Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters











Publication year range
1.
Neurotox Res ; 42(2): 16, 2024 Feb 20.
Article in English | MEDLINE | ID: mdl-38376791

ABSTRACT

Acetamiprid (ACE) and Imidacloprid (IMI) are widely-used neonicotinoid insecticides (NNIs) with functional activity at human acetylcholine nicotinic receptors and, therefore, with putative toxic effects. The objective of this study was the evaluation of the interactions between NNIs and α7-nAChR, as this receptor keeps intracellular Ca2+ ([Ca2+]i) to an optimum for an adequate neuronal functioning. Possible interactions between NNIs and the cryo-EM structure of the human α-7 nAChR were identified by molecular docking. Additionally, NNI effects were analyzed in neuroblastoma SH-SY5Y cells, as they naturally express α-7 nAChRs. Functional studies included proliferative/cytotoxic effects (MTT test) in undifferentiated SH-SY-5Y cells and indirect measurements of [Ca2+]i transients in retinoic acid-differentiated SH-SY-5Y cells loaded with Fluo-4 AM. Docking analysis showed that the binding of IMI and ACE occurred at the same aromatic cage that the specific α-7 nAChR agonist EVP-6124. IMI showed a better docking strength than ACE. According to the MTT assays, low doses (10-50 µM) of IMI better than ACE stimulated neuroblastoma cell proliferation. At higher doses (250-500 µM), IMI also prevailed over ACE and dose-dependently triggered more abrupt fluorescence changes due to [Ca2+]i mobilization in differentiated SH-SY5Y neurons. Indeed, only IMI blunted nicotine-evoked intracellular fluorescence stimulation (i.e., nicotine cross-desensitization). Summarizing, IMI demonstrated a superior docking strength and more robust cellular responses compared to ACE, which were likely associated with a stronger activity at α-7nAChRs. Through the interaction with α-7nAChRs, IMI would demonstrate its high neurotoxic potential for humans. More research is needed for investigating the proliferative effects of IMI in neuroblastoma cells.


Subject(s)
Insecticides , Neuroblastoma , Nitro Compounds , Receptors, Nicotinic , Humans , Calcium , Insecticides/toxicity , Molecular Docking Simulation , Nicotine/pharmacology , Neonicotinoids/toxicity
2.
Food Res Int ; 176: 113808, 2024 Jan.
Article in English | MEDLINE | ID: mdl-38163714

ABSTRACT

Hypothalamic inflammation and metabolic changes resulting from the consumption of high-fat diets have been linked to low grade inflammation and obesity. Inflammation impairs the hypothalamic expression of α7 nicotinic acetylcholine receptor (α7nAChR). The α7nAChR is described as the main component of the anti-inflammatory cholinergic pathway in different inflammation models. To assess whether the reduction in α7nAChR expression exacerbates hypothalamic inflammation induced by a high-fat diet (HFD), were used male and female global α7nAChR knockout mouse line in normal or high-fat diet for 4 weeks. Body weight gain, adiposity, glucose homeostasis, hypothalamic inflammation, food intake, and energy expenditure were evaluated. Insulin sensitivity was evaluated in neuronal cell culture. Consumption of an HFD for 4 weeks resulted in body weight gain and adiposity in male Chrna7-/- mice and the hypothalamus of male Chrna7-/- mice showed neuroinflammatory markers, with increased gene expression of pro-inflammatory cytokines and dysregulation in the nuclear factor kappa B pathway. Moreover, male Chrna7-/- mice consuming an HFD showed alterations in glucose homeostasis and serum of Chrna7-/- mice that consumed an HFD impaired insulin signalling in neuronal cell culture experiments. In general, female Chrna7-/- mice that consumed an HFD did not show the phenotypic and molecular changes found in male mice, indicating that there is sexual dimorphism in the analysed parameters. Thus, receptor deletion resulted in increased susceptibility to hypothalamic inflammation and metabolic damage associated with HFD consumption in male mice.


Subject(s)
Diet, High-Fat , alpha7 Nicotinic Acetylcholine Receptor , Male , Female , Animals , Mice , Diet, High-Fat/adverse effects , alpha7 Nicotinic Acetylcholine Receptor/genetics , alpha7 Nicotinic Acetylcholine Receptor/metabolism , Mice, Knockout , Obesity/genetics , Obesity/metabolism , Inflammation/metabolism , Weight Gain , Hypothalamus/metabolism , Phenotype , Glucose/metabolism
3.
Inflammation ; 47(3): 958-974, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38227123

ABSTRACT

Pulmonary emphysema is a primary component of chronic obstructive pulmonary disease (COPD), a life-threatening disorder characterized by lung inflammation and restricted airflow, primarily resulting from the destruction of small airways and alveolar walls. Cumulative evidence suggests that nicotinic receptors, especially the α7 subtype (α7nAChR), is required for anti-inflammatory cholinergic responses. We postulated that the stimulation of α7nAChR could offer therapeutic benefits in the context of pulmonary emphysema. To investigate this, we assessed the potential protective effects of PNU-282987, a selective α7nAChR agonist, using an experimental emphysema model. Male mice (C57BL/6) were submitted to a nasal instillation of porcine pancreatic elastase (PPE) (50 µl, 0.667 IU) to induce emphysema. Treatment with PNU-282987 (2.0 mg/kg, ip) was performed pre and post-emphysema induction by measuring anti-inflammatory effects (inflammatory cells, cytokines) as well as anti-remodeling and anti-oxidant effects. Elastase-induced emphysema led to an increase in the number of α7nAChR-positive cells in the lungs. Notably, both groups treated with PNU-282987 (prior to and following emphysema induction) exhibited a significant decrease in the number of α7nAChR-positive cells. Furthermore, both groups treated with PNU-282987 demonstrated decreased levels of macrophages, IL-6, IL-1ß, collagen, and elastic fiber deposition. Additionally, both groups exhibited reduced STAT3 phosphorylation and lower levels of SOCS3. Of particular note, in the post-treated group, PNU-282987 successfully attenuated alveolar enlargement, decreased IL-17 and TNF-α levels, and reduced the recruitment of polymorphonuclear cells to the lung parenchyma. Significantly, it is worth noting that MLA, an antagonist of α7nAChR, counteracted the protective effects of PNU-282987 in relation to certain crucial inflammatory parameters. In summary, these findings unequivocally demonstrate the protective abilities of α7nAChR against elastase-induced emphysema, strongly supporting α7nAChR as a pivotal therapeutic target for ameliorating pulmonary emphysema.


Subject(s)
Benzamides , Bridged Bicyclo Compounds , Mice, Inbred C57BL , Nicotinic Agonists , Pancreatic Elastase , Pulmonary Emphysema , alpha7 Nicotinic Acetylcholine Receptor , Animals , alpha7 Nicotinic Acetylcholine Receptor/agonists , alpha7 Nicotinic Acetylcholine Receptor/metabolism , Pulmonary Emphysema/drug therapy , Pulmonary Emphysema/chemically induced , Pulmonary Emphysema/metabolism , Pulmonary Emphysema/prevention & control , Mice , Benzamides/pharmacology , Benzamides/therapeutic use , Male , Bridged Bicyclo Compounds/pharmacology , Bridged Bicyclo Compounds/therapeutic use , Nicotinic Agonists/pharmacology , Nicotinic Agonists/therapeutic use , Lung/pathology , Lung/drug effects , Lung/metabolism , Anti-Inflammatory Agents/pharmacology , Anti-Inflammatory Agents/therapeutic use
4.
FASEB J ; 37(9): e23120, 2023 09.
Article in English | MEDLINE | ID: mdl-37527279

ABSTRACT

The α7nAChR is crucial to the anti-inflammatory reflex, and to the expression of neuropeptides that control food intake, but its expression can be decreased by environmental factors. We aimed to investigate whether microRNA modulation could be an underlying mechanism in the α7nAchR downregulation in mouse hypothalamus following a short-term exposure to an obesogenic diet. Bioinformatic analysis revealed Let-7 microRNAs as candidates to regulate Chrna7, which was confirmed by the luciferase assay. Mice exposed to an obesogenic diet for 3 days had increased Let-7a and decreased α7nAChR levels, accompanied by hypothalamic fatty acids and TNFα content. Hypothalamic neuronal cells exposed to fatty acids presented higher Let-7a and TNFα levels and lower Chrna7 expression, but when the cells were pre-treated with TLR4 inhibitor, Let-7a, TNFα, and Chrna7 were rescued to normal levels. Thus, the fatty acids overload trigger TNFα-induced Let-7 overexpression in hypothalamic neuronal cells, which negatively regulates α7nAChR, an event that can be related to hyperphagia and obesity predisposition in mice.


Subject(s)
Tumor Necrosis Factor-alpha , alpha7 Nicotinic Acetylcholine Receptor , Animals , Mice , alpha7 Nicotinic Acetylcholine Receptor/genetics , alpha7 Nicotinic Acetylcholine Receptor/metabolism , Tumor Necrosis Factor-alpha/metabolism , Fatty Acids , Down-Regulation , Hypothalamus/metabolism
5.
Brain Res ; 1727: 146567, 2020 01 15.
Article in English | MEDLINE | ID: mdl-31783002

ABSTRACT

New findings on neural regulation of immunity are allowing the design of novel pharmacological strategies to control inflammation and nociception. Herein, we report that choline, a 7-nicotinic acetylcholine receptor (α7nAChRs) agonist, prevents carrageenan-induced hyperalgesia without affecting inflammatory parameters (neutrophil migration or cytokine/chemokines production) or inducing sedation or even motor impairment. Choline also attenuates prostaglandin-E2 (PGE2)-induced hyperalgesia via α7nAChR activation and this antinociceptive effect was abrogated by administration of LNMMA (a nitric oxide synthase inhibitor), ODQ (an inhibitor of soluble guanylate cyclase; cGMP), andglibenclamide(an inhibitor of ATP-sensitive potassium channels). Furthermore, choline attenuates long-lasting Complete Freund's Adjuvant and incision-induced hyperalgesia suggesting its therapeutic potential to treat pain in rheumatoid arthritis or post-operative recovery, respectively. Our results suggest that choline modulates inflammatory hyperalgesia by activating the nitric oxide/cGMP/ATP-sensitive potassium channels without interfering in inflammatory events, and could be used in persistent pain conditions.


Subject(s)
Choline/pharmacology , Cyclic GMP/metabolism , Hyperalgesia/drug therapy , Inflammation/drug therapy , KATP Channels/metabolism , Nitric Oxide/metabolism , alpha7 Nicotinic Acetylcholine Receptor/agonists , Animals , Choline/therapeutic use , Dinoprostone/metabolism , Freund's Adjuvant , Male , Mice , Mice, Inbred BALB C
6.
Brain Behav Immun Health ; 2: 100034, 2020 Feb.
Article in English | MEDLINE | ID: mdl-38377429

ABSTRACT

Mental disorders (MDs) are highly prevalent and potentially debilitating complex disorders which causes remain elusive. Looking into deeper aspects of etiology or pathophysiology underlying these diseases would be highly beneficial, as the scarce knowledge in mechanistic and molecular pathways certainly represents an important limitation. Association between MDs and inflammation/neuroinflammation has been widely discussed and accepted by many, as high levels of pro-inflammatory cytokines were reported in patients with several MDs, such as schizophrenia (SCZ), bipolar disorder (BD) and major depression disorder (MDD), among others. Correlation of pro-inflammatory markers with symptoms intensity was also reported. However, the mechanisms underlying the inflammatory dysfunctions observed in MDs are not fully understood yet. In this context, microglial dysfunction has recently emerged as a possible pivotal player, as during the neuroinflammatory response, microglia can be over-activated, and excessive production of pro-inflammatory cytokines, which can modify the kynurenine and glutamate signaling, is reported. Moreover, microglial activation also results in increased astrocyte activity and consequent glutamate release, which are both toxic to the Central Nervous System (CNS). Also, as a result of increased microglial activation in MDs, products of the kynurenine pathway were shown to be changed, influencing then the dopaminergic, serotonergic, and glutamatergic signaling pathways. Therefore, in the present review, we aim to discuss how neuroinflammation impacts on glutamate and kynurenine signaling pathways, and how they can consequently influence the monoaminergic signaling. The consequent association with MDs main symptoms is also discussed. As such, this work aims to contribute to the field by providing insights into these alternative pathways and by shedding light on potential targets that could improve the strategies for pharmacological intervention and/or treatment protocols to combat the main pharmacologically unmatched symptoms of MDs, as the SCZ.

7.
Int J Mol Sci ; 19(5)2018 May 16.
Article in English | MEDLINE | ID: mdl-29772664

ABSTRACT

Macrophages are phagocytic immune cells that protect the body from foreign invaders and actively support the immune response by releasing anti- and proinflammatory cytokines. A seminal finding revolutionized the way macrophages are seen. The expression of the neuronal alpha7 nicotinic acetylcholine receptor (α7-nAChR) in macrophages led to the establishment of the cholinergic anti-inflammatory response (CAR) in which the activation of this receptor inactivates macrophage production of proinflammatory cytokines. This novel neuroimmune response soon began to emerge as a potential target to counteract inflammation during illness and infection states. Human immunodeficiency virus (HIV)-infected individuals suffer from chronic inflammation that persists even under antiretroviral therapy. Despite the CAR's importance, few studies involving macrophages have been performed in the HIV field. Evidence demonstrates that monocyte-derived macrophages (MDMs) recovered from HIV-infected individuals are upregulated for α7-nAChR. Moreover, in vitro studies demonstrate that addition of an HIV viral constituent, gp120IIIB, to uninfected MDMs also upregulates the α7-nAChR. Importantly, contrary to what was expected, activation of upregulated α7-nAChRs in macrophages does not reduce inflammation, suggesting a CAR disruption. Although it is reasonable to consider this receptor as a pharmacological target, additional studies are necessary since its activity seems to differ from that observed in neurons.


Subject(s)
HIV Infections/complications , Inflammation/etiology , Inflammation/metabolism , Macrophages/immunology , Macrophages/metabolism , Receptors, Nicotinic/metabolism , Acetylcholine/metabolism , Acetylcholine/pharmacology , Animals , Cytokines/metabolism , Drug Discovery , HIV Infections/drug therapy , HIV Infections/immunology , HIV Infections/virology , Humans , Immunity , Inflammation/pathology , Inflammation Mediators , Macrophages/drug effects , Monocytes/immunology , Monocytes/metabolism , Receptors, Nicotinic/genetics , alpha7 Nicotinic Acetylcholine Receptor/metabolism
8.
Front Immunol ; 8: 618, 2017.
Article in English | MEDLINE | ID: mdl-28620379

ABSTRACT

Schizophrenia is one of the most debilitating mental disorders and is aggravated by the lack of efficacious treatment. Although its etiology is unclear, epidemiological studies indicate that infection and inflammation during development induces behavioral, morphological, neurochemical, and cognitive impairments, increasing the risk of developing schizophrenia. The inflammatory hypothesis of schizophrenia is also supported by clinical studies demonstrating systemic inflammation and microglia activation in schizophrenic patients. Although elucidating the mechanism that induces this inflammatory profile remains a challenge, mounting evidence suggests that neuroimmune interactions may provide therapeutic advantages to control inflammation and hence schizophrenia. Recent studies have indicated that vagus nerve stimulation controls both peripheral and central inflammation via alpha-7 nicotinic acetylcholine receptor (α7nAChR). Other findings have indicated that vagal stimulation and α7nAChR-agonists can provide therapeutic advantages for neuropsychiatric disorders, such as depression and epilepsy. This review analyzes the latest results regarding: (I) the immune-to-brain pathogenesis of schizophrenia; (II) the regulation of inflammation by the autonomic nervous system in psychiatric disorders; and (III) the role of the vagus nerve and α7nAChR in schizophrenia.

9.
São Paulo; s.n; 2016. [137] p. ilus, tab, graf.
Thesis in Portuguese | LILACS | ID: biblio-870883

ABSTRACT

A lesão pulmonar aguda (LPA) é caracterizada por inflamação pulmonar de início súbito com recrutamento de polimorfonucleares e liberação de mediadores próinflamatórios. É uma condição grave que evolui com óbito em aproximadamente 40% dos casos. Diversos estudos que elucidaram a fisiopatologia da LPA, o tratamento ainda é insatisfatório. O sistema colinérgico anti-inflamatório foi descrito no pulmão e está relacionado a um reflexo via nervo vago que inibe a liberação de citocinas inflamatórias por efeitos relacionados a ação da acetilcolina em receptores nicotínicos. Nossa hipótese é de que a redução de VAChT, que está relacionada ao déficit na liberação de ACh, module a resposta inflamatória pulmonar em modelo de LPS. Objetivo: 1. Avaliar se a deficiência de VAChT modula a resposta pulmonar em animais geneticamente modificados; 2. Avaliar se a deficiência colinérgica induzida por redução de VAChT está envolvida na resposta pulmonar ao LPS e elucidar alguns mecanismos envolvidos; 3. Avaliar o potencial terapêutico do PNU, um agonista de alfa7nAChR nas alterações funcionais e histopatológicas em modelo de LPA em animais C57Bl6. Metodologia: Foram utilizados camundongos machos geneticamente modificados mutante (VAChT KDHOM) ou selvagem (WT) e C57BL/6. Inicialmente avaliamos a função pulmonar e a histopatologia pulmonar em animais VAChT KDHOM. Após, animais WT e VAChT KDHOM receberam instilação intranasal de LPS ou salina e a resposta inflamatória foi avaliada de 1,5h até 72 horas após. Ainda, foi avaliado a resposta pulmonar em VAChT KDHOM e WT após a instilação de LPS intraperitoneal. Por fim, animais C57BL/6 instilados com LPS intranasal, receberam tratamento prévio ou após com PNU, agonista do receptor nicotínico alfa7. Resultados: Animais mutante apresentaram maior quantidade de células recuperadas no lavado bronco alveolar (LBA) e aumento de citocinas próinflamatórias, aumento de edema peribrônquico e piora da função pulmonar. Ainda,...


Acute lung injury (ALI) is characterized by acute lung inflammation with recruitment of polymorphonuclear and release of proinflammatory mediators. It is a severe condition since leads to death 40% of the cases. Several studies have elucidated the pathophysiology of ALI, however the treatment is still unsatisfactory. The anti-inflammatory cholinergic system was described in the lung and is related to a vagal nerve reflex that inhibits the release of inflammatory cytokines by the action o ACh on nicotinic receptors. Our hypothesis is that the VAChT reduction, which is related to the deficit in the release of ACh, modulates the pulmonary inflammatory response in a model of LPS. Aim: 1. To assess whether VAChT deficiency modulates the pulmonary response in genetically modified animals; 2. Assess whether cholinergic deficiency induced reduction VAChT is involved in pulmonary response to LPS and elucidate some mechanisms involved; 3. To evaluate the therapeutic potential of PNU, an agonist alfa7nAChR, in functional and histological changes in C57BL6 mice with LPA. Methods: Mutant genetically modified male mice (VAChT KDHOM) or wild (WT) and C57BL/6 were used. First, we evaluated lung function and lung histopathology in VAChT KDHOM animals. After, WT animals and VAChT KDHOM received intranasal instillation of LPS or saline and the inflammatory response was assessed 1.5 hours to 72 hours. Moreover, the pulmonary response was evaluated in WT and VAChT KDHOM after instillation of LPS intraperitoneally. Finally, C57BL6 instilled with intranasal LPS received prior or post-treatment with PNU, an alfa7 nicotinic receptor agonist. Results: Mutant animals had higher number of cells recovered in brochoalveolar lavage (BAL) and increased pro-inflammatory cytokines, peribronchial edema and worsening of lung function. Still, there was an increase of NF_kB expression and reduction of JAK2. The VAChT deficiency induced increase in inflammatory cells...


Subject(s)
Animals , Male , Mice , Acetylcholine , Acute Lung Injury , Mice , Models, Animal , Pneumonia , Vesicular Acetylcholine Transport Proteins
10.
J Physiol Paris ; 108(4-6): 286-91, 2014.
Article in English | MEDLINE | ID: mdl-24819880

ABSTRACT

Central cholinergic system is critically involved in all known memory processes. Endogenous acetylcholine release by cholinergic neurons is necessary for modulation of acquisition, encoding, consolidation, reconsolidation, extinction, retrieval and expression. Experiments from our laboratory are mainly focused on elucidating the mechanisms by which acetylcholine modulates memory processes. Blockade of hippocampal alpha-7-nicotinic receptors (α7-nAChRs) with the antagonist methyllycaconitine impairs memory reconsolidation. However, the administration of a α7-nAChR agonist (choline) produce a paradoxical modulation, causing memory enhancement in mice trained with a weak footshock, but memory impairment in animals trained with a strong footshock. All these effects are long-lasting, and depend on the age of the memory trace. This review summarizes and discusses some of our recent findings, particularly regarding the involvement of α7-nAChRs on memory reconsolidation.


Subject(s)
Central Nervous System/drug effects , Cholinergic Agents/metabolism , Cholinergic Agents/pharmacology , Memory/drug effects , Neuropharmacology , Animals , Central Nervous System/metabolism , Humans , alpha7 Nicotinic Acetylcholine Receptor/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL