Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 168
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
Neurobiol Dis ; 147: 105157, 2021 01.
Artigo em Inglês | MEDLINE | ID: mdl-33129939

RESUMO

Inhibiting the activity of fatty-acid amide hydrolase (FAAH), the enzyme that deactivates the endocannabinoid anandamide, enhances anandamide-mediated signaling and holds promise as a molecular target for the treatment of human pathologies such as anxiety and pain. We have previously shown that the peripherally restricted FAAH inhibitor, URB937, prevents nitroglycerin-induced hyperalgesia - an animal model of migraine - and attenuates the activation of brain areas that are relevant for migraine pain, e.g. trigeminal nucleus caudalis and locus coeruleus. The current study is aimed at profiling the behavioral and biochemical effects of URB937 in animal models of acute and chronic migraine. We evaluated the effects of URB937 in two rat models that capture aspects of acute and chronic migraine, and are based on single or repeated administration of the vasodilating drug, nitroglycerin (NTG). In addition to nocifensive behavior, in trigeminal ganglia and medulla, we measured mRNA levels of neuropeptides and pro-inflammatory cytokines along with tissue levels of anandamide and palmitoylethanolamide (PEA), an endogenous agonist of peroxisome proliferator-activated receptor type-a (PPAR-a), which is also a FAAH substrate. In the acute migraine model, we also investigated the effect of subtype-selective antagonist for cannabinoid receptors 1 and 2 (AM251 and AM630, respectively) on nocifensive behavior and on levels of neuropeptides and pro-inflammatory cytokines. In the acute migraine paradigm, URB937 significantly reduced hyperalgesia in the orofacial formalin test when administered either before or after NTG. This effect was accompanied by an increase in anandamide and PEA levels in target neural tissue, depended upon CB1 receptor activation, and was associated with a decrease in calcitonin gene-related peptide (CGRP), substance P and cytokines TNF-alpha and IL-6 mRNA. Similar effects were observed in the chronic migraine paradigm, where URB937 counteracted NTG-induced trigeminal hyperalgesia and prevented the increase in neuropeptide and cytokine transcription. The results show that peripheral FAAH inhibition by URB937 effectively reduces both acute and chronic NTG-induced trigeminal hyperalgesia, likely via augmented anandamide-mediated CB1 receptor activation. These effects are associated with inhibition of neuropeptidergic and inflammatory pathways.


Assuntos
Amidoidrolases/antagonistas & inibidores , Canabinoides/farmacologia , Transtornos de Enxaqueca , Animais , Comportamento Animal/efeitos dos fármacos , Citocinas/efeitos dos fármacos , Citocinas/metabolismo , Modelos Animais de Doenças , Masculino , Bulbo/efeitos dos fármacos , Bulbo/metabolismo , Neuropeptídeos/efeitos dos fármacos , Neuropeptídeos/metabolismo , Ratos , Ratos Sprague-Dawley , Gânglio Trigeminal/efeitos dos fármacos , Gânglio Trigeminal/metabolismo
2.
Arch Insect Biochem Physiol ; 107(1): e21767, 2021 May.
Artigo em Inglês | MEDLINE | ID: mdl-33835527

RESUMO

Mythimna separata Walker (Lepidoptera: Noctuidae) is one of the major pests that can cause severe damage to grain crops. The development of low-toxicity and high-performance botanical insecticides is becoming the focus of new pesticide research to control M. separata. Tutin, a sesquiterpene lactone compound obtained from Coriaria sinica Maxim, a native Chinese poisonous plant, has antifeedant, absorption, and stomach poisoning against a variety of pests. To understand the toxic effect of tutin on M. separata larvae, we set out to determine their antifeedant, mortality, paralysis, weight change, and to examine the spreading of M. separata hemocytes under different concentrations of tutin treatment. Tissue distribution of the immune-associated gene growth-blocking peptide (GBP) and neuroglian peptide (Nrg) was detected by reverse transcription polymerase chain reaction (PCR). Furthermore, real-time quantitative PCR was carried out to determine the expression profiles of GBP and Nrg after different concentrations of tutin stimulation. Our results revealed that tutin exhibited significant antifeedant and insecticidal activities, paralysis, weight loss to M. separata. Besides, tutin significantly influenced on the morphology of hemocytes and enhanced the expression of GBP and Nrg in M. separata.


Assuntos
Inseticidas/farmacologia , Mariposas/efeitos dos fármacos , Picrotoxina/análogos & derivados , Sesquiterpenos/farmacologia , Animais , Citocinas/efeitos dos fármacos , Citocinas/metabolismo , Hemócitos/efeitos dos fármacos , Proteínas de Insetos/efeitos dos fármacos , Proteínas de Insetos/metabolismo , Larva/efeitos dos fármacos , Larva/crescimento & desenvolvimento , Mariposas/crescimento & desenvolvimento , Neuropeptídeos/efeitos dos fármacos , Controle de Pragas , Picrotoxina/farmacologia
3.
Front Neuroendocrinol ; 55: 100799, 2019 10.
Artigo em Inglês | MEDLINE | ID: mdl-31614151

RESUMO

Hormonal contraceptives are frequently prescribed drugs among women, mainly for their reversible contraceptive purposes but also for beneficial effects in some gynecological pathologies. Despite extensive studies aimed at elucidating the physical effects of hormonal contraceptives and ameliorating some unwanted outcomes, little is known yet about the effects of these drugs on brain function and related behavior, which are known to be modulated by endogenous steroid hormones. We describe the current literature on preclinical studies in animals undertaken to investigate effects of hormonal contraceptives on brain function and behavior. These studies suggest that hormonal contraceptives influence neurohormones, neurotransmitters, neuropeptides, and emotional, cognitive, social and sexual behaviors. Animals allow examination of the basic biological mechanisms of these drugs, devoid of the psychological aspect often associated to hormonal contraceptives' use in women. Understanding the neurobiological effects of these drugs may improve women's health and may help women making informed choices on hormonal contraception.


Assuntos
Ansiedade , Comportamento Animal/efeitos dos fármacos , Encéfalo/efeitos dos fármacos , Contraceptivos Hormonais/farmacologia , Depressão , Aprendizagem/efeitos dos fármacos , Neuropeptídeos/efeitos dos fármacos , Neuroesteroides , Pregnanolona/farmacologia , Comportamento Sexual/efeitos dos fármacos , Comportamento Social , Estresse Psicológico , Transmissão Sináptica/efeitos dos fármacos , Animais , Ansiedade/induzido quimicamente , Ansiedade/metabolismo , Ansiedade/fisiopatologia , Depressão/induzido quimicamente , Depressão/metabolismo , Depressão/fisiopatologia , Feminino , Humanos , Estresse Psicológico/induzido quimicamente , Estresse Psicológico/tratamento farmacológico , Estresse Psicológico/metabolismo
4.
BMC Neurosci ; 21(1): 32, 2020 07 22.
Artigo em Inglês | MEDLINE | ID: mdl-32698770

RESUMO

BACKGROUND: Recent research has revealed that the community of microorganisms inhabiting the gut affects brain development, function and behaviour. In particular, disruption of the gut microbiome during critical developmental windows can have lasting effects on host physiology. Both antibiotic exposure and germ-free conditions impact the central nervous system and can alter multiple aspects of behaviour. Social impairments are typically displayed by antibiotic-treated and germ-free animals, yet there is a lack of understanding of the underlying neurobiological changes. Since the µ-opioid, oxytocin and vasopressin systems are key modulators of mammalian social behaviour, here we investigate the effect of experimentally manipulating the gut microbiome on the expression of these pathways. RESULTS: We show that social neuropeptide signalling is disrupted in germ-free and antibiotic-treated mice, which may contribute to the behavioural deficits observed in these animal models. The most notable finding is the reduction in neuroreceptor gene expression in the frontal cortex of mice administered an antibiotic cocktail post-weaning. Additionally, the changes observed in germ-free mice were generally in the opposite direction to the antibiotic-treated mice. CONCLUSIONS: Antibiotic treatment when young can impact brain signalling pathways underpinning social behaviour and pain regulation. Since antibiotic administration is common in childhood and adolescence, our findings highlight the potential adverse effects that antibiotic exposure during these key neurodevelopmental periods may have on the human brain, including the possible increased risk of neuropsychiatric conditions later in life. In addition, since antibiotics are often considered a more amenable alternative to germ-free conditions, our contrasting results for these two treatments suggest that they should be viewed as distinct models.


Assuntos
Antibacterianos/efeitos adversos , Encéfalo/efeitos dos fármacos , Neuropeptídeos/efeitos dos fármacos , Dor/tratamento farmacológico , Comportamento Social , Animais , Antibacterianos/metabolismo , Antibacterianos/farmacologia , Encéfalo/metabolismo , Microbioma Gastrointestinal/efeitos dos fármacos , Microbioma Gastrointestinal/fisiologia , Camundongos , Neuropeptídeos/metabolismo , Vasopressinas/metabolismo , Vasopressinas/farmacologia
5.
Horm Behav ; 125: 104825, 2020 09.
Artigo em Inglês | MEDLINE | ID: mdl-32771417

RESUMO

The endocannabinoid system (ECs) is a well known contributor to the hedonic regulation of food intake (FI) in mammals whereas in fish, the knowledge regarding hedonic mechanisms that control FI is limited. Previous studies reported the involvement of ECs in FI regulation in fish since anandamide (AEA) treatment induced enhanced FI and changes of mRNA abundance of appetite-related neuropeptides through cannabinoid receptor 1 (cnr1). However, no previous studies in fish evaluated the impact of palatable food like high-fat diets (HFD) on mechanisms involved in hedonic regulation of FI including the possible involvement of ECs. Therefore, we aimed to evaluate the effect of feeding a HFD on the response of ECs in rainbow trout (Oncorhynchus mykiss). First, we demonstrated a higher intake over 4 days of HFD compared with a control diet (CD). Then, we evaluated the postprandial response (1, 3 and 6 h) of components of the ECs in plasma, hypothalamus, and telencephalon after feeding fish with CD and HFD. The results obtained indicate that the increased FI of HFD occurred along with increased levels of 2-arachidonoylglycerol (2-AG) and AEA in plasma and in brain areas like hypothalamus and telencephalon putatively involved in hedonic regulation of FI in fish. Decreased mRNA abundance of EC receptors like cnr1, gpr55 and trpv1 suggest a feed-back counter-regulatory mechanism in response to the increased levels of EC. Furthermore, the results also suggest that neural activity players associated to FI regulation in mammals as cFOS, γ-Amino butyric acid (GABA) and brain derived neurotrophic factor (BDNF)/neurotrophic receptor tyrosine kinase (NTRK) systems could be involved in the hedonic eating response to a palatable diet in fish.


Assuntos
Dieta Hiperlipídica , Endocanabinoides/metabolismo , Oncorhynchus mykiss/metabolismo , Receptor CB1 de Canabinoide/metabolismo , Animais , Apetite/efeitos dos fármacos , Apetite/genética , Regulação do Apetite/efeitos dos fármacos , Regulação do Apetite/fisiologia , Encéfalo/efeitos dos fármacos , Encéfalo/metabolismo , Gorduras na Dieta/farmacologia , Ingestão de Alimentos/efeitos dos fármacos , Ingestão de Alimentos/genética , Metabolismo Energético/efeitos dos fármacos , Metabolismo Energético/genética , Regulação da Expressão Gênica/efeitos dos fármacos , Hipotálamo/efeitos dos fármacos , Hipotálamo/metabolismo , Neuropeptídeos/efeitos dos fármacos , Neuropeptídeos/genética , Neuropeptídeos/metabolismo , Oncorhynchus mykiss/fisiologia , Receptor CB1 de Canabinoide/genética , Telencéfalo/efeitos dos fármacos , Telencéfalo/metabolismo
6.
Headache ; 60(7): 1259-1272, 2020 07.
Artigo em Inglês | MEDLINE | ID: mdl-32602955

RESUMO

OBJECTIVE: To review the literature on the mechanism of action of onabotulinumtoxinA in chronic migraine. BACKGROUND: OnabotulinumtoxinA is a chronic migraine preventive treatment that significantly reduces headache frequency. The traditional mechanism described for onabotulinumtoxinA - reducing muscle contractions - is insufficient to explain its efficacy in migraine, which is primarily a sensory neurological disease. METHODS: A narrative literature review on the mechanism of action of onabotulinumtoxinA in chronic migraine. RESULTS: Following injection into tissues, onabotulinumtoxinA inhibits soluble N-ethylmaleimide-sensitive fusion attachment protein receptor (SNARE)-mediated vesicle trafficking by cleaving one of its essential proteins, soluble N-ethylmaleimide-sensitive fusion attachment protein (SNAP-25), which occurs in both motor and sensory nerves. OnabotulinumtoxinA inhibits regulated exocytosis of motor and sensory neurochemicals and proteins, as well as membrane insertion of peripheral receptors that convey pain from the periphery to the brain, because both processes are SNARE dependent. OnabotulinumtoxinA can decrease exocytosis of pro-inflammatory and excitatory neurotransmitters and neuropeptides such as substance P, calcitonin gene-related peptide, and glutamate from primary afferent fibers that transmit nociceptive pain and participate in the development of peripheral and central sensitization. OnabotulinumtoxinA also decreases the insertion of pain-sensitive ion channels such as transient receptor potential cation channel subfamily V member 1 (TRPV1) into the membranes of nociceptive neurons; this is likely enhanced in the sensitized neuron. For chronic migraine prevention, onabotulinumtoxinA is injected into 31-39 sites in 7 muscles of the head and neck. Sensory nerve endings of neurons whose cell bodies are located in trigeminal and cervical ganglia are distributed throughout the injected muscles, and are overactive in people with migraine. Through inhibition of these sensory nerve endings, onabotulinumtoxinA reduces the number of pain signals that reach the brain and consequently prevents activation and sensitization of central neurons postulated to be involved in migraine chronification. CONCLUSION: OnabotulinumtoxinA likely acts via sensory mechanisms to treat chronic migraine.


Assuntos
Toxinas Botulínicas Tipo A/farmacologia , Ácido Glutâmico/efeitos dos fármacos , Transtornos de Enxaqueca/prevenção & controle , Fármacos Neuromusculares/farmacologia , Neuropeptídeos/efeitos dos fármacos , Neurotransmissores/farmacologia , Proteínas SNARE/efeitos dos fármacos , Doença Crônica , Humanos
7.
Int J Neuropsychopharmacol ; 21(2): 175-186, 2018 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-29020410

RESUMO

Background: Previous studies have shown that a low dose of scopolamine produces rapid-acting antidepressant-like actions in rodents. Understanding the mechanisms underlying this effect and the dose-dependent variations of drug responses remains an important task. L-type voltage-dependent calcium channels were found to mediate rapid-acting antidepressant effects of certain medications (e.g., ketamine). Therefore, it is of great interest to determine the involvement of L-type voltage-dependent calcium channels in the action of scopolamine. Methods: Herein, we investigated the mechanisms underlying behavioral responses to various doses of scopolamine in mice to clarify the involvement of L-type voltage-dependent calcium channels in its modes of action. Open field test, novel object recognition test, and forced swimming test were performed on mice administered varied doses of scopolamine (0.025, 0.05, 0.1, 1, and 3 mg/kg, i.p.) alone or combined with L-type voltage-dependent calcium channel blocker verapamil (5 mg/kg, i.p.). Then, the changes in brain-derived neurotrophic factor and neuropeptide VGF (nonacronymic) levels in the hippocampus and prefrontal cortex of these mice were analyzed. Results: Low doses of scopolamine (0.025 and 0.05 mg/kg) produced significant antidepressant-like effects in the forced swimming test, while higher doses (1 and 3 mg/kg) resulted in significant memory deficits and depressive-like behaviors. Moreover, the behavioral changes in responses to various doses may be related to the upregulation (0.025 and 0.05 mg/kg) and downregulation (1 and 3 mg/kg) of brain-derived neurotrophic factor and VGF in the hippocampus and prefrontal cortex in mice. We further found that the rapid-acting antidepressant-like effects and the upregulation on brain-derived neurotrophic factor and VGF produced by a low dose of scopolamine (0.025 mg/kg) were completely blocked by verapamil. Conclusions: These results indicate that L-type voltage-dependent calcium channels are likely involved in the behavioral changes in response to various doses of scopolamine through the regulation of brain-derived neurotrophic factor and VGF levels.


Assuntos
Antidepressivos/farmacologia , Comportamento Animal/efeitos dos fármacos , Fator Neurotrófico Derivado do Encéfalo/efeitos dos fármacos , Bloqueadores dos Canais de Cálcio/farmacologia , Canais de Cálcio Tipo L/efeitos dos fármacos , Disfunção Cognitiva/induzido quimicamente , Depressão/induzido quimicamente , Hipocampo/efeitos dos fármacos , Neuropeptídeos/efeitos dos fármacos , Córtex Pré-Frontal/efeitos dos fármacos , Escopolamina/farmacologia , Verapamil/farmacologia , Animais , Antidepressivos/administração & dosagem , Bloqueadores dos Canais de Cálcio/administração & dosagem , Modelos Animais de Doenças , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Fatores de Crescimento Neural , Escopolamina/administração & dosagem , Verapamil/administração & dosagem
8.
Genet Mol Res ; 15(1): 15017362, 2016 Feb 26.
Artigo em Inglês | MEDLINE | ID: mdl-26985919

RESUMO

The hypothalamus is an important component of the nervous system, and neuropeptide Y (NPY), proopiomelanocortin (POMC), and neuromedin U (NMU) are key players in physiological regulation. Puerarin is important for nerve regulation. We investigated the effect of puerarin on the expression of NMU, NPY, and POMC genes in the hypothalamus. The results showed that the puerarin low-dose group and the other groups were significantly different (P < 0.05). However, there was no significant difference in NMU, POMC, and NPY among the groups.


Assuntos
Hipotálamo/metabolismo , Isoflavonas/farmacologia , Neuropeptídeo Y/genética , Neuropeptídeos/genética , Pró-Opiomelanocortina/genética , Animais , Regulação da Expressão Gênica , Hipotálamo/efeitos dos fármacos , Neuropeptídeo Y/efeitos dos fármacos , Neuropeptídeos/efeitos dos fármacos , Pró-Opiomelanocortina/efeitos dos fármacos , Ratos
10.
Neuro Endocrinol Lett ; 36(8): 767-70, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26921577

RESUMO

OBJECTIVE: The neonatal and/or prepubertal androgen milieu affects sexual maturation. In rodents, neonatal chronic testosterone treatment, which is used as a model of polycystic ovary syndrome (PCOS), results in the onset of vaginal opening occurring earlier in the pubertal period. DESIGN: In the present study, the changes in hypothalamic Kiss1 (a gonadotropin-releasing hormone (GnRH)-stimulating factor) and RF-amide related peptide (RFRP; a GnRH inhibitory factor) mRNA expression induced by testosterone treatment were examined in order to clarify whether these factors are involved in the testosterone-induced acceleration of sexual maturation. RESULTS: The onset of vaginal opening occurred earlier and uterine weight was increased in female rats subjected to chronic (from postnatal day 23 to day 31) testosterone treatment. Contrary to our expectations, the rats' hypothalamic Kiss1 and Kiss1 receptor mRNA levels were not changed, and their serum luteinizing hormone (LH) levels were decreased. Although hypothalamic RFRP mRNA expression was decreased in the testosterone-treated rats, this change was not reflected in their serum LH levels. CONCLUSIONS: These results indicate that the advancement of sexual maturation observed in chronic testosterone-treated rats might be caused by a peripheral, rather than a central, mechanism.


Assuntos
Androgênios/farmacologia , Hipotálamo/efeitos dos fármacos , Kisspeptinas/efeitos dos fármacos , Neuropeptídeos/efeitos dos fármacos , RNA Mensageiro/efeitos dos fármacos , Maturidade Sexual/efeitos dos fármacos , Testosterona/farmacologia , Vagina/efeitos dos fármacos , Animais , Feminino , Hormônio Liberador de Gonadotropina/efeitos dos fármacos , Hormônio Liberador de Gonadotropina/genética , Hormônio Liberador de Gonadotropina/metabolismo , Hipotálamo/metabolismo , Kisspeptinas/genética , Kisspeptinas/metabolismo , Neuropeptídeos/genética , Neuropeptídeos/metabolismo , RNA Mensageiro/metabolismo , Ratos , Ratos Sprague-Dawley , Receptores Acoplados a Proteínas G/efeitos dos fármacos , Receptores Acoplados a Proteínas G/genética , Receptores Acoplados a Proteínas G/metabolismo , Receptores de Kisspeptina-1 , Receptores de Neuropeptídeos/efeitos dos fármacos , Receptores de Neuropeptídeos/genética , Receptores de Neuropeptídeos/metabolismo
11.
Artigo em Zh | MEDLINE | ID: mdl-25916449

RESUMO

OBJECTIVE: To investigate the effects of polygala on leaning and memory and the expression of Microtubule associated protein on manganese poisoned mice. METHODS: 60 female Kunming mice were randomly and equally divided into 5 group. They are normal control group (CG), manganese poisoned group (MG), manganese poisoned with polygala high dose group (MHG), manganese poisoned with polygala middle dose group (MMG), manganese poisoned with polygala low dose group (MLG). The model of manganese poisoned mice was prepared of the way of intraperitoneal injection of manganese chloride (MnCl2 15 mg/kg), the spatial learning and memory ability was tested by Morris water maze, the Doublecortin (DCX) was tested by the way of immunofluorescent staining in the SVZ and SGZ. RESULT: In the navigation test, compared with MG, the escape latency of MHG, MMG and MLG were significantly decreased (P < 0.05), in space exploration experiments, MHG, MMG, MLG compared with MG, the number increased significantly across platforms (P < 0.05). compared with MG, the DCX expression of MHG, MMG and MLG were significantly increased (P < 0.05). CONCLUTION: The leaning and memory ability of manganese poisoned mice can be improved by the polygala, and the mechanism may be related to promote the expression of DCX and neurogenesis in the brain.


Assuntos
Intoxicação por Manganês/tratamento farmacológico , Proteínas Associadas aos Microtúbulos/efeitos dos fármacos , Neurogênese/efeitos dos fármacos , Neuropeptídeos/efeitos dos fármacos , Extratos Vegetais/farmacologia , Polygala/química , Animais , Proteínas do Domínio Duplacortina , Proteína Duplacortina , Feminino , Aprendizagem em Labirinto/efeitos dos fármacos , Memória/efeitos dos fármacos , Camundongos
12.
J Physiol ; 592(5): 915-26, 2014 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-24297853

RESUMO

The role of the small GTP-binding protein Rac1 in smooth muscle contraction was examined using small molecule inhibitors (EHT1864, NSC23766) and a novel smooth muscle-specific, conditional, Rac1 knockout mouse strain. EHT1864, which affects nucleotide binding and inhibits Rac1 activity, concentration-dependently inhibited the contractile responses induced by several different modes of activation (high-K+, phenylephrine, carbachol and protein kinase C activation by phorbol-12,13-dibutyrate) in several different visceral (urinary bladder, ileum) and vascular (mesenteric artery, saphenous artery, aorta) smooth muscle tissues. This contractile inhibition was associated with inhibition of the Ca2+ transient. Knockout of Rac1 (with a 50% loss of Rac1 protein) lowered active stress in the urinary bladder and the saphenous artery consistent with a role of Rac1 in facilitating smooth muscle contraction. NSC23766, which blocks interaction between Rac1 and some guanine nucleotide exchange factors, specifically inhibited the α1 receptor responses (phenylephrine) in vascular tissues and potentiated prostaglandin F2α and thromboxane (U46619) receptor responses. The latter potentiating effect occurred at lowered intracellular [Ca2+]. These results show that Rac1 activity is required for active contraction in smooth muscle, probably via enabling an adequate Ca2+ transient. At the same time, specific agonists recruit Rac1 signalling via upstream modulators, resulting in either a potentiation of contraction via Ca2+ mobilization (α1 receptor stimulation) or an attenuated contraction via inhibition of Ca2+ sensitization (prostaglandin and thromboxane receptors).


Assuntos
Sinalização do Cálcio/fisiologia , Contração Isométrica/fisiologia , Músculo Liso/fisiologia , Neuropeptídeos/metabolismo , Transdução de Sinais/fisiologia , Proteínas rac1 de Ligação ao GTP/metabolismo , Animais , Feminino , Técnicas In Vitro , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Neuropeptídeos/efeitos dos fármacos , Neuropeptídeos/genética , Proteínas rac1 de Ligação ao GTP/efeitos dos fármacos , Proteínas rac1 de Ligação ao GTP/genética
13.
J Am Soc Nephrol ; 24(7): 1114-26, 2013 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-23661807

RESUMO

Ras homolog enriched in brain (Rheb) is a small GTPase that regulates cell growth, differentiation, and survival by upregulating mammalian target of rapamycin complex 1 (mTORC1) signaling. The role of Rheb/mTORC1 signaling in the activation of kidney fibroblasts and the development of kidney fibrosis remains largely unknown. In this study, we found that Rheb/mTORC1 signaling was activated in interstitial myofibroblasts from fibrotic kidneys. Treatment of rat kidney interstitial fibroblasts (NRK-49F cell line) with TGFß1 also activated Rheb/mTORC1 signaling. Blocking Rheb/mTORC1 signaling with rapamycin or Rheb small interfering RNA abolished TGFß1-induced fibroblast activation. In a transgenic mouse, ectopic expression of Rheb activated kidney fibroblasts. These Rheb transgenic mice exhibited increased activation of mTORC1 signaling in both kidney tubular and interstitial cells as well as progressive interstitial renal fibrosis; rapamycin inhibited these effects. Similarly, mice with fibroblast-specific deletion of Tsc1, a negative regulator of Rheb, exhibited activated mTORC1 signaling in kidney interstitial fibroblasts and increased renal fibrosis, both of which rapamycin abolished. Taken together, these results suggest that Rheb/mTORC1 signaling promotes the activation of kidney fibroblasts and contributes to the development of interstitial fibrosis, possibly providing a therapeutic target for progressive renal disease.


Assuntos
Fibroblastos/metabolismo , Fibroblastos/patologia , Rim/metabolismo , Rim/patologia , Proteínas Monoméricas de Ligação ao GTP/metabolismo , Neuropeptídeos/metabolismo , Proteínas/metabolismo , Animais , Linhagem Celular , Modelos Animais de Doenças , Fibrose , Imuno-Histoquímica , Alvo Mecanístico do Complexo 1 de Rapamicina , Camundongos , Proteínas Monoméricas de Ligação ao GTP/efeitos dos fármacos , Complexos Multiproteicos , Neuropeptídeos/efeitos dos fármacos , Proteínas/efeitos dos fármacos , Proteína Enriquecida em Homólogo de Ras do Encéfalo , Ratos , Transdução de Sinais , Sirolimo/farmacologia , Serina-Treonina Quinases TOR , Fator de Crescimento Transformador beta1/farmacologia
14.
Eur Respir J ; 41(1): 165-76, 2013 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-22790920

RESUMO

The protective effects of prostacyclin and its stable analogue iloprost are mediated by elevation of intracellular cyclic AMP (cAMP) leading to enhancement of the peripheral actin cytoskeleton and cell-cell adhesive structures. This study tested the hypothesis that iloprost may exhibit protective effects against lung injury and endothelial barrier dysfunction induced by bacterial wall lipopolysaccharide (LPS). Endothelial barrier dysfunction was assessed by measurements of transendothelial permeability, morphologically and by analysis of LPS-activated inflammatory signalling. In vivo, C57BL/6J mice were challenged with LPS with or without iloprost or 8-bromoadenosine-3',5'-cyclic monophosphate (Br-cAMP) treatment. Lung injury was monitored by measurements of bronchoalveolar lavage protein content, cell count and Evans blue extravasation. Iloprost and Br-cAMP attenuated the disruption of the endothelial monolayer, and suppressed the activation of p38 mitogen-activated protein kinase (MAPK), the nuclear factor (NF)-κB pathway, Rho signalling, intercellular adhesion molecular (ICAM)-1 expression and neutrophil migration after LPS challenge. In vivo, iloprost was effective against LPS-induced protein and neutrophil accumulation in bronchoalveolar lavage fluid, and reduced myeloperoxidase activation, ICAM-1 expression and Evans blue extravasation in the lungs. Inhibition of Rac activity abolished the barrier-protective and anti-inflammatory effects of iloprost and Br-cAMP. Iloprost-induced elevation of intracellular cAMP triggers Rac signalling, which attenuates LPS-induced NF-κB and p38 MAPK inflammatory pathways and the Rho-dependent mechanism of endothelial permeability.


Assuntos
Iloprosta/uso terapêutico , Lesão Pulmonar/tratamento farmacológico , Pulmão/efeitos dos fármacos , Pulmão/fisiopatologia , Animais , Células Cultivadas , Endotélio/efeitos dos fármacos , Endotélio/fisiologia , Lipopolissacarídeos/administração & dosagem , Lesão Pulmonar/induzido quimicamente , Camundongos , Camundongos Endogâmicos C57BL , Neuropeptídeos/efeitos dos fármacos , Neuropeptídeos/fisiologia , Proteínas rac de Ligação ao GTP/efeitos dos fármacos , Proteínas rac de Ligação ao GTP/fisiologia , Proteínas rac1 de Ligação ao GTP
15.
J Surg Res ; 183(2): 798-807, 2013 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-23545410

RESUMO

Excessive neutrophil recruitment is a major feature in septic lung damage although the signaling mechanisms behind pulmonary infiltration of neutrophils in sepsis remain elusive. In the present study, we hypothesized that Rac1 might play an important role in pulmonary neutrophil accumulation and tissue injury in abdominal sepsis. Male C57BL/6 mice were treated with Rac1 inhibitor NSC23766 (5 mg/kg) before cecal ligation and puncture (CLP). Bronchoalveolar lavage fluid and lung tissue were collected for the quantification of neutrophil recruitment and edema and CXC chemokine formation. Blood was collected for the determination of Mac-1 on neutrophils and proinflammatory compounds in plasma. Gene expression of CXC chemokines and tumor necrosis factor alpha was determined by quantitative reverse transcription-polymerase chain reaction in alveolar macrophages. Rac1 activity was increased in lungs from septic animals, and NSC23766 significantly decreased pulmonary activity of Rac1 induced by CLP. Administration of NSC23766 markedly reduced CLP-triggered neutrophil infiltration, edema formation, and tissue damage in the lung. Inhibition of Rac1 decreased CLP-induced neutrophil expression of Mac-1 and pulmonary formation of CXC chemokines. Moreover, NSC23766 abolished the sepsis-evoked elevation of messenger RNA levels of CXC chemokines and tumor necrosis factor alpha in alveolar macrophages. Rac1 inhibition decreased the CLP-induced increase in plasma levels of high mobility group protein B1 and interleukin 6, indicating a role of Rac1 in systemic inflammation. In conclusion, our results demonstrate that Rac1 signaling plays a key role in regulating pulmonary infiltration of neutrophils and tissue injury via regulation of chemokine production in the lung and Mac-1 expression on neutrophils in abdominal sepsis. Thus, targeting Rac1 activity might be a useful strategy to protect the lung in abdominal sepsis.


Assuntos
Quimiocinas CXC/metabolismo , Antígeno de Macrófago 1/metabolismo , Neuropeptídeos/fisiologia , Pneumonia/microbiologia , Pneumonia/fisiopatologia , Sepse/complicações , Transdução de Sinais/fisiologia , Proteínas rac de Ligação ao GTP/fisiologia , Aminoquinolinas/farmacologia , Animais , Ceco/lesões , Movimento Celular/efeitos dos fármacos , Movimento Celular/fisiologia , Modelos Animais de Doenças , Proteína HMGB1/metabolismo , Técnicas In Vitro , Ligadura/efeitos adversos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Neuropeptídeos/antagonistas & inibidores , Neuropeptídeos/efeitos dos fármacos , Neutrófilos/efeitos dos fármacos , Neutrófilos/metabolismo , Neutrófilos/patologia , Pneumonia/patologia , Punções/efeitos adversos , Pirimidinas/farmacologia , Sepse/etiologia , Proteínas rac de Ligação ao GTP/antagonistas & inibidores , Proteínas rac de Ligação ao GTP/efeitos dos fármacos , Proteínas rac1 de Ligação ao GTP
16.
Int J Mol Sci ; 14(8): 16817-35, 2013 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-23955266

RESUMO

Trimethyltin (TMT) is an organotin compound exhibiting neurotoxicant effects selectively localized in the limbic system and especially marked in the hippocampus, in both experimental animal models and accidentally exposed humans. TMT administration causes selective neuronal death involving either the granular neurons of the dentate gyrus or the pyramidal cells of the Cornu Ammonis, with a different pattern of localization depending on the different species studied or the dosage schedule. TMT is broadly used to realize experimental models of hippocampal neurodegeneration associated with cognitive impairment and temporal lobe epilepsy, though the molecular mechanisms underlying the associated selective neuronal death are still not conclusively clarified. Experimental evidence indicates that TMT-induced neurodegeneration is a complex event involving different pathogenetic mechanisms, probably acting differently in animal and cell models, which include neuroinflammation, intracellular calcium overload, and oxidative stress. Microarray-based, genome-wide expression analysis has been used to investigate the molecular scenario occurring in the TMT-injured brain in different in vivo and in vitro models, producing an overwhelming amount of data. The aim of this review is to discuss and rationalize the state-of-the-art on TMT-associated genome wide expression profiles in order to identify comparable and reproducible data that may allow focusing on significantly involved pathways.


Assuntos
Perfilação da Expressão Gênica , Hipocampo/efeitos dos fármacos , Hipocampo/metabolismo , Doenças Neurodegenerativas/metabolismo , Compostos de Trimetilestanho/administração & dosagem , Animais , Linhagem Celular , Camundongos , Mitocôndrias/efeitos dos fármacos , Modelos Animais , Doenças Neurodegenerativas/induzido quimicamente , Doenças Neurodegenerativas/genética , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Neuropeptídeos/efeitos dos fármacos , Neurotoxinas/administração & dosagem , Estresse Oxidativo/efeitos dos fármacos , Ratos
17.
J Biol Chem ; 286(40): 34575-82, 2011 Oct 07.
Artigo em Inglês | MEDLINE | ID: mdl-21808066

RESUMO

Paclitaxel (Taxol) is one of the most effective treatment options for patients suffering from a variety of cancers. A major side effect seen in a high percentage of patients treated with paclitaxel is irreversible peripheral neuropathy. We previously reported that prolonged treatment with paclitaxel activates a calcium-dependent enzyme, calpain, which degrades neuronal calcium sensor 1 (NCS-1) and subsequent loss of intracellular calcium signaling. Because it appears that activation of calpain is an early step in this destructive cascade, we proposed that inhibition of calpain will protect against the unwanted side effects of paclitaxel treatment. First, NCS-1 levels and intracellular calcium signaling were found to be protected by the presence of lactacystin, a protesome inhibitor. To reinforce the role of calpain in this process, we showed that increased concentrations of calpastatin, a naturally occurring calpain inhibitor, were protective. Next, we tested two mutated versions of NCS-1 developed with point mutations at the P2 position of the calpain cleavage site of NCS-1 to decrease the likelihood of NCS-1 degradation. One mutant was cleaved more favorably by calpain compared with NCS-1 WT, whereas the other mutant was less favorably cleaved. Expression of either mutated version of NCS-1 in neuroblastoma cells protected intracellular calcium signals from paclitaxel-induced changes. These results support our hypothesis that it is possible to protect cells from paclitaxel-induced degradation of NCS-1 by inhibiting calpain activity.


Assuntos
Sinalização do Cálcio , Calpaína/metabolismo , Proteínas Sensoras de Cálcio Neuronal/metabolismo , Neuropeptídeos/metabolismo , Paclitaxel/farmacologia , Acetilcisteína/análogos & derivados , Acetilcisteína/farmacologia , Linhagem Celular Tumoral , Humanos , Mutação , Neuroblastoma/metabolismo , Proteínas Sensoras de Cálcio Neuronal/efeitos dos fármacos , Neurônios/patologia , Neuropeptídeos/efeitos dos fármacos , Mutação Puntual , Inibidores de Proteases/farmacologia , Isoformas de Proteínas , Transdução de Sinais , Termodinâmica
18.
Epilepsia ; 53 Suppl 9: 41-58, 2012 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-23216578

RESUMO

Synaptic transmission is the communication between a presynaptic and a postsynaptic neuron, and the subsequent processing of the signal. These processes are complex and highly regulated, reflecting their importance in normal brain functioning and homeostasis. Sustaining synaptic transmission depends on the continuing cycle of synaptic vesicle formation, release, and endocytosis, which requires proteins such as dynamin, syndapin, synapsin, and synaptic vesicle protein 2A. Synaptic transmission is regulated by diverse mechanisms, including presynaptic modulators of synaptic vesicle formation and release, postsynaptic receptors and signaling, and modulators of neurotransmission. Neurotransmitters released presynaptically can bind to their postsynaptic receptors, the inhibitory γ-aminobutyric acid (GABA)ergic receptors or the excitatory glutamate receptors. Once released, glutamate activates a variety of postsynaptic receptors including α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA), N-methyl-D-aspartate (NMDA), kainate, and metabotropic receptors. The activation of the receptors triggers downstream signaling cascades generating a vast array of effects, which can be modulated by a numerous auxiliary regulatory subunits. Moreover, different neuropeptides such as neuropeptide Y, brain-derived neurotrophic factor (BDNF), somatostatin, ghrelin, and galanin, act as regulators of diverse synaptic functions and along with the classic neurotransmitters. Abnormalities in the regulation of synaptic transmission play a critical role in the pathogenesis of numerous brain diseases, including epilepsy. This review focuses on the different mechanisms involved in the regulation of synaptic transmission, which may play a role in the pathogenesis of epilepsy: the presynaptic modulators of synaptic vesicle formation and release, postsynaptic receptors, and modulators of neurotransmission, including the mechanism by which drugs can modulate the frequency and severity of epileptic seizures.


Assuntos
Anticonvulsivantes/farmacologia , Epilepsia/genética , Epilepsia/fisiopatologia , Transmissão Sináptica , Animais , Anticonvulsivantes/uso terapêutico , Epilepsia/tratamento farmacológico , Humanos , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/metabolismo , Neuropeptídeos/efeitos dos fármacos , Neuropeptídeos/metabolismo , Receptores de Glutamato/efeitos dos fármacos , Receptores de Glutamato/genética , Receptores de Glutamato/metabolismo , Transmissão Sináptica/efeitos dos fármacos , Transmissão Sináptica/genética , Vesículas Sinápticas/efeitos dos fármacos , Vesículas Sinápticas/genética
19.
Fluids Barriers CNS ; 19(1): 7, 2022 Jan 21.
Artigo em Inglês | MEDLINE | ID: mdl-35062973

RESUMO

BACKGROUND: C-C chemokine receptor type 1 (CCR1) and its endogenous ligand, CCL5, participate in the pathogenesis of neuroinflammatory diseases. However, much remains unknown regarding CCL5/CCR1 signaling in blood-brain barrier (BBB) permeability after intracerebral hemorrhage (ICH). METHODS: A total of 250 CD1 male mice were used and ICH was induced via autologous whole blood injection. Either Met-RANTES, a selective CCR1 antagonist, or Met-RANTES combined with a Rac1 CRISPR activator was administered to the mice 1 h after ICH. Post-ICH assessments included neurobehavioral tests, brain water content, BBB integrity, hematoma volume, Western blot, and immunofluorescence staining. The CCR1 ligand, rCCL5, and SRC CRISPR knockout in naïve mice were used to further elucidate detrimental CCL5/CCR1/SRC signaling. RESULTS: Brain endogenous CCR1 and CCL5 were upregulated after ICH in mice with a peak at 24 h, and CCR1 was expressed in endothelial cells, astrocytes, and neurons. Met-R treatment reduced brain edema and neurobehavioral impairment, as well as preserved BBB integrity and tight junction protein expression in ICH mice. Met-R treatment decreased expression of p-SRC, Rac1, albumin, and MMP9, but increased claudin-5, occludin, and ZO-1 tight junction proteins after ICH. These effects were regressed using the Rac1 CRISPR activator. Administration of rCCL5 in naïve mice increased expression of p-SRC, Rac1, albumin, and MMP9, but decreased levels of claudin-5, occludin, and ZO-1 tight junction proteins. These effects in naïve mice were reversed with SRC CRISPR (KO). CONCLUSIONS: Our findings demonstrate that CCR5 inhibition by Met-R improves neurological deficits after ICH by preserving BBB integrity through inhibiting CCR1/SRC/Rac1 signaling pathway in mice. Thus, Met-R has therapeutic potential in the management of ICH patients.


Assuntos
Barreira Hematoencefálica/metabolismo , Antagonistas dos Receptores CCR5/farmacologia , Hemorragia Cerebral/tratamento farmacológico , Hemorragia Cerebral/metabolismo , Quimiocina CCL5/farmacologia , Neuropeptídeos/metabolismo , Receptores CCR1/metabolismo , Proteínas rac1 de Ligação ao GTP/metabolismo , Quinases da Família src/metabolismo , Animais , Barreira Hematoencefálica/efeitos dos fármacos , Quimiocina CCL5/administração & dosagem , Masculino , Camundongos , Neuropeptídeos/efeitos dos fármacos , Receptores CCR1/antagonistas & inibidores , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/fisiologia , Proteínas rac1 de Ligação ao GTP/efeitos dos fármacos , Quinases da Família src/efeitos dos fármacos
20.
FASEB J ; 24(8): 2670-9, 2010 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-20335227

RESUMO

The orexigenic effect of ghrelin is mediated by neuropeptide Y (NPY) and agouti-related protein (AgRP) in the hypothalamic arcuate nucleus (ARC). Recent evidence also indicates that ghrelin promotes feeding through a mechanism involving activation of hypothalamic AMP-activated protein kinase (AMPK) and inactivation of acetyl-CoA carboxylase and fatty acid synthase (FAS). This results in decreased hypothalamic levels of malonyl-CoA, increased carnitine palmitoyltransferase 1 (CPT1) activity, and mitochondrial production of reactive oxygen species. We evaluated whether these molecular events are part of a unique signaling cascade or whether they represent alternative pathways mediating the orexigenic effect of ghrelin. Moreover, we examined the gender dependency of these mechanisms, because recent evidence has proposed that ghrelin orexigenic effect is reduced in female rats. We studied in both genders the effect of ghrelin on the expression of AgRP and NPY, as well as their transcription factors: cAMP response-element binding protein (CREB and its phosphorylated form, pCREB), forkhead box O1 (FoxO1 and its phosphorylated form, pFoxO1), and brain-specific homeobox transcription factor (BSX). In addition, to establish a mechanistic link between ghrelin, fatty acid metabolism, and neuropeptides, we evaluated the effect of ghrelin after blockage of hypothalamic fatty acid beta oxidation, by using the CPT1 inhibitor etomoxir. Ghrelin-induced changes in the AMPK-CPT1 pathway are associated with increased levels of AgRP and NPY mRNA expression through modulation of BSX, pCREB, and FoxO1, as well as decreased expression of endoplasmic reticulum (ER) stress markers in a gender-independent manner. In addition, blockage of hypothalamic fatty acid beta oxidation prevents the ghrelin-promoting action on AgRP and NPY mRNA expression, also in a gender-independent manner. Notably, this effect is associated with decreased BSX expression and reduced food intake. Overall, our data suggest that BSX integrates changes in neuronal metabolic status with ARC-derived neuropeptides in a gender-independent manner.


Assuntos
Ácidos Graxos/metabolismo , Grelina/farmacologia , Proteínas de Homeodomínio/genética , Hipotálamo/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Neuropeptídeos/efeitos dos fármacos , Fatores de Transcrição/metabolismo , Proteína Relacionada com Agouti/efeitos dos fármacos , Proteína Relacionada com Agouti/genética , Animais , Ingestão de Alimentos , Feminino , Regulação da Expressão Gênica , Masculino , Neuropeptídeo Y/efeitos dos fármacos , Neuropeptídeo Y/genética , Neuropeptídeos/genética , Ratos , Fatores Sexuais , Fatores de Transcrição/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA