Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 614
Filtrar
1.
Heart Rhythm ; 21(5): 610-619, 2024 May.
Artigo em Inglês | MEDLINE | ID: mdl-38160759

RESUMO

BACKGROUND: Neural remodeling in the left stellate ganglion (LSG), as mediated by neuroimmune reactions, promotes cardiac sympathetic nerve activity (SNA) and thus increases the incidence of ventricular arrhythmias (VAs). Interleukin-6 (IL-6) is an important factor of the neuroimmune interaction. OBJECTIVE: The present study explored the effects of IL-6 on LSG hyperactivity and the incidence of VAs. METHODS: Eighteen beagles were randomly allocated to a control group (saline with myocardial infarction [MI], n = 6), adeno-associated virus (AAV) group (AAV with MI, n = 6), and IL-6 group (overexpression of IL-6 via AAV vector with MI, n = 6). Ambulatory electrocardiography was performed before and 30 days after AAV microinjection into the LSG. LSG function and ventricular electrophysiology were assessed at 31 days after surgery, and a canine MI model was established. Samples of the LSG were collected for immunofluorescence staining and molecular biological evaluation. Blood samples and 24-hour Holter data were obtained from 24 patients with acute MI on the day after they underwent percutaneous coronary intervention to assess the correlation between IL-6 levels and SNA. RESULTS: IL-6 overexpression increased cardiac SNA and worsened postinfarction VAs. Furthermore, sustained IL-6 overexpression enhanced LSG function, promoted expression of nerve growth factor, c-fos, and fos B in the LSG, and activated the signal transducer and activator of transcription 3/regulator of G protein signalling 4 signaling pathway. Clinical sample analysis revealed a correlation between serum IL-6 levels and heart rate variability frequency domain index as well as T-wave alternans. CONCLUSION: IL-6 levels are correlated with cardiac SNA. Chronic overexpression of IL-6 mediates LSG neural remodeling through the signal transducer and activator of transcription 3/regulator of G protein signalling 4 signaling pathway, elevating the risk of VA after MI.


Assuntos
Modelos Animais de Doenças , Interleucina-6 , Gânglio Estrelado , Animais , Cães , Interleucina-6/metabolismo , Gânglio Estrelado/metabolismo , Arritmias Cardíacas/etiologia , Masculino , Eletrocardiografia Ambulatorial/métodos , Isquemia Miocárdica/metabolismo , Isquemia Miocárdica/fisiopatologia , Sistema Nervoso Simpático/fisiopatologia , Sistema Nervoso Simpático/metabolismo , Neuroimunomodulação/fisiologia , Humanos , Taquicardia Ventricular/etiologia , Taquicardia Ventricular/fisiopatologia , Taquicardia Ventricular/metabolismo , Taquicardia Ventricular/terapia
2.
Front Immunol ; 12: 754208, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34733286

RESUMO

The autonomic nervous system has been studied for its involvement in the control of macrophages; however, the mechanisms underlying the interaction between the adrenergic receptors and alternatively activated macrophages (M2) remain obscure. Using FVB wild-type and beta 2 adrenergic receptors knockout, we found that ß2-AR deficiency alleviates hepatobiliary damage in mice infected with C. sinensis. Moreover, ß2-AR-deficient mice decrease the activation and infiltration of M2 macrophages and decrease the production of type 2 cytokines, which are associated with a significant decrease in liver fibrosis in infected mice. Our in vitro results on bone marrow-derived macrophages revealed that macrophages from Adrb2-/- mice significantly decrease M2 markers and the phosphorylation of ERK/mTORC1 induced by IL-4 compared to that observed in M2 macrophages from Adrb2+/+ . This study provides a better understanding of the mechanisms by which the ß2-AR enhances type 2 immune response through the ERK/mTORC1 signaling pathway in macrophages and their role in liver fibrosis.


Assuntos
Clonorquíase/complicações , Cirrose Hepática Biliar/imunologia , Cirrose Hepática/imunologia , Ativação de Macrófagos , Neuroimunomodulação/fisiologia , Receptores Adrenérgicos beta 2/fisiologia , Animais , Sistema Nervoso Autônomo/fisiopatologia , Ductos Biliares/parasitologia , Ductos Biliares/patologia , Células Cultivadas , Clonorquíase/imunologia , Clonorquíase/fisiopatologia , Citocinas/sangue , Humanos , Cirrose Hepática/etiologia , Cirrose Hepática/parasitologia , Cirrose Hepática/patologia , Cirrose Hepática Biliar/etiologia , Cirrose Hepática Biliar/parasitologia , Cirrose Hepática Biliar/patologia , Sistema de Sinalização das MAP Quinases , Macrófagos/classificação , Macrófagos/imunologia , Masculino , Alvo Mecanístico do Complexo 1 de Rapamicina/fisiologia , Camundongos Knockout , Receptores Adrenérgicos beta 2/deficiência , Organismos Livres de Patógenos Específicos
3.
Psychoneuroendocrinology ; 131: 105295, 2021 09.
Artigo em Inglês | MEDLINE | ID: mdl-34119855

RESUMO

The majority of COVID-19 survivors experience long-term neuropsychiatric symptoms such as fatigue, sleeping difficulties, depression and anxiety. We propose that neuroimmune cross-talk via inflammatory cytokines such as interleukin-6 (IL-6) could underpin these long-term COVID-19 symptoms. This hypothesis is supported by several lines of research, including population-based cohort and genetic Mendelian Randomisation studies suggesting that inflammation is associated with fatigue and sleeping difficulties, and that IL-6 could represent a possible causal driver for these symptoms. Immune activation following COVID-19 can disrupt T helper 17 (TH17) and regulatory T (Treg) cell responses, affect central learning and emotional processes, and lead to a vicious cycle of inflammation and mitochondrial dysfunction that amplifies the inflammatory process and results in immuno-metabolic constraints on neuronal energy metabolism, with fatigue being the ultimate result. Increased cytokine activity drives this process and could be targeted to interrupt it. Therefore, whether persistent IL-6 dysregulation contributes to COVID-19-related long-term fatigue, sleeping difficulties, depression, and anxiety, and whether targeting IL-6 pathways could be helpful for treatment and prevention of long COVID are important questions that require investigation. This line of research could inform new approaches for treatment and prevention of long-term neuropsychiatric symptoms of COVID-19. Effective treatment and prevention of this condition could also help to stem the anticipated rise in depression and other mental illnesses ensuing this pandemic.


Assuntos
COVID-19/complicações , Interleucina-6/fisiologia , Transtornos Mentais/etiologia , Animais , Ansiedade/epidemiologia , Ansiedade/etiologia , COVID-19/epidemiologia , COVID-19/etiologia , COVID-19/psicologia , Estudos de Coortes , Depressão/epidemiologia , Depressão/etiologia , Fadiga/epidemiologia , Fadiga/etiologia , Humanos , Interleucina-6/metabolismo , Interleucina-6/farmacologia , Transtornos Mentais/epidemiologia , Neuroimunomodulação/efeitos dos fármacos , Neuroimunomodulação/fisiologia , SARS-CoV-2/fisiologia , Transtornos do Sono-Vigília/epidemiologia , Transtornos do Sono-Vigília/etiologia , Sobreviventes/estatística & dados numéricos , Síndrome de COVID-19 Pós-Aguda
4.
Front Immunol ; 12: 649786, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33859641

RESUMO

Neuromodulation of the immune system has been proposed as a novel therapeutic strategy for the treatment of inflammatory conditions. We recently demonstrated that stimulation of near-organ autonomic nerves to the spleen can be harnessed to modulate the inflammatory response in an anesthetized pig model. The development of neuromodulation therapy for the clinic requires chronic efficacy and safety testing in a large animal model. This manuscript describes the effects of longitudinal conscious splenic nerve neuromodulation in chronically-implanted pigs. Firstly, clinically-relevant stimulation parameters were refined to efficiently activate the splenic nerve while reducing changes in cardiovascular parameters. Subsequently, pigs were implanted with a circumferential cuff electrode around the splenic neurovascular bundle connected to an implantable pulse generator, using a minimally-invasive laparoscopic procedure. Tolerability of stimulation was demonstrated in freely-behaving pigs using the refined stimulation parameters. Longitudinal stimulation significantly reduced circulating tumor necrosis factor alpha levels induced by systemic endotoxemia. This effect was accompanied by reduced peripheral monocytopenia as well as a lower systemic accumulation of CD16+CD14high pro-inflammatory monocytes. Further, lipid mediator profiling analysis demonstrated an increased concentration of specialized pro-resolving mediators in peripheral plasma of stimulated animals, with a concomitant reduction of pro-inflammatory eicosanoids including prostaglandins. Terminal electrophysiological and physiological measurements and histopathological assessment demonstrated integrity of the splenic nerves up to 70 days post implantation. These chronic translational experiments demonstrate that daily splenic nerve neuromodulation, via implanted electronics and clinically-relevant stimulation parameters, is well tolerated and is able to prime the immune system toward a less inflammatory, pro-resolving phenotype.


Assuntos
Terapia por Estimulação Elétrica/métodos , Endotoxemia/terapia , Neuroimunomodulação/fisiologia , Nervos Esplâncnicos/fisiologia , Baço/inervação , Animais , Modelos Animais de Doenças , Terapia por Estimulação Elétrica/instrumentação , Eletrodos Implantados , Endotoxemia/imunologia , Feminino , Inflamação/imunologia , Inflamação/terapia , Baço/imunologia , Sus scrofa
5.
Artigo em Inglês | MEDLINE | ID: mdl-33687904

RESUMO

Immune dysregulation, neuronal inflammation, and oligodendrocyte degradation are key causes for autoimmune disorders like multiple sclerosis (MS) and various other immune dysregulated neurodegenerative complications responsible for CNS-mediated immune responses. Sirtuin (SIRT-1) is a nicotinamide adenosine dinucleotide (NAD)-dependent transcriptional protein that deacetylases and removes acetyl groups from its transcription factors like P53, FOXO, NF-Κb, PGC-1α. SIRT-1 mediates a wide range of physiological functions, including gene transcription, metabolism, neuronal apoptosis, and glucose production. SIRT-1 dysregulation targets transcription factors, and other molecular alterations such as gene expression modification influence neuronal plasticity, inhibit Th17 cells, and interleukin-1ß can aggravate brain diseases. Preclinical and clinical findings show that the upregulation of SIRT-1 reduces autoimmunity, neurodegeneration, and neuroexcitation. Even though drugs are being developed for symptomatic therapies in clinical trials, there are particular pharmacological implications for improving post-operative conditions in neurodegenerative patients where intensive care is required. Understanding the SIRT-1 signaling and identifying immune-mediated neuron deterioration can detect major therapeutic interventions that could prevent neuro complications. Thus, in the current review, we have addressed the manifestations of disease by the downregulation of SIRT-1 that could potentially cause MS and other neurodegenerative disorders and provided data on existing available and effective drug therapies and disease management strategies.


Assuntos
Fatores Imunológicos/farmacologia , Esclerose Múltipla/metabolismo , Doenças Neurodegenerativas/metabolismo , Sirtuína 1/metabolismo , Animais , Humanos , Fatores Imunológicos/uso terapêutico , Esclerose Múltipla/genética , Esclerose Múltipla/imunologia , Doenças Neurodegenerativas/genética , Doenças Neurodegenerativas/imunologia , Neuroimunomodulação/efeitos dos fármacos , Neuroimunomodulação/fisiologia , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/fisiologia , Sirtuína 1/efeitos dos fármacos , Sirtuína 1/genética
6.
J Neuroimmunol ; 353: 577501, 2021 04 15.
Artigo em Inglês | MEDLINE | ID: mdl-33571815

RESUMO

The endocannabinoid system (ECS) is a complex physiological network involved in creating homeostasis and maintaining human health. Studies of the last 40 years have shown that endocannabinoids (ECs), a group of bioactive lipids, together with their set of receptors, function as one of the most important physiologic systems in human body. ECs and cannabinoid receptors (CBRs) are found throughout the body: in the brain tissues, immune cells, and in the peripheral organs and tissues as well. In recent years, ECs have emerged as key modulators of affect, neurotransmitter release, immune function, and several other physiological functions. This modulatory homoeostatic system operates in the regulation of brain activity and states of physical health and disease. In several research studies and patents the ECS has been recognised with neuro-protective properties thus it might be a target in neurodegenerative diseases. Most immune cells express these bioactive lipids and their receptors, recent data also highlight the immunomodulatory effects of endocannabinoids. Interplay of immune and nervous system has been recognized in past, recent studies suggest that ECS function as a bridge between neuronal and immune system. In several ongoing clinical trial studies, the ECS has also been placed in the anti-cancer drugs spotlight. This review summarizes the literature of cannabinoid ligands and their biosynthesis, cannabinoid receptors and their distribution, and the signaling pathways initiated by the binding of cannabinoid ligands to cannabinoid receptors. Further, this review highlights the functional role of cannabinoids and ECS in blood cell development, neuroimmune interactions and associated disorders. Moreover, we highlight the current state of knowledge of cannabinoid ligands as the mediators of neuroimmune interactions, which can be therapeutically effective for neuro-immune disorders and several diseases associated with neuroinflammation.


Assuntos
Endocanabinoides/fisiologia , Hematopoese/fisiologia , Neuroimunomodulação/fisiologia , Animais , Homeostase/fisiologia , Humanos , Receptores de Canabinoides/metabolismo
7.
Int J Mol Sci ; 22(3)2021 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-33535595

RESUMO

Pain can be induced by tissue injuries, diseases and infections. The interactions between the peripheral nervous system (PNS) and immune system are primary actions in pain sensitizations. In response to stimuli, nociceptors release various mediators from their terminals that potently activate and recruit immune cells, whereas infiltrated immune cells further promote sensitization of nociceptors and the transition from acute to chronic pain by producing cytokines, chemokines, lipid mediators and growth factors. Immune cells not only play roles in pain production but also contribute to PNS repair and pain resolution by secreting anti-inflammatory or analgesic effectors. Here, we discuss the distinct roles of four major types of immune cells (monocyte/macrophage, neutrophil, mast cell, and T cell) acting on the PNS during pain process. Integration of this current knowledge will enhance our understanding of cellular changes and molecular mechanisms underlying pain pathogenies, providing insights for developing new therapeutic strategies.


Assuntos
Dor Crônica/sangue , Dor Crônica/imunologia , Sistema Imunitário , Neuroimunomodulação/fisiologia , Sistema Nervoso Periférico/imunologia , Animais , Humanos , Inflamação , Macrófagos/citologia , Macrófagos/metabolismo , Mastócitos/citologia , Camundongos , Monócitos/citologia , Neutrófilos/citologia , Neutrófilos/metabolismo , Nociceptividade , Manejo da Dor , Sistema Nervoso Periférico/metabolismo , Transdução de Sinais , Linfócitos T/citologia
8.
J Neuroimmune Pharmacol ; 16(2): 454-469, 2021 06.
Artigo em Inglês | MEDLINE | ID: mdl-32648088

RESUMO

Deep brain stimulation of the subthalamic nucleus (DBS-STN) is an effective treatment for advanced motor symptoms of Parkinson's disease (PD). Recently, a connection between the limbic part of the STN and side effects of DBS-STN has been increasingly recognized. Animal studies have shown that DBS-STN influences behavior and provokes neurochemical changes in regions of the limbic system. Some of these regions, which are activated during DBS-STN, are involved in neuroimmunomodulation. The therapeutic effects of DBS-STN in PD treatment are clear, but the influence of DBS-STN on peripheral immunity has not been reported so far. In this study, we examined the effects of unilateral DBS-STN applied in male Wistar rats with 6-hydroxydopamine PD model (DBS-6OHDA) and rats without nigral dopamine depletion (DBS) on corticosterone (CORT) plasma concentration, blood natural killer cell cytotoxicity (NKCC), leukocyte numbers, lymphocyte population and apoptosis numbers, plasma interferon gamma (IFN-γ), interleukin 6 (IL-6), and tumor necrosis factor (TNF-α) concentration. The same peripheral immune parameters we measured also in non-stimulated rats with PD model (6OHDA). We observed peripheral immunity changes related to PD model. The NKCC and percentage of T cytotoxic lymphocytes were enhanced, while the level of lymphocyte apoptosis was down regulated in 6OHDA and DBS-6OHDA groups. After DBS-STN (DBS-6OHDA and DBS groups), the plasma CORT and TNF-α were elevated, the number of NK cells and percentage of apoptosis were increased, while the number of B lymphocytes was decreased. We also found, changes in plasma IFN-γ and IL-6 levels in all the groups. These results suggest potential peripheral immunomodulative effects of DBS-STN in the rat model of PD. However, further studies are necessary to explain these findings and their clinical implication. Graphical Abstract Influence of deep brain stimulation of the subthalamic nucleus on peripheral immunity in rat model of Parkinson's disease.


Assuntos
Corticosterona/sangue , Neuroimunomodulação/fisiologia , Transtornos Parkinsonianos/imunologia , Núcleo Subtalâmico/fisiologia , Animais , Estimulação Encefálica Profunda , Masculino , Ratos , Ratos Wistar
9.
Am J Psychiatry ; 178(4): 313-320, 2021 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-33207936

RESUMO

OBJECTIVE: Children exposed to severe, chronic stress are vulnerable to mental and physical health problems across the lifespan. To explain how these problems develop, the neuroimmune network hypothesis suggests that early-life stress initiates a positive feedback loop between peripheral inflammatory cells and networked brain regions involved in threat and reward processing. The authors sought to test this hypothesis by studying a sample of urban children from diverse socioeconomic backgrounds. METHODS: The authors examined the basic predictions of the neuroimmune network hypothesis in 207 children (mean age=13.9 years, 63% female; 33% Black; 30% Hispanic), focusing on poverty as a stressor. The children had fasting blood drawn to quantify five inflammatory biomarkers-C-reactive protein, tumor necrosis factor-α, and interleukins-6, -8, and -10-which were averaged to form a composite score. Children also completed two functional MRI tasks, which measured amygdala responsivity to angry facial expressions and ventral striatum responsivity to monetary rewards. RESULTS: Poverty status and neural responsivity interacted statistically to predict inflammation. Among children living in poverty, amygdala threat responsivity was positively associated with inflammation, and the same was true for ventral striatum responsivity to reward. As children's socioeconomic conditions improved, these brain-immune associations became weaker. In sensitivity analyses, these patterns were robust to alternative measures of socioeconomic status and were independent of age, sex, racial and ethnic identity, and pubertal status. The associations were also condition specific; no interactions were apparent for amygdala responsivity to neutral faces, or striatal responsivity to monetary losses. CONCLUSIONS: These findings suggest that childhood poverty is associated with accentuated neural-immune signaling, consistent with the neuroimmune network hypothesis.


Assuntos
Tonsila do Cerebelo/diagnóstico por imagem , Inflamação/imunologia , Neuroimunomodulação/imunologia , Pobreza , Recompensa , Estresse Psicológico/imunologia , Estresse Psicológico/fisiopatologia , Estriado Ventral/diagnóstico por imagem , Adolescente , Experiências Adversas da Infância , Tonsila do Cerebelo/fisiopatologia , Ira , Encéfalo/diagnóstico por imagem , Encéfalo/fisiopatologia , Proteína C-Reativa/imunologia , Expressão Facial , Retroalimentação Fisiológica , Feminino , Neuroimagem Funcional , Humanos , Interleucina-10/imunologia , Interleucina-6/imunologia , Interleucina-8/imunologia , Imageamento por Ressonância Magnética , Masculino , Neuroimunomodulação/fisiologia , Fator de Necrose Tumoral alfa/imunologia , Estriado Ventral/fisiopatologia
10.
Life Sci ; 267: 118958, 2021 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-33383054

RESUMO

AIMS: Spinal cord injury (SCI) is a major cause of long-term physical impairment. Currently, treatment for SCI is limited to supportive measures, which can lead to permanent disability, representing a serious social burden. The present study aimed to evaluate the inflammatory microenvironment effects of human umbilical cord mesenchymal stem cells (HUCMSCs)+ Ultrashort Wave (USW) therapy on SCI and reveal possible mechanisms. MAIN METHODS: Low-dose USW was treated one day after SCI, and HUCMSCs suspension was transferred to the lesion using a micro-syringe 7 days after SCI. The functional effects of HUCMSCs and USW, separately and combinedly, were measured, together with the infiltration of CD3+ cells, formation of A1 astrocytes and activation of NUR77/ NF-κB pathway. KEY FINDINGS: Our results showed that HUCMSCs+USW therapy improved motor function of SCI rat, together with decreased infiltration of CD3+ T cells, and decreased induction of microglia and A1 astrocytes. And also USW treatment played a very important role on decreasing the infiltration of CD3+ T cells and IBA-1+ cells. Reduced production of pro-inflammatory cytokines IL-1ß and IL-6 was also observed in rats receiving HUCMSCs+USW therapy, medicated by NUR77/NF-κB pathway. SIGNIFICANCE: These findings indicated that HUCMSCs+USW therapy could attenuate inflammatory microenvironment through NUR77/NF-κB signaling pathway, which might contribute to its better outcome.


Assuntos
Transplante de Células-Tronco de Sangue do Cordão Umbilical/métodos , Células-Tronco Mesenquimais/efeitos da radiação , Traumatismos da Medula Espinal/terapia , Animais , Astrócitos/metabolismo , Citocinas/metabolismo , Feminino , Inflamação/patologia , Células-Tronco Mesenquimais/metabolismo , Células-Tronco Mesenquimais/fisiologia , Microglia/metabolismo , NF-kappa B/metabolismo , Neuroimunomodulação/fisiologia , Estresse Oxidativo/efeitos dos fármacos , Ratos , Ratos Sprague-Dawley , Terapia por Ondas Curtas/métodos , Transdução de Sinais/efeitos dos fármacos , Medula Espinal/metabolismo , Traumatismos da Medula Espinal/metabolismo , Cordão Umbilical
11.
J Clin Endocrinol Metab ; 106(2): e415-e429, 2021 01 23.
Artigo em Inglês | MEDLINE | ID: mdl-33104773

RESUMO

CONTEXT: The relationship between the endocrine system and autoimmunity has been recognized for a long time and one of the best examples of autoimmune endocrine disease is autoimmune hypophysitis. A better understanding of autoimmune mechanisms and radiological, biochemical, and immunological developments has given rise to the definition of new autoimmune disorders including autoimmunity-related hypothalamic-pituitary disorders. However, whether hypothalamitis may occur as a distinct entity is still a matter of debate. EVIDENCE ACQUISITION: Here we describe a 35-year-old woman with growing suprasellar mass, partial empty sella, central diabetes insipidus, hypopituitarism, and hyperprolactinemia. EVIDENCE SYNTHESIS: Histopathologic examination of surgically removed suprasellar mass revealed lymphocytic infiltrate suggestive of an autoimmune disease with hypothalamic involvement. The presence of antihypothalamus antibodies to arginine vasopressin (AVP)-secreting cells (AVPcAb) at high titers and the absence of antipituitary antibodies suggested the diagnosis of isolated hypothalamitis. Some similar conditions have sometimes been reported in the literature but the simultaneous double finding of lymphocytic infiltrate and the presence of AVPcAb so far has never been reported. CONCLUSIONS: We think that the hypothalamitis can be considered a new isolated autoimmune disease affecting the hypothalamus while the lymphocytic infundibuloneurohypophysitis can be a consequence of hypothalamitis with subsequent autoimmune involvement of the pituitary. To our knowledge this is the first observation of autoimmune hypothalamic involvement with central diabetes insipidus, partial empty sella, antihypothalamic antibodies and hypopituitarism.


Assuntos
Encefalite/diagnóstico , Doenças Hipotalâmicas/diagnóstico , Adulto , Doenças Autoimunes/classificação , Doenças Autoimunes/diagnóstico , Diabetes Insípido Neurogênico/diagnóstico , Diabetes Insípido Neurogênico/etiologia , Diagnóstico Diferencial , Encefalite/complicações , Doenças do Sistema Endócrino/classificação , Doenças do Sistema Endócrino/diagnóstico , Feminino , Humanos , Hiperprolactinemia/diagnóstico , Hiperprolactinemia/etiologia , Hipopituitarismo/diagnóstico , Hipopituitarismo/etiologia , Doenças Hipotalâmicas/complicações , Neuroimunomodulação/fisiologia , Sela Túrcica/patologia
12.
J Neurosci ; 41(5): 855-865, 2021 02 03.
Artigo em Inglês | MEDLINE | ID: mdl-33239404

RESUMO

Chronic pain, encompassing conditions, such as low back pain, arthritis, persistent post-surgical pain, fibromyalgia, and neuropathic pain disorders, is highly prevalent but remains poorly treated. The vast majority of therapeutics are directed solely at neurons, despite the fact that signaling between immune cells, glia, and neurons is now recognized as indispensable for the initiation and maintenance of chronic pain. This review highlights recent advances in understanding fundamental neuroimmune signaling mechanisms and novel therapeutic targets in rodent models of chronic pain. We further discuss new technological developments to study, diagnose, and quantify neuroimmune contributions to chronic pain in patient populations.


Assuntos
Autoanticorpos/imunologia , Dor Crônica/imunologia , Modelos Animais de Doenças , Neuroglia/imunologia , Neuroimunomodulação/fisiologia , Neurônios/imunologia , Animais , Autoanticorpos/metabolismo , Dor Crônica/metabolismo , Humanos , Fator 2 Relacionado a NF-E2/imunologia , Fator 2 Relacionado a NF-E2/metabolismo , Neuroglia/metabolismo , Neurônios/metabolismo , Roedores
13.
PLoS Biol ; 18(12): e3001008, 2020 12.
Artigo em Inglês | MEDLINE | ID: mdl-33315860

RESUMO

Changes to the structure of nodes of Ranvier in the normal-appearing white matter (NAWM) of multiple sclerosis (MS) brains are associated with chronic inflammation. We show that the paranodal domains in MS NAWM are longer on average than control, with Kv1.2 channels dislocated into the paranode. These pathological features are reproduced in a model of chronic meningeal inflammation generated by the injection of lentiviral vectors for the lymphotoxin-α (LTα) and interferon-γ (IFNγ) genes. We show that tumour necrosis factor (TNF), IFNγ, and glutamate can provoke paranodal elongation in cerebellar slice cultures, which could be reversed by an N-methyl-D-aspartate (NMDA) receptor blocker. When these changes were inserted into a computational model to simulate axonal conduction, a rapid decrease in velocity was observed, reaching conduction failure in small diameter axons. We suggest that glial cells activated by pro-inflammatory cytokines can produce high levels of glutamate, which triggers paranodal pathology, contributing to axonal damage and conduction deficits.


Assuntos
Esclerose Múltipla/patologia , Nós Neurofibrosos/patologia , Substância Branca/patologia , Adulto , Idoso , Idoso de 80 Anos ou mais , Axônios/patologia , Encéfalo/patologia , Sinapses Elétricas/patologia , Sinapses Elétricas/efeitos da radiação , Feminino , Humanos , Inflamação/patologia , Masculino , Microglia/patologia , Pessoa de Meia-Idade , Esclerose Múltipla/diagnóstico por imagem , Bainha de Mielina/patologia , Neuroglia/patologia , Neuroimunomodulação/imunologia , Neuroimunomodulação/fisiologia , Nós Neurofibrosos/fisiologia , Substância Branca/diagnóstico por imagem , Substância Branca/imunologia
14.
J Neuroinflammation ; 17(1): 368, 2020 Dec 02.
Artigo em Inglês | MEDLINE | ID: mdl-33267881

RESUMO

BACKGROUND: The carotid bodies and baroreceptors are sensors capable of detecting various physiological parameters that signal to the brain via the afferent carotid sinus nerve for physiological adjustment by efferent pathways. Because receptors for inflammatory mediators are expressed by these sensors, we and others have hypothesised they could detect changes in pro-inflammatory cytokine blood levels and eventually trigger an anti-inflammatory reflex. METHODS: To test this hypothesis, we surgically isolated the carotid sinus nerve and implanted an electrode, which could deliver an electrical stimulation package prior and following a lipopolysaccharide injection. Subsequently, 90 min later, blood was extracted, and cytokine levels were analysed. RESULTS: Here, we found that carotid sinus nerve electrical stimulation inhibited lipopolysaccharide-induced tumour necrosis factor production in both anaesthetised and non-anaesthetised conscious mice. The anti-inflammatory effect of carotid sinus nerve electrical stimulation was so potent that it protected conscious mice from endotoxaemic shock-induced death. In contrast to the mechanisms underlying the well-described vagal anti-inflammatory reflex, this phenomenon does not depend on signalling through the autonomic nervous system. Rather, the inhibition of lipopolysaccharide-induced tumour necrosis factor production by carotid sinus nerve electrical stimulation is abolished by surgical removal of the adrenal glands, by treatment with the glucocorticoid receptor antagonist mifepristone or by genetic inactivation of the glucocorticoid gene in myeloid cells. Further, carotid sinus nerve electrical stimulation increases the spontaneous discharge activity of the hypothalamic paraventricular nucleus leading to enhanced production of corticosterone. CONCLUSION: Carotid sinus nerve electrostimulation attenuates inflammation and protects against lipopolysaccharide-induced endotoxaemic shock via increased corticosterone acting on the glucocorticoid receptor of myeloid immune cells. These results provide a rationale for the use of carotid sinus nerve electrostimulation as a therapeutic approach for immune-mediated inflammatory diseases.


Assuntos
Seio Carotídeo/fisiologia , Inflamação/metabolismo , Células Mieloides/metabolismo , Neuroimunomodulação/fisiologia , Animais , Seio Carotídeo/inervação , Estimulação Elétrica , Feminino , Inflamação/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Células Mieloides/imunologia , Receptores de Glucocorticoides
15.
Neuroreport ; 31(15): 1084-1089, 2020 10 14.
Artigo em Inglês | MEDLINE | ID: mdl-32881777

RESUMO

Cervical nerve root injury induces a host of inflammatory mediators in the spinal cord that initiate and maintain neuronal hyperexcitability and pain. Secretory phospholipase A2 (sPLA2) is an enzyme that has been implicated as a mediator of pain onset and maintenance in inflammation and neural injury. Although sPLA2 modulates nociception and excitatory neuronal signaling in vitro, its effects on neuronal activity and central sensitization early after painful nerve root injury are unknown. This study investigated whether inhibiting spinal sPLA2 at the time of nerve root compression (NRC) modulates the pain, dorsal horn hyperexcitability, and spinal genes involved in glutamate signaling, nociception, and inflammation that are seen early after injury. Rats underwent a painful C7 NRC injury with immediate intrathecal administration of the sPLA2 inhibitor thioetheramide-phosphorlycholine. Additional groups underwent either injury alone or sham surgery. One day after injury, behavioral sensitivity, spinal neuronal excitability, and spinal cord gene expression for glutamate receptors (mGluR5 and NR1) and transporters (GLT1 and EAAC1), the neuropeptide substance P, and pro-inflammatory cytokines (TNFα, IL1α, and IL1ß) were assessed. Treatment with the sPLA2 inhibitor prevented mechanical allodynia, attenuated neuronal hyperexcitability in the spinal dorsal horn, restored the proportion of spinal neurons classified as wide dynamic range, and reduced genes for mGluR5, substance P, IL1α, and IL1ß to sham levels. These findings indicate spinal regulation of central sensitization after painful neuropathy and suggest that spinal sPLA2 is implicated in those early spinal mechanisms of neuronal excitability, perhaps via glutamate signaling, neurotransmitters, or inflammatory cascades.


Assuntos
Genes Reguladores/fisiologia , Síndromes de Compressão Nervosa/enzimologia , Neuroimunomodulação/fisiologia , Fosfolipases A2 Secretórias/antagonistas & inibidores , Fosfolipases A2 Secretórias/metabolismo , Raízes Nervosas Espinhais/enzimologia , Animais , Genes Reguladores/efeitos dos fármacos , Injeções Espinhais , Masculino , Síndromes de Compressão Nervosa/tratamento farmacológico , Síndromes de Compressão Nervosa/genética , Neuroimunomodulação/efeitos dos fármacos , Dor/tratamento farmacológico , Dor/enzimologia , Dor/genética , Doenças do Sistema Nervoso Periférico/tratamento farmacológico , Doenças do Sistema Nervoso Periférico/enzimologia , Doenças do Sistema Nervoso Periférico/genética , Fosfatidilcolinas/administração & dosagem , Radiculopatia/tratamento farmacológico , Radiculopatia/enzimologia , Radiculopatia/genética , Ratos , Ratos Sprague-Dawley , Raízes Nervosas Espinhais/efeitos dos fármacos
16.
J Neuroimmunol ; 348: 577382, 2020 11 15.
Artigo em Inglês | MEDLINE | ID: mdl-32919148

RESUMO

The cells of the immune and neuronal systems share different receptors for cytokines or neurotransmitters, producing feedback responses between both systems. Cytokines such as IL-1ß and TNF-α can induce inflammation; however, the secretion of these molecules can be modulated by anti-inflammatory cytokines, as is the case for TGF-ß, as well as by different hormones or neurotransmitters such as the γ-aminobutyric acid (GABA). In this study, we evaluated the secretion of IL-1ß, TNF-α, and TGF-ß under basal conditions, in the head of the kidney, spleen, thymus, and serum of the Nile tilapia, as well as their release induced by different sub-basal increases of GABA. We found that at the higher dose of GABA these cytokines were synthesised at a higher concentration compared to the control group. These results may suggest that there is feedback between both systems and that GABA plays a role in the modulation of the immune response.


Assuntos
Ciclídeos/imunologia , Interleucina-1beta/biossíntese , Tecido Linfoide/metabolismo , Fator de Crescimento Transformador beta/biossíntese , Fator de Necrose Tumoral alfa/biossíntese , Ácido gama-Aminobutírico/metabolismo , Animais , Proteínas de Peixes/imunologia , Proteínas de Peixes/metabolismo , Neuroimunomodulação/fisiologia
17.
Am J Ther ; 27(5): e468-e476, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32897982

RESUMO

BACKGROUND: Pain and depression have a high impact on caring for the people who need palliative care, but both of these are neglected compared with the approach for other symptoms encountered by these patients. AREAS OF UNCERTAINTY: There are few studies in humans that support the existence of common neural circuits between depression and pain that also explore the use of drugs with effects in both conditions. More knowledge is needed about the relationship of these clinical entities that will lead to the optimization of the treatment and improvement of quality of life. DATA SOURCES: We conducted a search in PubMed to identify relevant articles and reviews that have been published in the last 5 years, concerning the topic of common pathways between depression and pain (2014-April 2019). THERAPEUTIC ADVANCES: The connections between the 2 clinical entities start at the level of the cortical regions. The hippocampus is the main site of neural changes, modification of the immune system, neuromodulators, neurotransmitters, and signaling pathways implicated in both conditions. Increased levels of peripheral proinflammatory cytokines and neuroinflammatory changes are related to the physiopathology of these entities. Inflammation links depression and pain by altering neural circuits and changes in their common cortical regions. Antidepressants are used to treat depression and chronic, pain but more experimental studies are needed to determine which antidepressant drugs are the most effective in treating the 2 entities. CONCLUSIONS: Pharmacological and nonpharmacological interventions targeting cortical changes in pain and depression are promising, but more clinical studies are needed to validate their usefulness.


Assuntos
Antidepressivos/administração & dosagem , Dor Crônica/terapia , Depressão/terapia , Neuralgia/terapia , Cuidados Paliativos/métodos , Dor Crônica/complicações , Dor Crônica/fisiopatologia , Dor Crônica/psicologia , Depressão/complicações , Depressão/fisiopatologia , Depressão/psicologia , Hipocampo/efeitos dos fármacos , Hipocampo/patologia , Hipocampo/fisiopatologia , Humanos , Mediadores da Inflamação/metabolismo , Rede Nervosa/efeitos dos fármacos , Rede Nervosa/fisiopatologia , Neuralgia/complicações , Neuralgia/fisiopatologia , Neuralgia/psicologia , Neuroimunomodulação/efeitos dos fármacos , Neuroimunomodulação/fisiologia , Neuropeptídeos/metabolismo , Qualidade de Vida , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/imunologia
18.
Int J Mol Sci ; 21(18)2020 Sep 05.
Artigo em Inglês | MEDLINE | ID: mdl-32899565

RESUMO

We studied 114 primitive cerebral neoplasia, that were surgically treated, and underwent radiotherapy (RT), and compared their results to those obtained by 190 patients diagnosed with subcortical vascular dementia (sVAD). Patients with any form of primitive cerebral neoplasia underwent whole-brain radiotherapy. All the tumor patients had regional field partial brain RT, which encompassed each tumor, with an average margin of 2.6 cm from the initial target tumor volume. We observed in our patients who have been exposed to a higher dose of RT (30-65 Gy) a cognitive and behavior decline similar to that observed in sVAD, with the frontal dysexecutive syndrome, apathy, and gait alterations, but with a more rapid onset and with an overwhelming effect. Multiple mechanisms are likely to be involved in radiation-induced cognitive impairment. The active site of RT brain damage is the white matter areas, particularly the internal capsule, basal ganglia, caudate, hippocampus, and subventricular zone. In all cases, radiation damage inside the brain mainly focuses on the cortical-subcortical frontal loops, which integrate and process the flow of information from the cortical areas, where executive functions are "elaborated" and prepared, towards the thalamus, subthalamus, and cerebellum, where they are continuously refined and executed. The active mechanisms that RT drives are similar to those observed in cerebral small vessel disease (SVD), leading to sVAD. The RT's primary targets, outside the tumor mass, are the blood-brain barrier (BBB), the small vessels, and putative mechanisms that can be taken into account are oxidative stress and neuro-inflammation, strongly associated with the alteration of NMDA receptor subunit composition.


Assuntos
Encefalopatias/patologia , Disfunção Cognitiva/patologia , Porencefalia/patologia , Adulto , Barreira Hematoencefálica/patologia , Encéfalo/patologia , Córtex Cerebral/patologia , Doenças de Pequenos Vasos Cerebrais/complicações , Doenças de Pequenos Vasos Cerebrais/patologia , Demência Vascular/patologia , Feminino , Humanos , Doença Iatrogênica/prevenção & controle , Masculino , Pessoa de Meia-Idade , Neuroimunomodulação/fisiologia , Estresse Oxidativo/fisiologia , Porencefalia/etiologia , Radioterapia/efeitos adversos , Substância Branca/patologia
19.
Neurotherapeutics ; 17(3): 886-899, 2020 07.
Artigo em Inglês | MEDLINE | ID: mdl-32696274

RESUMO

The current crises in opioid abuse and chronic pain call for the development of nonopioid and nonpharmacological therapeutics for pain relief. Neuromodulation-based approaches, such as spinal cord stimulation, dorsal root ganglion simulation, and nerve stimulation including vagus nerve stimulation, have shown efficacy in achieving pain control in preclinical and clinical studies. However, the mechanisms by which neuromodulation alleviates pain are not fully understood. Accumulating evidence suggests that neuromodulation regulates inflammation and neuroinflammation-a localized inflammation in peripheral nerves, dorsal root ganglia/trigeminal ganglia, and spinal cord/brain-through neuro-immune interactions. Specialized proresolving mediators (SPMs) such as resolvins, protectins, maresins, and lipoxins are lipid molecules produced during the resolution phase of inflammation and exhibit multiple beneficial effects in resolving inflammation in various animal models. Recent studies suggest that SPMs inhibit inflammatory pain, postoperative pain, neuropathic pain, and cancer pain in rodent models via immune, glial, and neuronal modulations. It is noteworthy that sham surgery is sufficient to elevate resolvin levels and may serve as a model of resolution. Interestingly, it has been shown that the vagus nerve produces SPMs and vagus nerve stimulation (VNS) induces SPM production in vitro. In this review, we discuss how neuromodulation such as VNS controls pain via immunomodulation and neuro-immune interactions and highlight possible involvement of SPMs. In particular, we demonstrate that VNS via auricular electroacupuncture effectively attenuates chemotherapy-induced neuropathic pain. Furthermore, auricular stimulation is able to increase resolvin levels in mice. Thus, we propose that neuromodulation may control pain and inflammation/neuroinflammatioin via SPMs. Finally, we discuss key questions that remain unanswered in our understanding of how neuromodulation-based therapies provide short-term and long-term pain relief.


Assuntos
Mediadores da Inflamação/antagonistas & inibidores , Mediadores da Inflamação/metabolismo , Neuroimunomodulação/fisiologia , Manejo da Dor/métodos , Dor/metabolismo , Analgésicos/farmacologia , Analgésicos/uso terapêutico , Animais , Ácidos Docosa-Hexaenoicos/metabolismo , Ácidos Docosa-Hexaenoicos/uso terapêutico , Humanos , Metabolismo dos Lipídeos/efeitos dos fármacos , Metabolismo dos Lipídeos/fisiologia , Lipoxinas/metabolismo , Lipoxinas/uso terapêutico , Neuroimunomodulação/efeitos dos fármacos , Estimulação do Nervo Vago/métodos
20.
Expert Opin Biol Ther ; 20(12): 1461-1475, 2020 12.
Artigo em Inglês | MEDLINE | ID: mdl-32700610

RESUMO

INTRODUCTION: A large volume of data indicates that the known thymic hormones, thymulin, thymopoietin, thymosin-α, thymosin-ß, and thymic humoral factor-y2, exhibit different spectra of activities. Although large in volume, available data are rather fragmented, resulting in a lack of understanding of the role played by thymic hormones in immune homeostasis. AREA COVERED: Existing data compartmentalizes the effect of thymic peptides into 2 categories: influence on immune cells and interconnection with neuroendocrine systems. The current study draws attention to a third aspect of the thymic peptide effect that has not been clarified yet, wherein ubiquitous and highly abundant intranuclear precursors of so called 'thymic peptides' play a fundamental role in all somatic cells. EXPERT OPINION: Our analysis indicated that, under certain stress-related conditions, these precursors are cleaved to form immunologically active peptides that rapidly leave the nucleus and intracellular spaces, to send 'distress signals' to the immune system, thereby acting as stress sensors. We propose that these peptides may form a link between somatic cells and immune as well as neuroendocrine systems. This model may provide a better understanding of the mechanisms underlying immune homeostasis, leading thereby to the development of new therapeutic regimes utilizing the characteristics of thymic peptides.


Assuntos
Fragmentos de Peptídeos/fisiologia , Precursores de Proteínas/fisiologia , Estresse Fisiológico/imunologia , Timo/metabolismo , Hormônios do Timo/fisiologia , Animais , Homeostase/imunologia , Humanos , Neuroimunomodulação/fisiologia , Fragmentos de Peptídeos/metabolismo , Hormônios Peptídicos/metabolismo , Hormônios Peptídicos/fisiologia , Precursores de Proteínas/metabolismo , Hormônios do Timo/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA