Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 128
Filtrar
2.
Scand J Immunol ; 94(4): e13092, 2021 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-34780075

RESUMO

The cholinergic anti-inflammatory pathway (CAP) is a classic neuroimmune pathway, consisting of the vagus nerve, acetylcholine (ACh)-the pivotal neurotransmitter of the vagus nerve-and its receptors. This pathway can activate and regulate the activities of immune cells, inhibit cell proliferation and differentiation, as well as suppress cytokine release, thereby playing an anti-inflammatory role, and widely involved in the occurrence and development of various diseases; recent studies have demonstrated that the CAP may be a new target for the treatment of autoimmune rheumatic diseases. In this review, we will summarize the latest progress with the view of figuring out the role of the cholinergic pathway and how it interacts with inflammatory reactions in several autoimmune rheumatic diseases, and many advances are results from a wide range of experiments performed in vitro and in vivo.


Assuntos
Doenças Autoimunes/etiologia , Doenças Reumáticas/etiologia , Acetilcolina/imunologia , Animais , Artrite Reumatoide/etiologia , Artrite Reumatoide/imunologia , Doenças Autoimunes/imunologia , Humanos , Inflamação/imunologia , Lúpus Eritematoso Sistêmico/etiologia , Lúpus Eritematoso Sistêmico/imunologia , Neuroimunomodulação , Osteoartrite/etiologia , Osteoartrite/imunologia , Receptores Colinérgicos/imunologia , Doenças Reumáticas/imunologia , Escleroderma Sistêmico/etiologia , Escleroderma Sistêmico/imunologia , Síndrome de Sjogren/etiologia , Síndrome de Sjogren/imunologia , Espondiloartropatias/etiologia , Espondiloartropatias/imunologia , Nervo Vago/imunologia
3.
Proc Natl Acad Sci U S A ; 118(20)2021 05 18.
Artigo em Inglês | MEDLINE | ID: mdl-33972441

RESUMO

Neuromodulation of immune function by stimulating the autonomic connections to the spleen has been demonstrated in rodent models. Consequently, neuroimmune modulation has been proposed as a new therapeutic strategy for the treatment of inflammatory conditions. However, demonstration of the translation of these immunomodulatory mechanisms in anatomically and physiologically relevant models is still lacking. Additionally, translational models are required to identify stimulation parameters that can be transferred to clinical applications of bioelectronic medicines. Here, we performed neuroanatomical and functional comparison of the mouse, rat, pig, and human splenic nerve using in vivo and ex vivo preparations. The pig was identified as a more suitable model of the human splenic innervation. Using functional electrophysiology, we developed a clinically relevant marker of splenic nerve engagement through stimulation-dependent reversible reduction in local blood flow. Translation of immunomodulatory mechanisms were then assessed using pig splenocytes and two models of acute inflammation in anesthetized pigs. The pig splenic nerve was shown to locally release noradrenaline upon stimulation, which was able to modulate cytokine production by pig splenocytes. Splenic nerve stimulation was found to promote cardiovascular protection as well as cytokine modulation in a high- and a low-dose lipopolysaccharide model, respectively. Importantly, splenic nerve-induced cytokine modulation was reproduced by stimulating the efferent trunk of the cervical vagus nerve. This work demonstrates that immune responses can be modulated by stimulation of spleen-targeted autonomic nerves in translational species and identifies splenic nerve stimulation parameters and biomarkers that are directly applicable to humans due to anatomical and electrophysiological similarities.


Assuntos
Sistema Imunitário/inervação , Imunomodulação/efeitos dos fármacos , Baço/imunologia , Sistema Nervoso Simpático/imunologia , Nervo Vago/imunologia , Animais , Feminino , Expressão Gênica , Humanos , Sistema Imunitário/efeitos dos fármacos , Inflamação , Interleucina-6/genética , Interleucina-6/imunologia , Lipopolissacarídeos/farmacologia , Camundongos , Microcirculação/efeitos dos fármacos , Microcirculação/genética , Microcirculação/imunologia , Norepinefrina/farmacologia , Ratos , Especificidade da Espécie , Baço/efeitos dos fármacos , Baço/inervação , Baço/patologia , Suínos , Sistema Nervoso Simpático/efeitos dos fármacos , Fator de Necrose Tumoral alfa/genética , Fator de Necrose Tumoral alfa/imunologia , Nervo Vago/efeitos dos fármacos , Estimulação do Nervo Vago/métodos
4.
FASEB J ; 35(3): e21320, 2021 03.
Artigo em Inglês | MEDLINE | ID: mdl-33660333

RESUMO

Influenza A virus (IAV) is rapidly detected in the airways by the immune system, with resident parenchymal cells and leukocytes orchestrating viral sensing and the induction of antiviral inflammatory responses. The airways are innervated by heterogeneous populations of vagal sensory neurons which also play an important role in pulmonary defense. How these neurons respond to IAV respiratory infection remains unclear. Here, we use a murine model to provide the first evidence that vagal sensory neurons undergo significant transcriptional changes following a respiratory IAV infection. RNA sequencing on vagal sensory ganglia showed that IAV infection induced the expression of many genes associated with an antiviral and pro-inflammatory response and this was accompanied by a significant increase in inflammatory cell recruitment into the vagal ganglia. Assessment of gene expression in single-vagal sensory neurons confirmed that IAV infection induced a neuronal inflammatory phenotype, which was most prominent in bronchopulmonary neurons, and also evident in some neurons innervating other organs. The altered transcriptome could be mimicked by intranasal treatment with cytokines and the lung homogenates of infected mice, in the absence of infectious virus. These data argue that IAV pulmonary infection and subsequent inflammation induces vagal sensory ganglia neuroinflammation and this may have important implications for IAV-induced morbidity.


Assuntos
Inflamação/imunologia , Vírus da Influenza A , Pulmão/inervação , Infecções por Orthomyxoviridae/imunologia , Células Receptoras Sensoriais/imunologia , Nervo Vago/imunologia , Animais , Feminino , Pulmão/virologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Células Receptoras Sensoriais/metabolismo , Transcrição Gênica , Nervo Vago/metabolismo
5.
Virulence ; 12(1): 360-376, 2021 12.
Artigo em Inglês | MEDLINE | ID: mdl-33380272

RESUMO

Abnormalities in CD4+ T cell (Th cell) differentiation play an important role in the pathogenesis of viral myocarditis (VMC). Our previous studies demonstrated that activation of the cholinergic anti-inflammatory pathway (CAP) alleviated the inflammatory response. In addition, we observed that right cervical vagotomy aggravates VMC by inhibiting CAP. However, the vagus nerve's effect on differentiation of CD4+ T cells has not been studied in VMC mice to date. In this study, we investigated the effects of cervical vagotomy and the α7nAChR agonist pnu282987 on CD4+ T cell differentiation in a murine myocarditis model (BALB/c) infected with coxsackievirus B3 (CVB3). Splenic CD4+ T cells from CVB3-induced mice obtained and cultured to investigate the potential mechanism of CD4+ T cell differentiation. Each Th cell subset was analyzed by flow cytometry. Our results showed that right cervical vagotomy increased proportions of Th1 and Th17 cells and decreased proportions of Th2 and Treg cells in the spleen. Vagotomy-induced upregulation of T-bet, Ror-γ, IFN-γ, and IL-17 expression while downregulating the expression of Gata3, Foxp3, and IL-4 in the heart. In addition, we observed upregulated levels of proinflammatory cytokines, aggravated myocardial lesions and cellular infiltration, and worsened cardiac function in VMC mice. Pnu282987 administration reversed these outcomes. Furthermore, vagotomy inhibited JAK2-STAT3 activation and enhanced NF-κB activation in splenic CD4+ T cells. The CD4+ T cell differentiation was related to JAK2-STAT3 and NF-κB signal pathways. In conclusion, vagus nerve modulates the inflammatory response by regulating CD4+ T cell differentiation in response to VMC.


Assuntos
Linfócitos T CD4-Positivos/fisiologia , Diferenciação Celular/imunologia , Infecções por Coxsackievirus/imunologia , Enterovirus Humano B/imunologia , Miocardite/imunologia , Miocardite/virologia , Nervo Vago/imunologia , Doença Aguda , Animais , Linfócitos T CD4-Positivos/imunologia , Citocinas/imunologia , Enterovirus Humano B/classificação , Masculino , Camundongos , Camundongos Endogâmicos BALB C
6.
Pharmacol Ther ; 222: 107794, 2021 06.
Artigo em Inglês | MEDLINE | ID: mdl-33310156

RESUMO

Obesity and the metabolic syndrome (MetS), which have reached pandemic proportions significantly increase the risk for type 2 diabetes, cardiovascular disease, and other serious conditions. Recent data with COVID-19 patients indicate that obesity also is a significant risk factor for this novel viral disease and poor outcome of associated critical illness. These findings considerably change the view of obesity as a driver of serious, but slowly-progressing chronic diseases, and emphasize the urgency to explore new therapeutic approaches. Inflammation is a recognized driver of metabolic derangements in obesity and MetS, and a core feature of COVID-19 pathobiology. Recent advances in our understanding of inflammatory regulation have highlighted the role of the nervous system and the vagus nerve-based inflammatory reflex. Current bioelectronic and pharmacological therapeutic explorations centered on the inflammatory reflex offer new approaches for conditions characterized by immune and metabolic dysregulation and for ameliorating the escalating burden of obesity, MetS, and COVID-19.


Assuntos
COVID-19 , Inflamação , Obesidade , Nervo Vago/imunologia , COVID-19/imunologia , COVID-19/metabolismo , Humanos , Inflamação/imunologia , Inflamação/metabolismo , Síndrome Metabólica/imunologia , Obesidade/epidemiologia , Obesidade/imunologia , Obesidade/terapia , SARS-CoV-2
7.
Int J Mol Sci ; 21(19)2020 Oct 02.
Artigo em Inglês | MEDLINE | ID: mdl-33023273

RESUMO

Anorexia nervosa represents a severe mental disorder associated with food avoidance and malnutrition. In patients suffering from anorexia nervosa, cardiovascular complications are the main reason leading to morbidity and mortality. However, the origin and pathological mechanisms leading to higher cardiovascular risk in anorexia nervosa are still unclear. In this aspect, the issue of exact pathological mechanisms as well as sensitive biomarkers for detection of anorexia nervosa-linked cardiovascular risk are discussed. Therefore, this review synthesised recent evidence of dysfunction in multiple neuroendocrine axes and alterations in the immune system that may represent anorexia nervosa-linked pathological mechanisms contributing to complex cardiovascular dysregulation. Further, this review is focused on identification of non-invasive biomarkers for the assessment of increased cardiovascular risk in anorexia nervosa that can be linked to a clinical application. Complex non-invasive assessment of cardiovascular autonomic regulation-cardiac vagal control (heart rate variability), sympathetic vascular activity (blood pressure variability), and cardiovascular reflex control (baroreflex sensitivity)-could represent a promising tool for early diagnosis, personalized therapy, and monitoring of therapeutic interventions in anorexia nervosa particularly at a vulnerable adolescent age.


Assuntos
Anorexia Nervosa/fisiopatologia , Sistema Nervoso Autônomo/fisiopatologia , Doenças Cardiovasculares/fisiopatologia , Sistemas Neurossecretores/fisiopatologia , Adolescente , Anorexia Nervosa/complicações , Anorexia Nervosa/imunologia , Pressão Sanguínea , Doenças Cardiovasculares/complicações , Doenças Cardiovasculares/imunologia , Feminino , Frequência Cardíaca , Humanos , Sistema Imunitário/fisiopatologia , Nervo Vago/imunologia , Nervo Vago/fisiopatologia
8.
Front Immunol ; 11: 595342, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33633726

RESUMO

The outbreak of coronavirus disease 2019 (COVID-19) underlined the urgent need for alleviating cytokine storm. We propose here that activating the cholinergic anti-inflammatory pathway (CAP) is a potential therapeutic strategy. However, there is currently no approved drugs targeting the regulatory pathway. It is evident that nicotine, anisodamine and some herb medicine, activate the CAP and exert anti-inflammation action in vitro and in vivo. As the vagus nerve affects both inflammation and specific immune response, we propose that vagus nerve stimulation by invasive or non-invasive devices and acupuncture at ST36, PC6, or GV20, are also feasible approaches to activate the CAP and control COVID-19. It is worth to investigate the efficacy and safety of the strategy in patients with COVID-19.


Assuntos
COVID-19/terapia , Síndrome da Liberação de Citocina/terapia , Neuroimunomodulação/imunologia , Estimulação do Nervo Vago/métodos , Nervo Vago/imunologia , Acupuntura , Anti-Inflamatórios/farmacologia , Citocinas/sangue , Medicamentos de Ervas Chinesas/farmacologia , Humanos , Inflamação/terapia , Nicotina/farmacologia , SARS-CoV-2 , Alcaloides de Solanáceas/farmacologia
9.
Artigo em Inglês | MEDLINE | ID: mdl-31138538

RESUMO

Bioelectronic medicine is an evolving field in which new insights into the regulatory role of the nervous system and new developments in bioelectronic technology result in novel approaches in disease diagnosis and treatment. Studies on the immunoregulatory function of the vagus nerve and the inflammatory reflex have a specific place in bioelectronic medicine. These studies recently led to clinical trials with bioelectronic vagus nerve stimulation in inflammatory diseases and other conditions. Here, we outline key findings from this preclinical and clinical research. We also point to other aspects and pillars of interdisciplinary research and technological developments in bioelectronic medicine.


Assuntos
Inflamação/fisiopatologia , Inflamação/terapia , Pesquisa Translacional Biomédica , Nervo Vago/fisiologia , Animais , Doenças Autoimunes/fisiopatologia , Doenças Autoimunes/terapia , Modelos Animais de Doenças , Humanos , Neuroimunomodulação , Reflexo , Nervo Vago/anatomia & histologia , Nervo Vago/imunologia
10.
Brain Behav Immun ; 80: 238-246, 2019 08.
Artigo em Inglês | MEDLINE | ID: mdl-30885844

RESUMO

The autonomic nervous system innervates all lymphoid tissues including the spleen therefore providing a link between the central nervous system and the immune system. The only known mechanism of neural inhibition of inflammation in the spleen relies on the production of norepinephrine by splenic catecholaminergic fibers which binds to ß2-adrenergic receptors (ß 2-ARs) of CD4+ T cells. These CD4+ T cells trigger the release of acetylcholine that inhibits the secretion of inflammatory cytokines by macrophages through α7 nicotinic acetylcholine receptor (α7nAchRs) signaling. While the vagal anti-inflammatory pathway has been extensively studied in rodents, it remains to be determined whether it coexists with other neural pathways. Here, we have found that three nerve branches project to the spleen in mice. While two of these nerves are associated with an artery and contain catecholaminergic fibers, the third is located at the apex of the spleen and contain both catecholaminergic and cholinergic fibers. We found that electrical stimulation of the apical nerve, but not the arterial nerves, inhibited inflammation independently of lymphocytes. In striking contrast to the anti-inflammatory pathway mechanism described so far, we also found that the inhibition of inflammation by apical nerve electrical stimulation relied on signaling by both ß 2-ARs and α7nAchRs in myeloid cells, with these two signaling pathways acting in parallel. Most importantly, apical splenic nerve electrical stimulation mitigated clinical symptoms in a mouse model of rheumatoid arthritis further providing the proof-of-concept that such an approach could be beneficial in patients with Immune-mediated inflammatory diseases.


Assuntos
Células Mieloides/imunologia , Receptores Adrenérgicos/imunologia , Receptores Nicotínicos/imunologia , Baço/imunologia , Baço/inervação , Acetilcolina/metabolismo , Animais , Estimulação Elétrica , Feminino , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Norepinefrina/metabolismo , Baço/fisiopatologia , Fator de Necrose Tumoral alfa/imunologia , Nervo Vago/imunologia , Estimulação do Nervo Vago
11.
Neuroscience ; 401: 59-72, 2019 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-30641114

RESUMO

Depression or stress is reportedly related to the overflow of inflammatory factors in the body and T cells were reported to play important roles in balancing the release of inflammatory factors through vagus nerve circuit. However, few works have been conducted to find if natural killer (NK) cells can also exert the similar function in the reported vagus nerve circuit as T cells and if there was any relationship between depression and this function. In the present study, the behavioral tests on BALB/c mice indicated that the depressant-like symptoms could be improved and simultaneously the concentrations of inflammatory factors in peripheral blood could be reduced significantly by adoptively transferring NK cells into stressed BALB/c mice. The results revealed that NK cells could control the release of inflammatory factors secreted by macrophages and ß2-AR (ß2-adrenergic receptor) on the NK cells were of great importance. Behavioral tests on NCG mice indicated that the antidepressant-like effects of NK cells notably declined after adoptively transferring NK cells with ß2-AR deficiency or with ChAT (choline acetyltransferase) deficiency into stressed NCG mice. Simultaneously, the anti-inflammatory effects also declined significantly both in vivo and in vitro, which indicated that the antidepressant-like property of NK cells may be related to its ability of controlling the release of inflammatory factors. Taken together, we find that NK cells may balance the release of inflammatory factors in our body by transporting the information between the terminal vagal branches and macrophages, which is the mechanism that NK cells may exert antidepressant-like effects.


Assuntos
Antidepressivos/imunologia , Citocinas/metabolismo , Inflamação/imunologia , Células Matadoras Naturais/imunologia , Animais , Antidepressivos/metabolismo , Comportamento Animal , Colina O-Acetiltransferase/metabolismo , Repetições Palindrômicas Curtas Agrupadas e Regularmente Espaçadas , Citocinas/farmacocinética , Inflamação/patologia , Células Matadoras Naturais/patologia , Macrófagos/imunologia , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Receptores Adrenérgicos beta 2/metabolismo , Estresse Psicológico/tratamento farmacológico , Nervo Vago/imunologia
12.
Artigo em Inglês | MEDLINE | ID: mdl-30126836

RESUMO

Recent studies have shown renal protective effects of bioelectric approaches, including ultrasound treatment, electrical vagus nerve stimulation, and optogenetic brainstem C1 neuron stimulation. The renal protection acquired by all three modalities was lost in splenectomized mice and/or α7 subunit of the nicotinic acetylcholine receptor-deficient mice. C1 neuron-mediated renal protection was blocked by ß2-adrenergic receptor antagonist. These findings indicate that all three methods commonly, at least partially, activate the cholinergic anti-inflammatory pathway, a well-studied neuroimmune pathway. In this article, we summarize the current understanding of neuroimmune axis-mediated kidney protection in preclinical models of acute kidney injury by these three modalities. Examination of the differences among these three modalities might lead to a further elucidation of the neuroimmune axis involved in renal protection and is of interest for developing new therapeutic approaches.


Assuntos
Injúria Renal Aguda/patologia , Neuroimunomodulação/imunologia , Injúria Renal Aguda/imunologia , Animais , Sistema Nervoso Autônomo/imunologia , Sistema Nervoso Autônomo/fisiopatologia , Humanos , Inflamação/patologia , Camundongos , Nervo Vago/imunologia , Nervo Vago/fisiopatologia
13.
Immunol Lett ; 202: 38-43, 2018 10.
Artigo em Inglês | MEDLINE | ID: mdl-30077536

RESUMO

Accumulating evidence points to a beneficial effect ofvagus nerve activity in tumor development. The vagus nerve is proposed to slow tumorigenesis because of its anti-inflammatory properties mediated through ACh and the α7nAChR. Since α7nAChRs are widely expressed by many types of immune cells we hypothesized that the vagus nerve affects the tumor microenvironment and anticancer immunity. We found direct evidence in studies using animal cancer models that vagus nerve stimulation alters immunological responses relevant to the tumor microenvironment. Also studies in pathologies other than cancer suggest a role for the vagus nerve in altering immunological responses relevant to anticancer immunity. These results provide a rationale to expect that vagus nerve stimulation, in combination with conventional cancer treatments, may improve the prognosis of cancer patients by promoting anticancer immunity.


Assuntos
Neoplasias/imunologia , Neoplasias/terapia , Nervo Vago/imunologia , Animais , Antineoplásicos/imunologia , Antineoplásicos/uso terapêutico , Humanos , Microambiente Tumoral/imunologia , Estimulação do Nervo Vago/métodos , Receptor Nicotínico de Acetilcolina alfa7/imunologia
14.
Am J Physiol Gastrointest Liver Physiol ; 315(5): G651-G658, 2018 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-30001146

RESUMO

Improved understanding of neuroimmune communication and the neural regulation of immunity and inflammation has recently led to proposing the concept of the "neuroimmune communicatome." This advance is based on experimental evidence for an organized and brain-integrated reflex-like relationship and dialogue between the nervous and the immune systems. A key circuitry in this communicatome is provided by efferent vagus nerve fibers and cholinergic signaling. Inflammation and metabolic alterations coexist in many disorders affecting the liver and the gastrointestinal (GI) tract, including obesity, metabolic syndrome, fatty liver disease, liver injury, and liver failure, as well as inflammatory bowel disease. Here, we outline mechanistic insights regarding the role of the vagus nerve and cholinergic signaling in the regulation of inflammation linked to metabolic derangements and the pathogenesis of these disorders in preclinical settings. Recent clinical advances using this knowledge in novel therapeutic neuromodulatory approaches within the field of bioelectronic medicine are also briefly summarized.


Assuntos
Neurônios Colinérgicos/fisiologia , Trato Gastrointestinal/fisiologia , Fígado/fisiologia , Neuroimunomodulação , Nervo Vago/fisiologia , Animais , Trato Gastrointestinal/imunologia , Humanos , Fígado/imunologia , Nervo Vago/imunologia
15.
Brain Behav Immun ; 73: 441-449, 2018 10.
Artigo em Inglês | MEDLINE | ID: mdl-29883598

RESUMO

Electrical stimulation of the vagus nerve (VNS) is a novel strategy used to treat inflammatory conditions. Therapeutic VNS activates both efferent and afferent fibers; however, the effects attributable to vagal afferent stimulation are unclear. Here, we tested if selective activation of afferent fibers in the abdominal vagus suppresses systemic inflammation. In urethane-anesthetized rats challenged with lipopolysaccharide (LPS, 60 µg/kg, i.v.), abdominal afferent VNS (2 Hz for 20 min) reduced plasma tumor necrosis factor alpha (TNF) levels 90 min later by 88% compared with unmanipulated animals. Pre-cutting the cervical vagi blocked this anti-inflammatory action. Interestingly, the surgical procedure to expose and prepare the abdominal vagus for afferent stimulation ('vagal manipulation') also had an anti-inflammatory action. Levels of the anti-inflammatory cytokine IL-10 were inversely related to those of TNF. Prior bilateral section of the splanchnic sympathetic nerves reversed the anti-inflammatory actions of afferent VNS and vagal manipulation. Sympathetic efferent activity in the splanchnic nerve was shown to respond reflexly to abdominal vagal afferent stimulation. These data demonstrate that experimentally activating abdominal vagal afferent fibers suppresses systemic inflammation, and that the efferent neural pathway for this action is in the splanchnic sympathetic nerves.


Assuntos
Inflamação/metabolismo , Nervos Esplâncnicos/fisiologia , Nervo Vago/fisiologia , Abdome/inervação , Vias Aferentes/metabolismo , Vias Aferentes/fisiologia , Animais , Anti-Inflamatórios/farmacologia , Citocinas , Modelos Animais de Doenças , Inflamação/imunologia , Interleucina-10/análise , Interleucina-10/sangue , Lipopolissacarídeos/farmacologia , Masculino , Vias Neurais , Ratos , Ratos Sprague-Dawley , Nervos Esplâncnicos/imunologia , Sistema Nervoso Simpático , Fator de Necrose Tumoral alfa/análise , Fator de Necrose Tumoral alfa/sangue , Nervo Vago/imunologia , Estimulação do Nervo Vago/métodos
16.
J Neurovirol ; 24(3): 379-381, 2018 06.
Artigo em Inglês | MEDLINE | ID: mdl-29532442

RESUMO

Vernet syndrome is a unilateral palsy of glossopharyngeal, vagus, and accessory nerves. Varicella zoster virus (VZV) infection has rarely been described as a possible cause. A 76-year-old man presented with 1-week-long symptoms of dysphonia, dysphagia, and weakness of the right shoulder elevation, accompanied by a mild right temporal parietal headache with radiation to the ipsilateral ear. Physical examination showed signs compatible with a right XI, X, and XI cranial nerves involvement and also several vesicular lesions in the right ear's concha. He had a personal history of poliomyelitis and chickenpox. Laringoscopy demonstrated right vocal cord palsy. Brain MRI showed thickening and enhancement of right lower cranial nerves and an enhancing nodular lesion in the ipsilateral jugular foramen, in T1 weighted images with gadolinium. Cerebrospinal fluid (CSF) analysis disclosed a mild lymphocytic pleocytosis and absence of VZV-DNA by PCR analysis. Serum VZV IgM and IgG antibodies were positive. The patient had a noticeable clinical improvement after initiation of acyclovir and prednisolone therapy. The presentation of a VZV infection with isolated IX, X, and XI cranial nerves palsy is extremely rare. In our case, the diagnosis of Vernet syndrome as a result of VZV infection was made essentially from clinical findings and supported by analytical and imaging data.


Assuntos
Encéfalo/virologia , Doenças dos Nervos Cranianos/virologia , Herpesvirus Humano 3/imunologia , Infecção pelo Vírus da Varicela-Zoster/virologia , Paralisia das Pregas Vocais/virologia , Nervo Acessório/diagnóstico por imagem , Nervo Acessório/imunologia , Nervo Acessório/fisiopatologia , Nervo Acessório/virologia , Idoso , Encéfalo/diagnóstico por imagem , Encéfalo/imunologia , Encéfalo/fisiopatologia , Doenças dos Nervos Cranianos/diagnóstico por imagem , Doenças dos Nervos Cranianos/imunologia , Doenças dos Nervos Cranianos/fisiopatologia , Nervo Glossofaríngeo/diagnóstico por imagem , Nervo Glossofaríngeo/imunologia , Nervo Glossofaríngeo/fisiopatologia , Nervo Glossofaríngeo/virologia , Herpesvirus Humano 3/isolamento & purificação , Humanos , Imageamento por Ressonância Magnética , Masculino , Nervo Vago/diagnóstico por imagem , Nervo Vago/imunologia , Nervo Vago/fisiopatologia , Nervo Vago/virologia , Infecção pelo Vírus da Varicela-Zoster/diagnóstico por imagem , Infecção pelo Vírus da Varicela-Zoster/imunologia , Infecção pelo Vírus da Varicela-Zoster/fisiopatologia , Paralisia das Pregas Vocais/diagnóstico por imagem , Paralisia das Pregas Vocais/imunologia , Paralisia das Pregas Vocais/fisiopatologia
17.
Adv Physiol Educ ; 41(4): 578-593, 2017 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-29138216

RESUMO

The autonomic nervous system is a powerful regulator of circulatory adjustments to acute hemodynamic stresses. Here we focus on new concepts that emphasize the chronic influence of the sympathetic and parasympathetic systems on cardiovascular pathology. The autonomic neurohumoral system can dramatically influence morbidity and mortality from cardiovascular disease through newly discovered influences on the innate and adaptive immune systems. Specifically, the end-organ damage in heart failure or hypertension may be worsened or alleviated by pro- or anti-inflammatory pathways of the immune system, respectively, that are activated through neurohumoral transmitters. These concepts provide a major new perspective on potentially life-saving therapeutic interventions in the deadliest of diseases.


Assuntos
Sistema Nervoso Autônomo/fisiologia , Doenças Cardiovasculares/imunologia , Sistema Imunitário/fisiologia , Imunidade Inata/fisiologia , Animais , Sistema Nervoso Autônomo/fisiopatologia , Barorreflexo/fisiologia , Doenças Cardiovasculares/fisiopatologia , Sistema Cardiovascular/imunologia , Sistema Cardiovascular/fisiopatologia , Citocinas/imunologia , Citocinas/metabolismo , Hemodinâmica/fisiologia , Humanos , Sistema Imunitário/fisiopatologia , Neurotransmissores/imunologia , Neurotransmissores/metabolismo , Nervo Vago/imunologia , Nervo Vago/metabolismo
18.
Physiol Res ; 66(Suppl 2): S139-S145, 2017 09 22.
Artigo em Inglês | MEDLINE | ID: mdl-28937230

RESUMO

Inflammation and other immune responses are involved in the variety of diseases and disorders. The acute response to endotoxemia includes activation of innate immune mechanisms as well as changes in autonomic nervous activity. The autonomic nervous system and the inflammatory response are intimately linked and sympathetic and vagal nerves are thought to have anti-inflammation functions. The basic functional circuit between vagus nerve and inflammatory response was identified and the neuroimmunomodulation loop was called cholinergic anti-inflammatory pathway. Unique function of vagus nerve in the anti-inflammatory reflex arc was found in many experimental and pre-clinical studies. They brought evidence on the cholinergic signaling interacting with systemic and local inflammation, particularly suppressing immune cells function. Pharmacological/electrical modulation of vagal activity suppressed TNF-alpha and other proinflammatory cytokines production and had beneficial therapeutic effects. Many questions related to mapping, linking and targeting of vagal-immune interactions have been elucidated and brought understanding of its basic physiology and provided the initial support for development of Tracey´s inflammatory reflex. This review summarizes and critically assesses the current knowledge defining cholinergic anti-inflammatory pathway with main focus on studies employing an experimental approach and emphasizes the potential of modulation of vagally-mediated anti-inflammatory pathway in the treatment strategies.


Assuntos
Anti-Inflamatórios/imunologia , Neurônios Colinérgicos/imunologia , Neuroimunomodulação/fisiologia , Transdução de Sinais/fisiologia , Nervo Vago/imunologia , Animais , Anti-Inflamatórios/metabolismo , Colinérgicos/imunologia , Colinérgicos/metabolismo , Neurônios Colinérgicos/metabolismo , Humanos , Inflamação/imunologia , Inflamação/metabolismo , Nervo Vago/metabolismo
19.
J Immunol ; 198(9): 3389-3397, 2017 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-28416717

RESUMO

The field of immunology is principally focused on the molecular mechanisms by which hematopoietic cells initiate and maintain innate and adaptive immunity. That cornerstone of attention has been expanded by recent discoveries that neuronal signals occupy a critical regulatory niche in immunity. The discovery is that neuronal circuits operating reflexively regulate innate and adaptive immunity. One particularly well-characterized circuit regulating innate immunity, the inflammatory reflex, is dependent upon action potentials transmitted to the reticuloendothelial system via the vagus and splenic nerves. This field has grown significantly with the identification of several other reflexes regulating discrete immune functions. As outlined in this review, the delineation of these mechanisms revealed a new understanding of immunity, enabled a first-in-class clinical trial using bioelectronic devices to inhibit cytokines and inflammation in rheumatoid arthritis patients, and provided a mosaic view of immunity as the integration of hematopoietic and neural responses to infection and injury.


Assuntos
Alergia e Imunologia , Inflamação Neurogênica , Neurociências , Baço/inervação , Nervo Vago/imunologia , Imunidade Adaptativa , Animais , Citocinas/metabolismo , Humanos , Imunidade Inata , Neuroimunomodulação , Reflexo/imunologia
20.
Immunity ; 46(1): 92-105, 2017 01 17.
Artigo em Inglês | MEDLINE | ID: mdl-28065837

RESUMO

Uncovering mechanisms that control immune responses in the resolution of bacterial infections is critical for the development of new therapeutic strategies that resolve infectious inflammation without unwanted side effects. We found that disruption of the vagal system in mice delayed resolution of Escherichia coli infection. Dissection of the right vagus decreased peritoneal group 3 innate lymphoid cell (ILC3) numbers and altered peritoneal macrophage responses. Vagotomy resulted in an inflammatory peritoneal lipid mediator profile characterized by reduced concentrations of pro-resolving mediators, including the protective immunoresolvent PCTR1, along with elevated inflammation-initiating eicosanoids. We found that acetylcholine upregulated the PCTR biosynthetic pathway in ILC3s. Administration of PCTR1 or ILC3s to vagotomized mice restored tissue resolution tone and host responses to E. coli infections. Together these findings elucidate a host protective mechanism mediated by ILC3-derived pro-resolving circuit, including PCTR1, that is controlled by local neuronal output to regulate tissue resolution tone and myeloid cell responses.


Assuntos
Ácidos Docosa-Hexaenoicos/imunologia , Mediadores da Inflamação/imunologia , Linfócitos/imunologia , Peritonite/imunologia , Nervo Vago/imunologia , Animais , Separação Celular , Modelos Animais de Doenças , Infecções por Escherichia coli/imunologia , Citometria de Fluxo , Humanos , Masculino , Camundongos , Vagotomia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA