Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 85
Filtrar
1.
J Invest Dermatol ; 141(7): 1687-1698, 2021 07.
Artigo em Inglês | MEDLINE | ID: mdl-33600826

RESUMO

BACKGROUND: The pathogenesis of rosacea is incompletely understood. Signaling neuropeptides, including PACAP, a regulator of vasodilation and edema, are upregulated in rosacea skin. Here, we evaluated PACAP38-induced rosacea features and examined whether a 5-HT1B/1D receptor agonist could reduce these features. METHODS: A total of 35 patients with erythematotelangiectatic rosacea received an intravenous infusion of 10 pmol/kg/minute of PACAP38 followed by an intravenous infusion of 4 mg sumatriptan or placebo (saline) on two study days in a double-blind, randomized, placebo-controlled, and cross-over trial. RESULTS: PACAP38 increased facial skin blood flow by 90%, dilated the superficial temporal artery by 56%, and induced prolonged flushing and facial edema. Compared with placebo, sumatriptan reduced PACAP38-induced facial skin blood flow for 50 minutes (P = 0.023), constricted the superficial temporal artery for 80 minutes (P = 0.010), and reduced duration of flushing (P = 0.001) and facial edema (P < 0.001). CONCLUSIONS: We established a clinical experimental model of rosacea features and showed that sumatriptan was able to attenuate PACAP38-induced rosacea flushing and edema. Findings support a key role of PACAP38 in rosacea flushing pathogenesis. It remains unknown whether PACAP38 inhibition can improve rosacea. TRIAL REGISTER: The trial was registered at ClinicalTrials.govNCT03878784 in March 2019.


Assuntos
Edema/tratamento farmacológico , Rubor/tratamento farmacológico , Polipeptídeo Hipofisário Ativador de Adenilato Ciclase/imunologia , Rosácea/tratamento farmacológico , Sumatriptana/farmacologia , Adulto , Estudos Cross-Over , Método Duplo-Cego , Edema/imunologia , Face , Feminino , Rubor/imunologia , Humanos , Infusões Intravenosas , Masculino , Pessoa de Meia-Idade , Polipeptídeo Hipofisário Ativador de Adenilato Ciclase/administração & dosagem , Rosácea/imunologia , Sumatriptana/uso terapêutico , Resultado do Tratamento , Adulto Jovem
2.
Front Immunol ; 12: 782566, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34992601

RESUMO

Host inflammatory immune response comprises an essential element of the bone healing process, where M2 polarization allegedly contributes to a favorable healing outcome. In this context, immunoregulatory molecules that modulate host response, including macrophage polarization, are considered potential targets for improving bone healing. This study aims to evaluate the role of the immunoregulatory molecules VIP (Vasoactive intestinal peptide) and PACAP (Pituitary adenylate cyclase activating polypeptide), which was previously described to favor the development of the M2 phenotype, in the process of alveolar bone healing in C57Bl/6 (WT) mice. Experimental groups were submitted to tooth extraction and maintained under control conditions or treated with VIP or PACAP were evaluated by microtomographic (µCT), histomorphometric, immunohistochemical, and molecular analysis at 0, 3, 7, and 14 days to quantify tissue healing and host response indicators at the healing site. Gene expression analysis demonstrates the effectiveness of VIP or PACAP in modulating host response, evidenced by the early dominance of an M2-type response, which was paralleled by a significant increase in M2 (CD206+) in treated groups. However, despite the marked effect of M1/M2 balance in the healing sites, the histomorphometric analysis does not reveal an equivalent/corresponding modulation of the healing process. µCT reveals a slight increase in bone matrix volume and the trabecular thickness number in the PACAP group, while histomorphometric analyzes reveal a slight increase in the VIP group, both at a 14-d time-point; despite the increased expression of osteogenic factors, osteoblastic differentiation, activity, and maturation markers in both VIP and PACAP groups. Interestingly, a lower number of VIP and PACAP immunolabeled cells were observed in the treated groups, suggesting a reduction in endogenous production. In conclusion, while both VIP and PACAP treatments presented a significant immunomodulatory effect with potential for increased healing, no major changes were observed in bone healing outcome, suggesting that the signals required for bone healing under homeostatic conditions are already optimal, and additional signals do not improve an already optimal process. Further studies are required to elucidate the role of macrophage polarization in the bone healing process.


Assuntos
Processo Alveolar/lesões , Ativação de Macrófagos/efeitos dos fármacos , Polipeptídeo Hipofisário Ativador de Adenilato Ciclase/administração & dosagem , Peptídeo Intestinal Vasoativo/administração & dosagem , Cicatrização/imunologia , Processo Alveolar/efeitos dos fármacos , Processo Alveolar/imunologia , Processo Alveolar/cirurgia , Animais , Diferenciação Celular/efeitos dos fármacos , Diferenciação Celular/imunologia , Modelos Animais de Doenças , Feminino , Imunomodulação/efeitos dos fármacos , Masculino , Camundongos , Osteoblastos/fisiologia , Osteogênese/efeitos dos fármacos , Osteogênese/imunologia , Extração Dentária/efeitos adversos , Cicatrização/efeitos dos fármacos , Microtomografia por Raio-X
3.
Addict Biol ; 26(3): e12972, 2021 05.
Artigo em Inglês | MEDLINE | ID: mdl-33020973

RESUMO

While limited research has implicated the neuropeptide, pituitary adenylate cyclase-activating polypeptide (PACAP), in problematic alcohol use, the brain regions and isoforms involved in this effect remain to be determined. One region that has been found both to exhibit PACAP binding and, separately, to be involved in ethanol drinking is the nucleus accumbens (NAc). Thus, this study sought to characterize the effect of the PACAP isoforms in the NAc on ethanol drinking under the intermittent-access two-bottle-choice paradigm, in male and female Long-Evans rats. With microinjection into the medial NAc shell, PACAP-27 but not PACAP-38 was found to dose-dependently reduce binge-like ethanol drinking. In contrast, the PACAP receptor antagonist, PACAP (6-27), but not PACAP (6-38), enhanced ethanol drinking. This effect of PACAP was substance specific, as neither isoform in the NAc shell affected binge-like sucrose drinking. It was also anatomically specific, as PACAP-38 rather than PACAP-27 suppressed ethanol drinking when injected into the NAc core, and PACAP-27 instead enhanced drinking when injected into the caudal third of the medial NAc shell. Finally, while PACAP-38 in the NAc shell affected stress-related exploratory behavior, reducing time spent in the light chamber of a light-dark box, PACAP-27 did not significantly affect behavior in a light-dark box or open field. Together, these results, showing that PACAP-27 in the NAc shell attenuates binge-like ethanol drinking without affecting select stress-related behaviors, suggest that compounds related to this PACAP isoform should be investigated as potential novel therapeutics for the treatment of alcohol use disorder.


Assuntos
Consumo de Bebidas Alcoólicas/tratamento farmacológico , Núcleo Accumbens/efeitos dos fármacos , Polipeptídeo Hipofisário Ativador de Adenilato Ciclase/administração & dosagem , Animais , Feminino , Masculino , Microinjeções , Isoformas de Proteínas , Ratos , Ratos Long-Evans
4.
Cephalalgia ; 40(13): 1474-1488, 2020 11.
Artigo em Inglês | MEDLINE | ID: mdl-32962406

RESUMO

BACKGROUND: Previously reported increases in serum levels of vasodilating neuropeptides pituitary adenylate cyclase-activating peptide-38 (PACAP38) and vasoactive intestinal peptide (VIP) during attacks of cluster headache could indicate their involvement in cluster headache attack initiation. We investigated the attack-inducing effects of PACAP38 and vasoactive intestinal peptide in cluster headache, hypothesising that PACAP38, but not vasoactive intestinal peptide, would induce cluster-like attacks in episodic active phase and chronic cluster headache patients. METHODS: In a double-blind crossover study, 14 episodic cluster headache patients in active phase, 15 episodic cluster headache patients in remission phase and 15 chronic cluster headache patients were randomly allocated to receive intravenous infusion of PACAP38 (10 pmol/kg/min) or vasoactive intestinal peptide (8 pmol/kg/min) over 20 min on two study days separated by at least 7 days. We recorded headache intensity, incidence of cluster-like attacks, cranial autonomic symptoms and vital signs using a questionnaire (0-90 min). RESULTS: In episodic cluster headache active phase, PACAP38 induced cluster-like attacks in 6/14 patients and vasoactive intestinal peptide induced cluster-like attacks in 5/14 patients (p = 1.000). In chronic cluster headache, PACAP38 and vasoactive intestinal peptide both induced cluster-like attacks in 7/15 patients (p = 0.765). In episodic cluster headache remission phase, neither PACAP38 nor vasoactive intestinal peptide induced cluster-like attacks. CONCLUSIONS: Contrary to our hypothesis, attack induction was lower than expected and roughly equal by PACAP38 and vasoactive intestinal peptide in episodic active phase and chronic cluster headache patients, which contradicts the PAC1-receptor as being solely responsible for attack induction.Trial registration: clinicaltrials.gov (identifier NCT03814226).


Assuntos
Cefaleia Histamínica/tratamento farmacológico , Polipeptídeo Hipofisário Ativador de Adenilato Ciclase/administração & dosagem , Peptídeo Intestinal Vasoativo/administração & dosagem , Adenilil Ciclases , Adolescente , Adulto , Idoso , Estudos Cross-Over , Método Duplo-Cego , Feminino , Humanos , Infusões Intravenosas , Masculino , Pessoa de Meia-Idade , Polipeptídeo Hipofisário Ativador de Adenilato Ciclase/farmacologia , Resultado do Tratamento , Vasodilatação/efeitos dos fármacos , Vasodilatadores/farmacologia
5.
Neurotherapeutics ; 17(4): 1954-1972, 2020 10.
Artigo em Inglês | MEDLINE | ID: mdl-32918234

RESUMO

Brain edema is a vital contributor to early brain injury after subarachnoid hemorrhage (SAH), which is responsible for prolonged hospitalization and poor outcomes. Pharmacological therapeutic targets on edema formation have been the focus of research for decades. Pituitary adenylate cyclase-activating polypeptide (PACAP) has been shown to participate in neural development and brain injury. Here, we used PACAP knockout CRISPR to demonstrate that endogenous PACAP plays an endogenous neuroprotective role against brain edema formation after SAH in rats. The exogenous PACAP treatment provided both short- and long-term neurological benefits by preserving the function of the blood-brain barrier and glymphatic system after SAH. Pretreatment of inhibitors of PACAP receptors showed that the PACAP-involved anti-edema effect and neuroprotection after SAH was facilitated by the selective PACAP receptor (PAC1). Further administration of adenylyl cyclase (AC) inhibitor and sulfonylurea receptor 1 (SUR1) CRISPR activator suggested that the AC-cyclic adenosine monophosphate (cAMP)-protein kinase A (PKA) axis participated in PACAP signaling after SAH, which inhibited the expression of edema-related proteins, SUR1 and aquaporin-4 (AQP4), through SUR1 phosphorylation. Thus, PACAP may serve as a potential clinical treatment to alleviate brain edema in patients with SAH.


Assuntos
Barreira Hematoencefálica/efeitos dos fármacos , Edema Encefálico/tratamento farmacológico , Sistema Glinfático/efeitos dos fármacos , Polipeptídeo Hipofisário Ativador de Adenilato Ciclase/administração & dosagem , Hemorragia Subaracnóidea/tratamento farmacológico , Animais , Barreira Hematoencefálica/metabolismo , Barreira Hematoencefálica/patologia , Edema Encefálico/metabolismo , Edema Encefálico/patologia , Sistema Glinfático/metabolismo , Sistema Glinfático/patologia , Masculino , Ratos , Ratos Sprague-Dawley , Hemorragia Subaracnóidea/metabolismo , Hemorragia Subaracnóidea/patologia
6.
Immunohorizons ; 4(4): 191-200, 2020 04 17.
Artigo em Inglês | MEDLINE | ID: mdl-32303568

RESUMO

The purpose of this study was to investigate whether pituitary adenylate cyclase-activating polypeptide (PACAP) prevents mortality due to sepsis in mice. Mice were given PACAP at designated time points before or after cecal ligation and puncture (CLP), and organ injury and mortality were investigated. Serum inflammatory and anti-inflammatory cytokine levels were assessed after CLP. Plasma corticosterone and adrenocorticotropic hormone levels were also measured. Isolated tissue macrophages (Mfs) were incubated with or without PACAP, and production of cytokines was measured. Activation of NF-κB was investigated in tissue Mfs isolated from CLP animal in the presence and absence PACAP in vitro. PACAP treatment significantly prevented acute lung injury and mortality after CLP. Plasma endotoxin levels and bacterial load were not different between PACAP-treated and nontreated groups. Increased serum TNF-α and HMGB1 levels in animals treated with vehicle were significantly blunted in PACAP-treated animals after CLP. Furthermore, serum IL-10 levels were significantly greater in the PACAP-treated group compared with the vehicle group. Production of HMGB1 and TNF-α by isolated hepatic Mfs was significantly inhibited in the presence of PACAP, whereas production of IL-10 by isolated hepatic Mfs and interstitial lung Mfs was significantly increased. Plasma corticosterone and adrenocorticotropic hormone levels were significantly greater in the animals treated with PACAP compared with vehicle after CLP. Activation of NF-κB was significantly inhibited by PACAP in the hepatic Mfs compared with other tissue Mfs. PACAP prevents mortality due to septic peritonitis by inhibiting inflammation via NF-κB activation and possible effects on the brain.


Assuntos
Lesão Pulmonar Aguda/etiologia , Lesão Pulmonar Aguda/prevenção & controle , Peritonite/complicações , Peritonite/tratamento farmacológico , Polipeptídeo Hipofisário Ativador de Adenilato Ciclase/administração & dosagem , Sepse/complicações , Sepse/tratamento farmacológico , Lesão Pulmonar Aguda/mortalidade , Hormônio Adrenocorticotrópico/sangue , Animais , Células Cultivadas , Corticosterona/sangue , Citocinas/sangue , Modelos Animais de Doenças , Proteína HMGB1/metabolismo , Macrófagos/efeitos dos fármacos , Macrófagos/imunologia , Camundongos , Camundongos Endogâmicos C57BL , NF-kappa B/metabolismo , Peritonite/sangue , Peritonite/mortalidade , Sepse/sangue , Sepse/mortalidade , Transdução de Sinais/efeitos dos fármacos , Resultado do Tratamento , Fator de Necrose Tumoral alfa/metabolismo
7.
J Headache Pain ; 21(1): 19, 2020 Feb 24.
Artigo em Inglês | MEDLINE | ID: mdl-32093617

RESUMO

BACKGROUND: Pituitary adenylate cyclase-activating polypeptide-38 (PACAP38) induces headache in healthy volunteers but the precise mechanisms by which PACAP38 leads to headache are unclear. We investigated the headache preventive effect of sumatriptan and ketorolac on PACAP38-induced headache in healthy volunteers. In addition, we explored contribution of vascular mechanisms to PACAP38-induced headache using high resolution magnetic resonance angiography. METHODS: Thirty-four healthy volunteers were divided in two groups (A and B) and received infusion of PACAP38 (10 picomol/kg/min) over 20 min. Group A was pretreated with intravenous sumatriptan (4 mg) or ketorolac (30 mg) 20 min before infusion of PACAP38. Group B received infusion of sumatriptan or ketorolac as post-treatment 90 min after infusion of PACAP38. In both experiments, we used a randomized, double-blind, cross-over design. We recorded headache characteristics and circumference of extra-intracerebral arteries. RESULTS: We found no difference in AUC (0-6 h) of PACAP38-induced headache in group A, pretreated with sumatriptan or ketorolac (p = 0.297). There was no difference between sumatriptan and ketorolac in PACAP38-induced circumference change (AUCBaseline-110 min) of MMA (p = 0.227), STA (p = 0.795) and MCA (p = 0.356). In group B, post-treatment with ketorolac reduced PACAP38-headache compared to sumatriptan (p < 0.001). Post-treatment with sumatriptan significantly reduced the circumference of STA (p = 0.039) and MMA (p = 0.015) but not of MCA (p = 0.981) compared to ketorolac. In an explorative analysis, we found that pre-treatment with sumatriptan reduced PACAP38-induced headache compared to no treatment (AUC0-90min). CONCLUSIONS: Post-treatment with ketorolac was more effective in attenuating PACAP38-induced headache compared to sumatriptan. Ketorolac exerted its effect without affecting PACAP38-induced arterial dilation, whereas sumatriptan post-treatment attenuated PACAP38-induced dilation of MMA and STA. Pre-treatment with sumatriptan attenuated PACAP38-induced headache without affecting PACAP38-induced arterial dilation. Our findings suggest that ketorolac and sumatriptan attenuated PACAP38-induced headache in healthy volunteers without vascular effects. TRIAL REGISTRATION: Clinicaltrials.gov (NCT03585894). Registered 13 July 2018.


Assuntos
Cefaleia/induzido quimicamente , Cefaleia/tratamento farmacológico , Cetorolaco/administração & dosagem , Medição da Dor/efeitos dos fármacos , Polipeptídeo Hipofisário Ativador de Adenilato Ciclase/efeitos adversos , Sumatriptana/administração & dosagem , Adulto , Anti-Inflamatórios não Esteroides/administração & dosagem , Estudos Cross-Over , Método Duplo-Cego , Feminino , Cefaleia/diagnóstico , Humanos , Infusões Intravenosas , Angiografia por Ressonância Magnética , Masculino , Modelos Teóricos , Medição da Dor/métodos , Polipeptídeo Hipofisário Ativador de Adenilato Ciclase/administração & dosagem , Agonistas do Receptor 5-HT1 de Serotonina/administração & dosagem , Vasodilatadores/administração & dosagem , Vasodilatadores/efeitos adversos
8.
Front Endocrinol (Lausanne) ; 11: 585082, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33551991

RESUMO

Intranasal (IN) administration appears to be a suitable route for clinical use as it allows direct delivery of bioactive molecules to the central nervous system, reducing systemic exposure and sides effects. Nevertheless, only some molecules can be transported to the brain from the nasal cavity. This led us to compare the efficiency of an IN, intravenous (IV), and intraperitoneal (IP) administration of pituitary adenylate cyclase-activating polypeptide (PACAP) after transient or permanent middle cerebral artery occlusion (MCAO) in C57BL/6 mice. The results show that the neuroprotective effect of PACAP is much more efficient after IN administration than IV injection while IP injection had no effect. IN administration of PACAP reduced the infarct volume when injected within 6 h after the reperfusion and improved functional recovery up to at least 1 week after the ischemia.


Assuntos
Infarto Encefálico/tratamento farmacológico , Infarto da Artéria Cerebral Média/complicações , Fármacos Neuroprotetores/administração & dosagem , Neurotransmissores/administração & dosagem , Polipeptídeo Hipofisário Ativador de Adenilato Ciclase/administração & dosagem , Recuperação de Função Fisiológica , Administração Intranasal , Animais , Infarto Encefálico/etiologia , Infarto Encefálico/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL
9.
Cephalalgia ; 40(1): 57-67, 2020 01.
Artigo em Inglês | MEDLINE | ID: mdl-31299857

RESUMO

INTRODUCTION: Pituitary adenylate cyclase-activating polypeptide (PACAP) is found in two functional isoforms, namely PACAP38 and PACAP27. The migraine-inducing properties of PACAP38 are well studied. However, it is not known whether the lesser-known and under-studied protein isoform, PACAP27, can also induce migraine attacks. Here, we studied the effect of human PACAP27 infusion on induction of migraine in a provocation model. METHODS: In a crossover study, 20 migraine without aura patients were randomly assigned to receive human PACAP27 (10 picomol/kg/min) or saline (placebo) infusion over 20 min. We recorded the migraine and associated symptoms. RESULTS: All patients completed the study. PACAP27 provoked migraine-like attacks in 11 patients (55%) and two developed attacks after placebo (10%) (p = 0.022). The headache intensity and duration after PACAP27 was significantly greater compared to placebo (p = 0.003). CONCLUSION: PACAP27 triggers migraine attacks without aura. These novel data strengthen the role of PACAP and its receptors in migraine pathogenesis.


Assuntos
Enxaqueca sem Aura/induzido quimicamente , Enxaqueca sem Aura/diagnóstico , Polipeptídeo Hipofisário Ativador de Adenilato Ciclase/administração & dosagem , Polipeptídeo Hipofisário Ativador de Adenilato Ciclase/efeitos adversos , Vasodilatadores/administração & dosagem , Vasodilatadores/efeitos adversos , Adolescente , Adulto , Velocidade do Fluxo Sanguíneo/efeitos dos fármacos , Velocidade do Fluxo Sanguíneo/fisiologia , Estudos Cross-Over , Método Duplo-Cego , Feminino , Humanos , Infusões Intravenosas , Masculino , Pessoa de Meia-Idade , Adulto Jovem
10.
Int J Mol Sci ; 20(19)2019 Oct 07.
Artigo em Inglês | MEDLINE | ID: mdl-31591326

RESUMO

Pituitary adenylate cyclase-activating polypeptide (PACAP) is a pleiotropic neuropeptide with a widespread distribution throughout the entire body including the urinary system. PACAP exerts protective actions in different injury models related to several organ systems. Its protective effect is mainly based on its antiapoptotic, anti-inflammatory and antioxidant effects. The present review aims to summarize the effects of PACAP in pathologies associated with inflammation and oxidative stress-induced damage in the kidney. Both in vitro and in vivo data are available proving its protective actions against oxidative stress, hypoxia, renal ischemia/reperfusion, diabetic nephropathy, myeloma kidney injury, amyloidosis and different types of drug-induced nephropathies. Data showing the nephroprotection by PACAP emphasize the potential of PACAP's therapeutic use in various renal pathologies.


Assuntos
Anti-Inflamatórios/administração & dosagem , Rim/lesões , Polipeptídeo Hipofisário Ativador de Adenilato Ciclase/administração & dosagem , Animais , Anti-Inflamatórios/farmacologia , Antioxidantes/administração & dosagem , Antioxidantes/farmacologia , Apoptose/efeitos dos fármacos , Sobrevivência Celular , Citocinas/metabolismo , Humanos , Rim/efeitos dos fármacos , Estresse Oxidativo/efeitos dos fármacos , Polipeptídeo Hipofisário Ativador de Adenilato Ciclase/farmacologia
11.
Neuropharmacology ; 160: 107761, 2019 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-31493466

RESUMO

Anxiety-related disorders are the most prevalent mental disorders in the world and they are characterized by abnormal responses to stressors. Pituitary adenylate cyclase-activating polypeptide (PACAP) is a neuropeptide highly expressed in the extended amygdala, a brain macrostructure involved in the response to threat that includes the central nucleus of the amygdala (CeA) and the bed nucleus of the stria terminalis (BNST). The aim of this series of experiments was to systematically elucidate the role of the PACAP system of the CeA and BNST under both control, unstressed conditions and after the presentation of a stressor in rats. For this purpose, we used the acoustic startle response (ASR), an unconscious response to sudden acoustic stimuli sensitive to changes in stress which can be used as an operationalization of the hypervigilance present in anxiety- and trauma-related disorders. We found that infusion of PACAP, but not the related peptide vasoactive intestinal peptide (VIP), into either the CeA or the BNST causes a dose-dependent increase in ASR. In addition, while infusion of the antagonist PACAP(6-38) into either the CeA or the BNST does not affect ASR in non-stressed conditions, it prevents the sensitization of ASR induced by an acute footshock stress. Finally, we found that footshock stress induces a significant increase in PACAP, but not VIP, levels in both of these brain areas. Altogether, these data show that the PACAP system of the extended amygdala contributes to stress-induced hyperarousal and suggest it as a potential novel target for the treatment of stress-related disorders.


Assuntos
Núcleo Central da Amígdala/efeitos dos fármacos , Polipeptídeo Hipofisário Ativador de Adenilato Ciclase/farmacologia , Reflexo de Sobressalto/efeitos dos fármacos , Núcleos Septais/efeitos dos fármacos , Estresse Psicológico , Animais , Ansiedade/metabolismo , Ansiedade/patologia , Comportamento Animal/efeitos dos fármacos , Núcleo Central da Amígdala/metabolismo , Masculino , Polipeptídeo Hipofisário Ativador de Adenilato Ciclase/administração & dosagem , Ratos , Ratos Wistar , Núcleos Septais/metabolismo , Peptídeo Intestinal Vasoativo/metabolismo
12.
Peptides ; 121: 170134, 2019 11.
Artigo em Inglês | MEDLINE | ID: mdl-31449829

RESUMO

Pituitary adenylate cyclase-activating polypeptide (PACAP) has emerged as an important signaling peptide in migraine pathogenesis. Recently, we have shown that the less-abundant PACAP isoform, PACAP27, induced migraine and headache in patients equipotently to PACAP38. The present study examined the effect of PACAP27 on cerebral hemodynamics in healthy volunteers using high resolution magnetic resonance angiography (MRA). Eighteen healthy volunteers received infusion of PACAP27 (10 pmol/kg/min) or placebo over 20 min and were scanned repeatedly in fixed intervals for 5 h in a double-blind, randomized, placebo-controlled study. The circumference of extra-intracerebral arteries was measured and compared with PACAP38 data. We found significant dilation of middle meningeal artery (MMA) (p = 0.019), superficial temporal artery (p = 0.001) and external carotid artery (p = 0.039) after PACAP27 infusion compared to placebo. Whereas the middle cerebral artery (MCA) (p = 0.011) and internal carotid artery (ICA) (pICAcervical = 0.015, pICAcerebral = 0.019) were constricted. No effects on basilar artery (p = 0.708) and cavernous portion of ICA were found. Post hoc analyses revealed significant larger area under the curve for MMA after PACAP38 compared to PACAP27 (p = 0.033). We also found that PACAP27 induced headache in nine out of twelve (75%) volunteers and one (17%) after placebo. In conclusion, PACAP27 induced headache and dilated extracerebral arteries (>5 h) and slightly constricted MCA in healthy volunteers. Post hoc analysis of PACAP38 data compared with PACAP27 showed that PACAP isoforms dilates MMA with significantly different magnitude.


Assuntos
Circulação Cerebrovascular/efeitos dos fármacos , Cefaleia/fisiopatologia , Polipeptídeo Hipofisário Ativador de Adenilato Ciclase/efeitos adversos , Vasoconstrição/efeitos dos fármacos , Vasodilatação/efeitos dos fármacos , Adolescente , Adulto , Área Sob a Curva , Artérias Carótidas/diagnóstico por imagem , Artérias Carótidas/efeitos dos fármacos , Artérias Carótidas/fisiologia , Circulação Cerebrovascular/fisiologia , Método Duplo-Cego , Feminino , Cefaleia/induzido quimicamente , Cefaleia/diagnóstico por imagem , Voluntários Saudáveis , Humanos , Angiografia por Ressonância Magnética , Masculino , Artérias Meníngeas/diagnóstico por imagem , Artérias Meníngeas/efeitos dos fármacos , Artérias Meníngeas/fisiologia , Artéria Cerebral Média/diagnóstico por imagem , Artéria Cerebral Média/efeitos dos fármacos , Artéria Cerebral Média/fisiologia , Polipeptídeo Hipofisário Ativador de Adenilato Ciclase/administração & dosagem , Artérias Temporais/diagnóstico por imagem , Artérias Temporais/efeitos dos fármacos , Artérias Temporais/fisiologia , Vasoconstrição/fisiologia , Vasodilatação/fisiologia
13.
Artigo em Inglês | MEDLINE | ID: mdl-31192159

RESUMO

Pituitary Adenylate Cyclase-Activating Polypeptide (PACAP) is an endogenous neuropeptide with distinct functions including the regulation of inflammatory processes. PACAP is able to modify the immune response by directly regulating macrophages and monocytes inhibiting the production of inflammatory cytokines, chemokines and free radicals. Here, we analyzed the effect of exogenous PACAP on peripheral immune cell subsets upon acute infection with the parasite Toxoplasma gondii (T. gondii). PACAP administration was followed by diminished innate immune cell recruitment to the peritoneal cavity of T. gondii-infected mice. PACAP did not directly interfere with parasite replication, instead, indirectly reduced parasite burden in mononuclear cell populations by enhancing their phagocytic capacity. Although proinflammatory cytokine levels were attenuated in the periphery upon PACAP treatment, interleukin (IL)-10 and Transforming growth factor beta (TGF-ß) remained stable. While PACAP modulated VPAC1 and VPAC2 receptors in immune cells upon binding, it also increased their expression of brain-derived neurotrophic factor (BDNF). In addition, the expression of p75 neurotrophin receptor (p75NTR) on Ly6Chi inflammatory monocytes was diminished upon PACAP administration. Our findings highlight the immunomodulatory effect of PACAP on peripheral immune cell subsets during acute Toxoplasmosis, providing new insights about host-pathogen interaction and the effects of neuropeptides during inflammation.


Assuntos
Interações Hospedeiro-Patógeno/imunologia , Imunomodulação , Neuropeptídeos/imunologia , Polipeptídeo Hipofisário Ativador de Adenilato Ciclase/administração & dosagem , Polipeptídeo Hipofisário Ativador de Adenilato Ciclase/imunologia , Toxoplasmose/imunologia , Animais , Antígenos Ly , Fator Neurotrófico Derivado do Encéfalo/metabolismo , Quimiocinas/metabolismo , Citocinas/metabolismo , Modelos Animais de Doenças , Feminino , Imunidade Inata , Inflamação , Interleucina-10 , Macrófagos , Camundongos , Camundongos Endogâmicos C57BL , Fatores de Crescimento Neural , Receptores Tipo II de Peptídeo Intestinal Vasoativo , Receptores Tipo I de Polipeptídeo Intestinal Vasoativo , Toxoplasma , Regulação para Cima
14.
Sci Rep ; 9(1): 9183, 2019 06 24.
Artigo em Inglês | MEDLINE | ID: mdl-31235716

RESUMO

The blood-brain barrier (BBB) regulates the traffic of molecules into the central nervous system (CNS) and also limits the drug delivery. Due to their flexible properties, liposomes are an attractive tool to deliver drugs across the BBB. We previously characterized gH625, a peptide derived from Herpes simplex virus 1. The present study investigates the efficiency of liposomes functionalized on their surface with gH625 to promote the brain uptake of neuroprotective peptide PACAP (pituitary adenylate cyclase-activating polypeptide). Using a rat in vitro BBB model, we showed that the liposomes preparations were non-toxic for the endothelial cells, as assessed by analysis of tight junction protein ZO1 organization and barrier integrity. Next, we found that gH625 improves the transfer of liposomes across endothelial cell monolayers, resulting in both low cellular uptake and increased transport of PACAP. Finally, in vivo results demonstrated that gH625 ameliorates the efficiency of liposomes to deliver PACAP to the mouse brain after intravenous administration. gH625-liposomes improve both PACAP reaching and crossing the BBB, as showed by the higher number of brain cells labelled with PACAP. gH625-liposomes represent a promising strategy to deliver therapeutic agents to CNS and to provide an effective imaging and diagnostic tool for the brain.


Assuntos
Barreira Hematoencefálica/efeitos dos fármacos , Sistemas de Liberação de Medicamentos , Lipossomos/farmacocinética , Peptídeos/farmacocinética , Polipeptídeo Hipofisário Ativador de Adenilato Ciclase/farmacocinética , Proteínas do Envelope Viral/farmacocinética , Administração Intravenosa , Animais , Transporte Biológico , Células Cultivadas , Células Endoteliais/citologia , Células Endoteliais/metabolismo , Camundongos , Polipeptídeo Hipofisário Ativador de Adenilato Ciclase/administração & dosagem , Ratos , Ratos Wistar
15.
Neurobiol Learn Mem ; 157: 24-34, 2019 01.
Artigo em Inglês | MEDLINE | ID: mdl-30458282

RESUMO

The stress-related neuropeptide pituitary adenylate cyclase-activating polypeptide (PACAP) is implicated in neuromodulation of learning and memory. PACAP can alter synaptic plasticity and has direct actions on neurons in the amygdala and hippocampus that could contribute to its acute and persistent effects on the consolidation and expression of conditioned fear. We recently demonstrated that intracerebroventricular (ICV) infusion of PACAP prior to fear conditioning (FC) results in initial amnestic-like effects followed by hyper-expression of conditioned freezing with repeated testing, and analyses of immediate-early gene c-Fos expression suggested that the central nucleus of the amygdala (CeA), but not the lateral/basolateral amygdala (LA/BLA) or hippocampus, are involved in these PACAP effects. Here, we extend that work by examining the expression of the synaptic plasticity marker activity-regulated cytoskeleton-associated protein (Arc/Arg 3.1) after PACAP administration and FC. Male Sprague-Dawley rats were implanted with cannula for ICV infusion of PACAP-38 (1.5 µg) or vehicle followed by FC and tests for conditioned freezing. One hour after FC, Arc protein expression was significantly elevated in the CeA and bed nucleus of the stria terminalis (BNST), interconnected structures that are key elements of the extended amygdala, in rats that received the combination of PACAP + FC. In contrast, Arc expression within the subdivisions of the hippocampus, or the LA/BLA, were unchanged. A subpopulation of Arc-positive cells in both the CeA and BNST also express PKCdelta, an intracellular marker that has been used to identify microcircuits that gate conditioned fear in the CeA. Consistent with our previous findings, on the following day conditioned freezing behavior was reduced in rats that had been given the combination of PACAP + FC-an amnestic-like effect-and Arc expression levels had returned to baseline. Given the established role of Arc in modifying synaptic plasticity and memory formation, our findings suggest that PACAP-induced overexpression of Arc following fear conditioning may disrupt neuroplastic changes within populations of CeA and BNST neurons normally responsible for encoding fear-related cues that, in this case, results in altered fear memory consolidation. Hence, PACAP systems may represent an axis on which stress and experience-driven neurotransmission converge to alter emotional memory, and mediate pathologies that are characteristic of psychiatric illnesses such as post-traumatic stress disorder.


Assuntos
Tonsila do Cerebelo/metabolismo , Proteínas do Citoesqueleto/metabolismo , Medo/fisiologia , Proteínas do Tecido Nervoso/metabolismo , Neurônios/metabolismo , Polipeptídeo Hipofisário Ativador de Adenilato Ciclase/metabolismo , Núcleos Septais/metabolismo , Tonsila do Cerebelo/efeitos dos fármacos , Animais , Condicionamento Clássico , Masculino , Plasticidade Neuronal , Neurônios/efeitos dos fármacos , Polipeptídeo Hipofisário Ativador de Adenilato Ciclase/administração & dosagem , Ratos Sprague-Dawley , Núcleos Septais/efeitos dos fármacos
16.
Curr Pharm Des ; 24(33): 3892-3904, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30417775

RESUMO

Pituitary adenylate cyclase activating polypeptide (PACAP) is a neuropeptide with diverse actions, including strong neurotrophic and neuroprotective effects. The aim of our present review is to provide a summary of the different approaches how in vivo neuroprotective effects can be achieved, emphasizing the potential translational values for future therapeutic applications. In the central nervous system, PACAP has been shown to have in vivo protective effects in models of cerebral ischemia, Parkinson's and Alzheimer's disease, Huntington chorea, traumatic brain and spinal cord injury and different retinal pathologies. PACAP passes through the bloodbrain barrier and therefore, systemic administration can affect the nervous system and lead to neuroprotective effects. This review summarizes results obtained in neuronal injury studies via local, such as intracerebral, intrathecal, intracerebroventricular, intravitreal and systemic treatments, such as intravenous, intraperitoneal and subcutaneous administration of PACAP. A few other options are summarized, like intranasal and eye drops treatments, as well as difficulties and side effects of different treatments are also discussed.


Assuntos
Neuroproteção/efeitos dos fármacos , Fármacos Neuroprotetores/farmacologia , Polipeptídeo Hipofisário Ativador de Adenilato Ciclase/farmacologia , Doença de Alzheimer/tratamento farmacológico , Animais , Lesões Encefálicas Traumáticas/tratamento farmacológico , Isquemia Encefálica/tratamento farmacológico , Humanos , Doença de Huntington/tratamento farmacológico , Fármacos Neuroprotetores/administração & dosagem , Doença de Parkinson/tratamento farmacológico , Polipeptídeo Hipofisário Ativador de Adenilato Ciclase/administração & dosagem , Traumatismos da Medula Espinal/tratamento farmacológico
17.
Mol Neurobiol ; 55(11): 8263-8277, 2018 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-29526016

RESUMO

Deficits in hippocampal synaptic plasticity result in cognitive impairment in Huntington's disease (HD). Pituitary adenylate cyclase-activating polypeptide (PACAP) is a neuropeptide that exerts neuroprotective actions, mainly through the PAC1 receptor. However, the role of PACAP in cognition is poorly understood, and no data exists in the context of Huntington's disease (HD). Here, we investigated the ability of PACAP receptor stimulation to enhance memory development in HD. First, we observed a hippocampal decline of all three PACAP receptor expressions, i.e., PAC1, VPAC1, and VPAC2, in two different HD mouse models, R6/1 and HdhQ7/Q111, from the onset of cognitive dysfunction. In hippocampal post-mortem human samples, we found a specific decrease of PAC1, without changes in VPAC1 and VPAC2 receptors. To determine whether activation of PACAP receptors could contribute to improve memory performance, we conducted daily intranasal administration of PACAP38 to R6/1 mice at the onset of cognitive impairment for seven days. We found that PACAP treatment rescued PAC1 level in R6/1 mice, promoted expression of the hippocampal brain-derived neurotrophic factor, and reduced the formation of mutant huntingtin aggregates. Furthermore, PACAP administration counteracted R6/1 mice memory deficits as analyzed by the novel object recognition test and the T-maze spontaneous alternation task. Importantly, the effect of PACAP on cognitive performance was associated with an increase of VGlut-1 and PSD95 immunolabeling in hippocampus of R6/1 mice. Taken together, these results suggest that PACAP, acting through stimulation of PAC1 receptor, may have a therapeutic potential to counteract cognitive deficits induced in HD.


Assuntos
Hipocampo/fisiopatologia , Doença de Huntington/fisiopatologia , Memória/fisiologia , Plasticidade Neuronal/efeitos dos fármacos , Polipeptídeo Hipofisário Ativador de Adenilato Ciclase/farmacologia , Administração Intranasal , Animais , Fator Neurotrófico Derivado do Encéfalo/metabolismo , Transtornos Cognitivos/fisiopatologia , Modelos Animais de Doenças , Proteína 4 Homóloga a Disks-Large/metabolismo , Regulação da Expressão Gênica/efeitos dos fármacos , Hipocampo/patologia , Humanos , Proteína Huntingtina/metabolismo , Masculino , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Pessoa de Meia-Idade , Polipeptídeo Hipofisário Ativador de Adenilato Ciclase/administração & dosagem , Agregados Proteicos , Receptores de Polipeptídeo Hipofisário Ativador de Adenilato Ciclase/metabolismo , Proteína Vesicular 1 de Transporte de Glutamato/metabolismo
18.
Am J Physiol Heart Circ Physiol ; 314(3): H563-H572, 2018 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-29212793

RESUMO

Intermittent hypoxia causes a persistent increase in sympathetic activity that progresses to hypertension in chronic conditions such as obstructive sleep apnea. Pituitary adenylate cyclase-activating polypeptide (PACAP) is an excitatory neurotransmitter that causes long-lasting sympathetic excitation. We aimed to determine if intermittent activation of the rostral ventrolateral medulla (RVLM) causes PACAP-mediated elevation of sympathetic nerve activity, termed sympathetic long-term facilitation (sLTF). The role of PACAP in mediating sLTF in response to intermittent activation of the RVLM was investigated in urethane-anaesthetized and artificially ventilated rats ( n = 65, Sprague-Dawley). Bilateral RVLM microinjections of the PACAP type 1 receptor/vasoactive intestinal polypeptide receptor type 2 receptor antagonist PACAP-(6-38) [ n = 6, change (Δ): -16.4 ± 6.5%) or an ionotropic glutamate antagonist, kynurenate ( n = 6, Δ:-7.2 ± 2.3%), blocked the development of acute intermittent hypoxia-induced sLTF ( n = 6, Δ: 49.2 ± 14.2%). Intermittent RVLM microinjections of glutamate caused sLTF ( n = 5, Δ: 56.9 ± 14.7%) that was abolished by PACAP-(6-38) pretreatment ( n = 5, Δ:-1.2 ± 4.7%). Conversely, intermittent microinjections of PACAP in the RVLM did not elicit sLTF. Intermittent bilateral disinhibition of the RVLM by microinjection of γ-aminobutyric acid in the caudal ventrolateral medulla did not elicit sLTF. Direct activation of RVLM neurons is crucial for the development of sLTF. PACAP and glutamate act synergistically in the RVLM, with both being necessary for the sLTF response. We found that activation of glutamate but not PACAP receptors is necessary and sufficient to generate sLTF, even in the absence of intermittent hypoxia. Our results demonstrate that PACAP within the RVLM may contribute to the development of obstructive sleep apnea -induced hypertension. NEW & NOTEWORTHY Pharmacological blockade of either pituitary adenylate cyclase-activating polypeptide (PACAP) or ionotropic glutamate receptors in the rostral ventrolateral medulla prevents development of sympathetic long-term facilitation. PACAP receptor inhibition prevents the occurrence of hypoxia-induced peripheral chemoreflex sensitization. Thus, PACAP receptors may be a potential therapeutic target serving to reduce heightened sympathetic tone and hypersensitized cardiovascular reflexes.


Assuntos
Hipóxia/fisiopatologia , Ácido Cinurênico/administração & dosagem , Potenciação de Longa Duração/efeitos dos fármacos , Bulbo/efeitos dos fármacos , Fragmentos de Peptídeos/administração & dosagem , Polipeptídeo Hipofisário Ativador de Adenilato Ciclase/administração & dosagem , Apneia Obstrutiva do Sono/fisiopatologia , Sistema Nervoso Simpático/efeitos dos fármacos , Doença Aguda , Animais , Modelos Animais de Doenças , Ácido Glutâmico/administração & dosagem , Hipertensão/etiologia , Hipertensão/fisiopatologia , Hipóxia/complicações , Hipóxia/metabolismo , Masculino , Bulbo/metabolismo , Bulbo/fisiopatologia , Microinjeções , Ratos Sprague-Dawley , Receptores de Glutamato/efeitos dos fármacos , Receptores de Glutamato/metabolismo , Receptores de Polipeptídeo Hipofisário Ativador de Adenilato Ciclase/efeitos dos fármacos , Receptores de Polipeptídeo Hipofisário Ativador de Adenilato Ciclase/metabolismo , Apneia Obstrutiva do Sono/complicações , Apneia Obstrutiva do Sono/metabolismo , Sistema Nervoso Simpático/metabolismo , Sistema Nervoso Simpático/fisiopatologia , Transmissão Sináptica/efeitos dos fármacos , Fatores de Tempo , Ácido gama-Aminobutírico/administração & dosagem
19.
Peptides ; 97: 64-69, 2017 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-28964802

RESUMO

Diabetic retinopathy (DR) is a microvascular complication of diabetes. Hyperglycemic/hypoxic microenvironment concurs to aberrant angiogenesis characterizing the pathology and activates many downstream target genes including inflammatory cytokines and vasoactive peptides, such as interleukin-1ß (IL-1ß) and vascular endothelial growth factor (VEGF). It has been largely demonstrated that pituitary adenylate cyclase-activating peptide (PACAP) plays a protective effect in DR. In the present study, we investigated the role of PACAP to protect retinal tissue through IL-1ß and VEGF expression. Diabetes was induced in rats by streptozotocin (STZ) injection, and one week later a single intravitreal injection of 100µM PACAP was administrated. Analyses of IL-1ß and VEGF levels were performed three weeks after diabetes induction. The results demonstrated that a single intraocular administration of PACAP significantly reduced the expression of IL-1ß in diabetic animals. Moreover, it affects VEGF and its receptors (VEGFRs) levels and interferes with their retinal layers distribution as showed by confocal microscopy analysis. In particular, PACAP treatment downregulates VEGF and VEGFRs that are increasingly expressed in STZ-treated animals as compared to controls. These results indicate that PACAP plays an important role to attenuate the early phase of DR.


Assuntos
Retinopatia Diabética/prevenção & controle , Interleucina-1beta/metabolismo , Polipeptídeo Hipofisário Ativador de Adenilato Ciclase/administração & dosagem , Fator A de Crescimento do Endotélio Vascular/metabolismo , Animais , Injeções Intravítreas , Masculino , Ratos , Ratos Sprague-Dawley , Receptores de Fatores de Crescimento do Endotélio Vascular/metabolismo , Retina/metabolismo , Estreptozocina/farmacologia , Fatores de Tempo
20.
Peptides ; 95: 25-32, 2017 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-28720396

RESUMO

Mitoxantrone (MXT) is an androstenedione that is used to treat cancers and progressive forms of multiple sclerosis; however, its use is limited by its cardiotoxicity. Pituitary adenylate cyclase activating polypeptide (PACAP) is a member of the secretin/growth hormone-releasing hormone/vasoactive intestinal peptide family and has many functions, including cytoprotection and immunosuppression. We tested the hypothesis that PACAP can protect against MXT-induced cardiotoxicity in mice. Female BALB/c mice were treated once weekly for 4 weeks with saline (n=14) or MXT (3mg/kg, i.p.; n=14). Half of the mice in each group received PACAP (10µg, i.p.) 1h before and 24 and 48h after MXT, while the remaining mice received injections of saline on the same schedule. Echocardiography was used to assess cardiac structure and function. In mice treated with MXT and saline, body weight was significantly reduced after the third dose of MXT. PACAP significantly attenuated the reduction in body weight; however, the weights did not return to control level. Compared to controls, MXT-treated mice had significantly increased left ventricular (LV) diameter and LV volume and decreased LV posterior wall thickness. Fractional shortening (FS) and ejection fraction (EF) were also significantly decreased. Treatment with PACAP prevented MXT-induced LV dilation and significantly attenuated the reductions in FS and EF, although FS and EF did not return to control level. PACAP38 did not prevent MXT-induced decreases in LV posterior wall thickness. MXT dose-dependently decreased the viability of cultured U937 (human leukemia) cells; PACAP did not protect cultured U937 cells from MXT-mediated cell death. In conclusion, PACAP can attenuate MXT-mediated LV dilation and dysfunction in mice.


Assuntos
Traumatismos Cardíacos/tratamento farmacológico , Mitoxantrona/efeitos adversos , Polipeptídeo Hipofisário Ativador de Adenilato Ciclase/administração & dosagem , Disfunção Ventricular Esquerda/tratamento farmacológico , Animais , Cardiotoxicidade/tratamento farmacológico , Cardiotoxicidade/patologia , Linhagem Celular Tumoral , Modelos Animais de Doenças , Traumatismos Cardíacos/induzido quimicamente , Traumatismos Cardíacos/patologia , Humanos , Camundongos , Mitoxantrona/uso terapêutico , Neoplasias/complicações , Neoplasias/tratamento farmacológico , Neoplasias/patologia , Substâncias Protetoras/administração & dosagem , Disfunção Ventricular Esquerda/induzido quimicamente , Disfunção Ventricular Esquerda/patologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA