Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Sensors (Basel) ; 23(7)2023 Mar 28.
Artículo en Inglés | MEDLINE | ID: mdl-37050605

RESUMEN

Research in the field of gathering and analyzing biological signals is growing. The sensors are becoming more available and more non-invasive for examining such signals, which in the past required the inconvenient acquisition of data. This was achieved mainly by the fact that biological sensors were able to be built into wearable and portable devices. The representation and analysis of EEGs (electroencephalograms) is nowadays commonly used in various application areas. The application of the use of the EEG signals to the field of automation is still an unexplored area and therefore provides opportunities for interesting research. In our research, we focused on the area of processing automation; especially the use of the EEG signals to bridge the communication between control of individual processes and a human. In this study, the real-time communication between a PLC (programmable logic controller) and BCI (brain computer interface) was investigated and described. In the future, this approach can help people with physical disabilities to control certain machines or devices and therefore it could find applicability in overcoming physical disabilities. The main contribution of the article is, that we have demonstrated the possibility of interaction between a person and a manipulator controlled by a PLC with the help of a BCI. Potentially, with the expansion of functionality, such solutions will allow a person with physical disabilities to participate in the production process.


Asunto(s)
Interfaces Cerebro-Computador , Personas con Discapacidad , Humanos , Electroencefalografía , Automatización , Lógica
2.
Neurogastroenterol Motil ; 26(9): 1238-47, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-24966010

RESUMEN

BACKGROUND: Postoperative ileus (POI) is characterized by impaired gastrointestinal motility resulting from intestinal handling-associated inflammation. The introduction of laparoscopic surgery has dramatically reduced the duration of POI. However, it remains unclear to what extent this results in a reduction of intestinal inflammation. The aim of the present study is to compare the degree of intestinal inflammation and gastrointestinal transit following laparoscopic surgery and open abdominal surgery. METHODS: Mice were subjected to laparoscopic surgery or laparotomy alone or, in combination with standardized intestinal manipulation of the small bowel (IM). Gastrointestinal transit and intestinal inflammation were assessed 24 h after surgery by the number of myeloperoxidase (MPO) positive cells and the level of cytokine expression. The recovery time and the degree of inflammation were also analyzed in patients subjected to colectomy under open conditions (laparotomy) or laparoscopic conditions. KEY RESULTS: Mice undergoing IM by laparotomy (open IM), but not by laparoscopy (Lap IM) developed a significant delay in gastrointestinal transit compared to laparotomy or laparoscopy alone. In addition, there was significant intestinal inflammation only after open IM. Similarly, cytokine levels in peritoneal lavage fluid were lower while recovery time was faster in patients subjected to colectomy under laparoscopic conditions compared to open colectomy. CONCLUSIONS & INFERENCES: Our data confirms that intestinal inflammation is underlying the delayed gastrointestinal transit observed after open surgery. Most importantly, we demonstrate that intestinal inflammation under laparoscopic conditions is significantly lower compared to open surgery, most likely explaining the faster recovery following laparoscopic surgery.


Asunto(s)
Enteritis/etiología , Ileus/etiología , Enfermedades del Yeyuno/etiología , Laparoscopía/efectos adversos , Animales , Modelos Animales de Enfermedad , Enteritis/metabolismo , Femenino , Tránsito Gastrointestinal , Humanos , Interleucinas/metabolismo , Enfermedades del Yeyuno/metabolismo , Ratones , Ratones Endogámicos C57BL
3.
PLoS One ; 9(1): e87785, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24489965

RESUMEN

BACKGROUND: Electrical stimulation of the vagus nerve suppresses intestinal inflammation and normalizes gut motility in a mouse model of postoperative ileus. The exact anatomical interaction between the vagus nerve and the intestinal immune system remains however a matter of debate. In the present study, we provide additional evidence on the direct and indirect vagal innervation of the spleen and analyzed the anatomical evidence for neuroimmune modulation of macrophages by vagal preganglionic and enteric postganglionic nerve fibers within the intestine. METHODS: Dextran conjugates were used to label vagal preganglionic (motor) fibers projecting to the small intestine and spleen. Moreover, identification of the neurochemical phenotype of the vagal efferent fibers and enteric neurons was performed by immunofluorescent labeling. F4/80 antibody was used to label resident macrophages. RESULTS: Our anterograde tracing experiments did not reveal dextran-labeled vagal fibers or terminals in the mesenteric ganglion or spleen. Vagal efferent fibers were confined within the myenteric plexus region of the small intestine and mainly endings around nNOS, VIP and ChAT positive enteric neurons. nNOS, VIP and ChAT positive fibers were found in close proximity of intestinal resident macrophages carrying α7 nicotinic receptors. Of note, VIP receptors were found on resident macrophages located in close proximity of VIP positive nerve fibers. CONCLUSION: In the present study, we show that the vagus nerve does not directly interact with resident macrophages in the gut or spleen. Instead, the vagus nerve preferentially interacts with nNOS, VIP and ChAT enteric neurons located within the gut muscularis with nerve endings in close proximity of the resident macrophages.


Asunto(s)
Intestino Delgado/inervación , Macrófagos/fisiología , Bazo/inervación , Nervio Vago/fisiología , Acetilcolina/metabolismo , Animales , Vías Eferentes , Femenino , Intestino Delgado/citología , Intestino Delgado/metabolismo , Ganglios Linfáticos/citología , Ratones , Ratones Endogámicos BALB C , Plexo Mientérico/citología , Plexo Mientérico/fisiología , Cuello , Factores de Crecimiento Nervioso/metabolismo , Óxido Nítrico Sintasa de Tipo I/metabolismo , Bazo/citología , Receptor Nicotínico de Acetilcolina alfa 7/metabolismo
4.
PLoS One ; 9(1): e85304, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24416383

RESUMEN

INTRODUCTION: Intestinal manipulation (IM) during abdominal surgery results in intestinal inflammation leading to hypomotility or ileus. Mast cell activation is thought to play a crucial role in the pathophysiology of postoperative ileus (POI). However, this conclusion was mainly drawn using mast cell-deficient mouse models with abnormal Kit signaling. These mice also lack interstitial cells of Cajal (ICC) resulting in aberrant gastrointestinal motility even prior to surgery, compromising their use as model to study POI. To avoid these experimental weaknesses we took advantage of a newly developed knock-in mouse model, Cpa3(Cre/+) , devoid of mast cells but with intact Kit signaling. DESIGN: The role of mast cells in the development of POI and intestinal inflammation was evaluated assessing gastrointestinal transit and muscularis externa inflammation after IM in two strains of mice lacking mast cells, i.e. Kit(W-sh/W-sh) and Cpa3(Cre/+) mice, and by use of the mast cell stabilizer cromolyn. RESULTS: Kit(W-sh/W-sh) mice lack ICC networks and already revealed significantly delayed gastrointestinal transit even before surgery. IM did not further delay intestinal transit, but induced infiltration of myeloperoxidase positive cells, expression of inflammatory cytokines and recruitment of monocytes and neutrophils into the muscularis externa. On the contrary, Cpa3(Cre/+) mice have a normal network of ICC and normal gastrointestinal. Surprisingly, IM in Cpa3(Cre/+) mice caused delay in gut motility and intestinal inflammation as in wild type littermates mice (Cpa3(+/+) ). Furthermore, treatment with the mast cell inhibitor cromolyn resulted in an inhibition of mast cells without preventing POI. CONCLUSIONS: Here, we confirm that IM induced mast cell degranulation. However, our data demonstrate that mast cells are not required for the pathogenesis of POI in mice. Although there might be species differences between mouse and human, our results argue against mast cell inhibitors as a therapeutic approach to shorten POI.


Asunto(s)
Procedimientos Quirúrgicos del Sistema Digestivo/efectos adversos , Tránsito Gastrointestinal , Ileus/patología , Mastocitos/citología , Complicaciones Posoperatorias/patología , Animales , Carboxipeptidasas A/genética , Carboxipeptidasas A/metabolismo , Degranulación de la Célula , Movimiento Celular , Cromolin Sódico/farmacología , Modelos Animales de Enfermedad , Expresión Génica , Humanos , Ileus/metabolismo , Inflamación/etiología , Inflamación/metabolismo , Inflamación/patología , Células Intersticiales de Cajal/metabolismo , Células Intersticiales de Cajal/patología , Mastocitos/efectos de los fármacos , Mastocitos/metabolismo , Ratones , Ratones Transgénicos , Monocitos/metabolismo , Monocitos/patología , Neutrófilos/metabolismo , Neutrófilos/patología , Complicaciones Posoperatorias/etiología , Complicaciones Posoperatorias/metabolismo , Proteínas Proto-Oncogénicas c-kit/genética , Proteínas Proto-Oncogénicas c-kit/metabolismo , Especificidad de la Especie
5.
Gut ; 63(6): 938-48, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23929694

RESUMEN

The cholinergic anti-inflammatory pathway (CAIP) has been proposed as a key mechanism by which the brain, through the vagus nerve, modulates the immune system in the spleen. Vagus nerve stimulation (VNS) reduces intestinal inflammation and improves postoperative ileus. We investigated the neural pathway involved and the cells mediating the anti-inflammatory effect of VNS in the gut. The effect of VNS on intestinal inflammation and transit was investigated in wild-type, splenic denervated and Rag-1 knockout mice. To define the possible role of α7 nicotinic acetylcholine receptor (α7nAChR), we used knockout and bone marrow chimaera mice. Anterograde tracing of vagal efferents, cell sorting and Ca(2+) imaging were used to reveal the intestinal cells targeted by the vagus nerve. VNS attenuates surgery-induced intestinal inflammation and improves postoperative intestinal transit in wild-type, splenic denervated and T-cell-deficient mice. In contrast, VNS is ineffective in α7nAChR knockout mice and α7nAChR-deficient bone marrow chimaera mice. Anterograde labelling fails to detect vagal efferents contacting resident macrophages, but shows close contacts between cholinergic myenteric neurons and resident macrophages expressing α7nAChR. Finally, α7nAChR activation modulates ATP-induced Ca(2+) response in small intestine resident macrophages. We show that the anti-inflammatory effect of the VNS in the intestine is independent of the spleen and T cells. Instead, the vagus nerve interacts with cholinergic myenteric neurons in close contact with the muscularis macrophages. Our data suggest that intestinal muscularis resident macrophages expressing α7nAChR are most likely the ultimate target of the gastrointestinal CAIP.


Asunto(s)
Macrófagos/metabolismo , Músculo Liso/citología , Estimulación del Nervio Vago , Nervio Vago/metabolismo , Receptor Nicotínico de Acetilcolina alfa 7/metabolismo , Animales , Desnervación Autonómica , Citocinas/genética , Enteritis/metabolismo , Tránsito Gastrointestinal , Expresión Génica , Macrófagos/citología , Ratones , Ratones Noqueados , Plexo Mientérico/metabolismo , Neuronas/metabolismo , Nicotina/farmacología , Peroxidasa/metabolismo , Transducción de Señal , Bazo/inervación , Receptor Nicotínico de Acetilcolina alfa 7/agonistas , Receptor Nicotínico de Acetilcolina alfa 7/genética
6.
PLoS One ; 8(11): e79264, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24223920

RESUMEN

BACKGROUND: The cholinergic anti-inflammatory pathway is an endogenous mechanism by which the autonomic nervous system attenuates macrophage activation via nicotinic acetylcholine receptors (nAChR). This concept has however not been demonstrated at a cellular level in intact tissue. To this end, we have studied the effect of nicotine on the activation of resident macrophages in a mouse stomach preparation by means of calcium imaging. METHODS: Calcium transients ([Ca(2+)]i) in resident macrophages were recorded in a mouse stomach preparation containing myenteric plexus and muscle layers by Fluo-4. Activation of macrophages was achieved by focal puff administration of ATP. The effects of nicotine on activation of macrophages were evaluated and the nAChR involved was pharmacologically characterized. The proximity of cholinergic nerves to macrophages was quantified by confocal microscopy. Expression of ß2 and α7 nAChR was evaluated by ß2 immunohistochemistry and fluorophore-tagged α-bungarotoxin. RESULTS: In 83% of macrophages cholinergic varicose nerve fibers were detected at distances <900 nm. The ATP induced [Ca(2+)]i increase was significantly inhibited in 65% or 55% of macrophages by 100 µM or 10 µM nicotine, respectively. This inhibitory effect was reversed by the ß2 nAChR preferring antagonist dihydro-ß-eryhtroidine but not by hexamethonium (non-selective nAChR-antagonist), mecamylamine (α3ß4 nAChR-preferring antagonist), α-bungarotoxin or methyllycaconitine (both α7 nAChR-preferring antagonist). Macrophages in the stomach express ß2 but not α7 nAChR at protein level, while those in the intestine express both receptor subunits. CONCLUSION: This study is the first in situ demonstration of an inhibition of macrophage activation by nicotine suggesting functional signaling between cholinergic neurons and macrophages in the stomach. The data suggest that the ß2 subunit of the nAChR is critically involved in the nicotine-induced inhibition of these resident macrophages.


Asunto(s)
Macrófagos/efectos de los fármacos , Nicotina/farmacología , Receptores Nicotínicos/metabolismo , Estómago/efectos de los fármacos , Adenosina Trifosfato/farmacología , Compuestos de Anilina/metabolismo , Animales , Calcio/metabolismo , Sistema Nervioso Entérico/citología , Sistema Nervioso Entérico/efectos de los fármacos , Sistema Nervioso Entérico/metabolismo , Estimulantes Ganglionares/farmacología , Mucosa Gástrica/metabolismo , Inmunohistoquímica , Activación de Macrófagos/efectos de los fármacos , Macrófagos/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Microscopía Confocal , Plexo Mientérico/efectos de los fármacos , Plexo Mientérico/metabolismo , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Estómago/inervación , Xantenos/metabolismo
7.
Gut ; 62(11): 1581-90, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-23242119

RESUMEN

OBJECTIVE: Intestinal inflammation resulting from manipulation-induced mast cell activation is a crucial mechanism in the pathophysiology of postoperative ileus (POI). Recently it has been shown that spleen tyrosine kinase (Syk) is involved in mast cell degranulation. Therefore, we have evaluated the effect of the Syk-inhibitor GSK compound 143 (GSK143) as potential treatment to shorten POI. DESIGN: In vivo: in a mouse model of POI, the effect of the Syk inhibitor (GSK143) was evaluated on gastrointestinal transit, muscular inflammation and cytokine production. In vitro: the effect of GSK143 and doxantrazole were evaluated on cultured peritoneal mast cells (PMCs) and bone marrow derived macrophages. RESULTS: In vivo: intestinal manipulation resulted in a delay in gastrointestinal transit at t=24 h (Geometric Center (GC): 4.4 ± 0.3). Doxantrazole and GSK143 significantly increased gastrointestinal transit (GC doxantrazole (10 mg/kg): 7.2 ± 0.7; GSK143 (1 mg/kg): 7.6 ± 0.6), reduced inflammation and prevented recruitment of immune cells in the intestinal muscularis. In vitro: in PMCs, substance P (0-90 µM) and trinitrophenyl (0-4 µg/ml) induced a concentration-dependent release of ß-hexosaminidase. Pretreatment with doxantrazole and GSK143 (0.03-10 µM) concentration dependently blocked substance P and trinitrophenyl induced ß-hexosaminidase release. In addition, GSK143 was able to reduce cytokine expression in endotoxin-treated bone marrow derived macrophages in a concentration-dependent manner. CONCLUSIONS: The Syk inhibitor GSK143 reduces macrophage activation and mast cell degranulation in vitro. In addition, it inhibits manipulation-induced intestinal muscular inflammation and restores intestinal transit in mice. These findings suggest that Syk inhibition may be a new tool to shorten POI.


Asunto(s)
Compuestos de Anilina/uso terapéutico , Ileus/prevención & control , Péptidos y Proteínas de Señalización Intracelular/antagonistas & inhibidores , Complicaciones Posoperatorias/prevención & control , Inhibidores de Proteínas Quinasas/uso terapéutico , Proteínas Tirosina Quinasas/antagonistas & inhibidores , Pirimidinas/uso terapéutico , Compuestos de Anilina/administración & dosificación , Compuestos de Anilina/farmacología , Animales , Degranulación de la Célula/efectos de los fármacos , Células Cultivadas , Citocinas/biosíntesis , Relación Dosis-Respuesta a Droga , Evaluación Preclínica de Medicamentos/métodos , Tránsito Gastrointestinal/efectos de los fármacos , Ileus/fisiopatología , Activación de Macrófagos/efectos de los fármacos , Mastocitos/efectos de los fármacos , Mastocitos/fisiología , Ratones , Ratones Endogámicos C57BL , Ovalbúmina/antagonistas & inhibidores , Ovalbúmina/farmacología , Inhibidores de Fosfodiesterasa/uso terapéutico , Complicaciones Posoperatorias/fisiopatología , Inhibidores de Proteínas Quinasas/administración & dosificación , Inhibidores de Proteínas Quinasas/farmacología , Pirimidinas/administración & dosificación , Pirimidinas/farmacología , Sustancia P/antagonistas & inhibidores , Sustancia P/farmacología , Quinasa Syk , Tioxantenos/uso terapéutico , Xantonas/uso terapéutico
8.
Nat Rev Gastroenterol Hepatol ; 9(11): 675-83, 2012 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-22801725

RESUMEN

Patients undergoing an abdominal surgical procedure develop a transient episode of impaired gastrointestinal motility or postoperative ileus. Importantly, postoperative ileus is a major determinant of recovery after intestinal surgery and leads to increased morbidity and prolonged hospitalization, which is a great economic burden to health-care systems. Although a variety of strategies reduce postoperative ileus, including multimodal postoperative rehabilitation (fast-track care) and minimally invasive surgery, none of these methods have been completely successful in shortening the duration of postoperative ileus. The aetiology of postoperative ileus is multifactorial, but insights into the pathogenesis of postoperative ileus have identified intestinal inflammation, triggered by surgical handling, as the main mechanism. The importance of this inflammatory response in postoperative ileus is underscored by the beneficial effect of pharmacological interventions that block the influx of leukocytes. New insights into the pathophysiology of postoperative ileus and the involvement of the innate and the adaptive (T-helper type 1 cell-mediated immune response) immune system offer interesting and important new approaches to prevent postoperative ileus. In this Review, we discuss the latest insights into the mechanisms behind postoperative ileus and highlight new strategies to intervene in the postoperative inflammatory cascade.


Asunto(s)
Ileus/tratamiento farmacológico , Ileus/fisiopatología , Complicaciones Posoperatorias/tratamiento farmacológico , Complicaciones Posoperatorias/fisiopatología , Inmunidad Adaptativa/fisiología , Ghrelina/agonistas , Humanos , Inmunidad Innata/fisiología , Inflamación/fisiopatología , Naftoquinonas/uso terapéutico , Agonistas del Receptor de Serotonina 5-HT4/uso terapéutico
9.
Artículo en Inglés | MEDLINE | ID: mdl-23320135

RESUMEN

INTRODUCTION: Postoperative ileus (POI) is characterized by a transient inhibition of coordinated motility of the gastrointestinal (GI) tract after abdominal surgery and leads to increased morbidity and prolonged hospitalization. Currently, intestinal manipulation of the intestine is widely used as a preclinical model of POI. The technique used to manipulate the intestine is however highly variable and difficult to standardize, leading to large variations and inconsistent findings between different investigators. Therefore, we developed a device by which a fixed and adjustable pressure can be applied during intestinal manipulation. METHODS: The standardized pressure manipulation method was developed using the purpose-designed device. First, the effect of graded manipulation was examined on postoperative GI transit. Next, this new technique was compared to the conventional manipulation technique used in previous studies. GI transit was measured by evaluating the intestinal distribution of orally gavaged fluorescein isothiocyanate (FITC)-labeled dextran. Infiltration of myeloperoxidase positive cells and cytokine production (ELISA) in the muscularis externa of the intestine were assessed. RESULTS: Increasing pressures resulted in a graded reduction of intestinal transit and was associated with intestinal inflammation as demonstrated by influx of leukocytes and increased levels of IL-6, IL-1ß and MCP-1 compared to control mice. With an applied pressure of 9 grams a similar delay in intestinal transit could be obtained with a smaller standard deviation, leading to a reduced intra-individual variation. CONCLUSIONS: This method provides a reproducible model with small variation to study the pathophysiology of POI and to evaluate new anti-inflammatory strategies.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...