 2537-51, 2010 May.
Artigo em Inglês | MEDLINE | ID: mdl-20231362

RESUMO

Vasoactive intestinal peptide (VIP) is a potent anti-inflammatory neuropeptide that, by inhibiting Th1-driven responses and inducing the emergence of regulatory T cells (T(reg)), has been proven successful in the induction of tolerance in various experimental models of autoimmune disorders. Here, we investigate the molecular mechanisms involved in VIP-induced tolerance. VIP treatment in the presence of T-cell receptor (TCR) signaling and CD28 costimulation induced cell cycle arrest in human T cells. VIP blocked G(1)/S transition and inhibited the synthesis of cyclins D3 and E and the activation of the cyclin-dependent kinases (CDKs) cdk2 and cdk4. This effect was accompanied by maintenance of threshold levels of the CDK inhibitor p27(kip1) and impairment of phosphatidylinositol 3-kinase (PI3K)-Akt signaling. Inhibition of interleukin 2 (IL-2) transcription and downregulation of signaling through NFAT, AP-1, and Ras-Raf paralleled the VIP-induced cell cycle arrest. Noteworthy from a functional point of view is the fact that VIP-treated T cells show a regulatory phenotype characterized by high expression of CD25, cytotoxic-T-lymphocyte-associated protein 4 (CTLA4), and Forkhead box protein 3 (FoxP3) and potent suppressive activities against effector T cells. CTLA4 appears to be critically involved in the generation and suppressive activities of VIP-induced T(reg). Finally, cyclic AMP (cAMP) and protein kinase A (PKA) activation seems to mediate the VIP-induced cell cycle arrest and T(reg) generation.


Assuntos
Ciclo Celular/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos , Subpopulações de Linfócitos T/metabolismo , Linfócitos T/efeitos dos fármacos , Linfócitos T/metabolismo , Peptídeo Intestinal Vasoativo/farmacologia , Antígenos CD/genética , Antígenos CD/metabolismo , Antígenos CD28/metabolismo , Complexo CD3/metabolismo , Antígeno CTLA-4 , Ciclo Celular/fisiologia , AMP Cíclico/metabolismo , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Inibidor de Quinase Dependente de Ciclina p27/genética , Inibidor de Quinase Dependente de Ciclina p27/metabolismo , Citocinas/metabolismo , Proteína Forkhead Box O1 , Fatores de Transcrição Forkhead/genética , Fatores de Transcrição Forkhead/metabolismo , Humanos , Interleucina-2/metabolismo , Ativação Linfocitária , Fragmentos de Peptídeos/genética , Fragmentos de Peptídeos/farmacologia , Receptores de Antígenos de Linfócitos T/metabolismo , Subpopulações de Linfócitos T/citologia , Subpopulações de Linfócitos T/efeitos dos fármacos , Linfócitos T/citologia , Peptídeo Intestinal Vasoativo/agonistas , Peptídeo Intestinal Vasoativo/antagonistas & inibidores , Peptídeo Intestinal Vasoativo/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA