Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 55
Filtrar
1.
Int Immunopharmacol ; 136: 112316, 2024 Jul 30.
Artículo en Inglés | MEDLINE | ID: mdl-38823183

RESUMEN

The objective of this study was to investigate the neuroimmune mechanisms implicated in the enhancement of gastrointestinal function through the administration of oral DHA. Mast cell-deficient mice (KitW-sh) and C57BL/6 mice were used to establish postoperative ileus (POI) models. To further validate our findings, we conducted noncontact coculture experiments involving dorsal root ganglion (DRG) cells, bone marrow-derived mast cells (BMMCs) and T84 cells. Furthermore, the results obtained from investigations conducted on animals and cells were subsequently validated through clinical trials. The administration of oral DHA had ameliorative effects on intestinal barrier injury and postoperative ileus. In a mechanistic manner, the anti-inflammatory effect of DHA was achieved through the activation of transient receptor potential ankyrin 1 (TRPA1) on DRG cells, resulting in the stabilization of mast cells and increasing interleukin 10 (IL-10) secretion in mast cells. Furthermore, the activation of the pro-repair WNT1-inducible signaling protein 1 (WISP-1) signaling pathways by mast cell-derived IL-10 resulted in an enhancement of the intestinal barrier integrity. The current study demonstrated that the neuroimmune interaction between mast cells and nerves played a crucial role in the process of oral DHA improving the intestinal barrier integrity of POI, which further triggered the activation of CREB/WISP-1 signaling in intestinal mucosal cells.


Asunto(s)
Ácidos Docosahexaenoicos , Ileus , Interleucina-10 , Mucosa Intestinal , Mastocitos , Ratones Endogámicos C57BL , Complicaciones Posoperatorias , Canal Catiónico TRPA1 , Animales , Mastocitos/efectos de los fármacos , Mastocitos/inmunología , Ácidos Docosahexaenoicos/farmacología , Ácidos Docosahexaenoicos/uso terapéutico , Canal Catiónico TRPA1/metabolismo , Ratones , Ileus/tratamiento farmacológico , Ileus/inmunología , Humanos , Mucosa Intestinal/efectos de los fármacos , Mucosa Intestinal/inmunología , Mucosa Intestinal/patología , Mucosa Intestinal/metabolismo , Masculino , Interleucina-10/metabolismo , Complicaciones Posoperatorias/tratamiento farmacológico , Complicaciones Posoperatorias/inmunología , Ganglios Espinales/metabolismo , Ganglios Espinales/efectos de los fármacos , Modelos Animales de Enfermedad , Técnicas de Cocultivo , Antiinflamatorios/farmacología , Antiinflamatorios/uso terapéutico
2.
Acupunct Med ; 42(3): 123-132, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38813841

RESUMEN

BACKGROUND: Electroacupuncture (EA) has been reported to improve intestinal motility in mice with postoperative ileus (POI). Previous studies, however, have yielded heterogeneous results regarding the effect of EA on POI. METHODS: Herein, a POI mouse model was constructed by intestinal manipulation. To evaluate the effect of EA treatment on colonic transit, the levels of inflammatory markers (macrophage inflammatory protein (MIP)-1α, interleukin (IL)-1ß, IL-6, monocyte chemotactic protein (MCP)-1 and intercellular adhesion molecule (ICAM)-1) were detected by enzyme-linked immunosorbent assay (ELISA); immune cell infiltration was detected by immunohistochemical staining of myeloperoxidase (MPO), ectodysplasin (ED)-1 and ED-2, and the percentage of CD4+ interferon (IFN)-γ+ Th1 cells and IFN-γ secretion levels were determined. Activated Th1 cells and pentoxifylline, a cell differentiation inhibitor, were used to assess the role of Th1 cells in EA treatment of POI. Neostigmine administration and unilateral vagotomy were performed to confirm whether the effects of EA treatment on Th1 cells were mediated by the vagus nerve (VN). RESULTS: The results revealed that EA treatment at ST36 improved POI, as indicated by a decreased level of inflammatory-related markers and immune cell infiltration and shortened colonic transit time. The activated Th1 cells abolished the effects of EA treatment on POI. The effects of EA treatment on POI were enhanced by stimulation of the VN along with a decreased level of Th1 cells, but these effects were abolished by vagotomy along with an increased percentage of Th1 cells; this result indicates that the VN mediates the role of Th1 cells in the effects of EA treatment of POI. CONCLUSION: Our findings showed that the effects of EA treatment of POI were mainly mediated by Th1 cells through the stimulation of the VN and inhibition of the inflammatory response.


Asunto(s)
Electroacupuntura , Ileus , Complicaciones Posoperatorias , Células TH1 , Nervio Vago , Animales , Células TH1/inmunología , Ratones , Ileus/terapia , Ileus/inmunología , Nervio Vago/inmunología , Masculino , Humanos , Complicaciones Posoperatorias/terapia , Complicaciones Posoperatorias/inmunología , Ratones Endogámicos C57BL , Modelos Animales de Enfermedad , Interferón gamma/metabolismo , Interferón gamma/inmunología , Interleucina-6/metabolismo , Interleucina-6/inmunología , Molécula 1 de Adhesión Intercelular/metabolismo , Molécula 1 de Adhesión Intercelular/genética , Interleucina-1beta/metabolismo , Inflamación/terapia
3.
Int J Med Sci ; 18(13): 3014-3025, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34220330

RESUMEN

Rationale: Postoperative ileus (POI) is a frequent complication arising after gastrointestinal surgery but pathogenesis of POI is still not fully understood. While Th1 immune cells are implicated in POI, the involvement of Th2 cells has not yet been clarified. Given the impact of reactive oxygen species (ROS) in the regulation of Th1 and Th2 balance, we hypothesized that not only Th1 but also Th2 immune response can be involved in the development of experimental POI. Methods: The intestinal transit test was performed using carbon gum arabic. Electron microscopy was employed to assess tissue morphology and the presence of immune cells. Cytokines, IgE and ROS were measured. Immune cells from Peyer's patches were analyzed by Flow Cytometry and toluidine blue staining was used for detection of mast cells. Transcriptional factors were analyzed by Western blot. Results: POI is associated with an increase in both Th2 cytokines and Th2 cells. We have further demonstrated that POI induces a Th2-dependent activation of memory and non-memory B cells. This was accompanied by an increase in a number of mast cells in the colon of POI mice as well by an increased IgE and histamine plasma levels. We found that POI-induced accumulation of ROS was associated with an increased expression of the transcriptional factors HMBGI, NF-κB, and p38. This increased expression seemed to be associated with a Th2 response. Conclusion: Th2 immune response can be involved in the activation of mast cells in POI, which was associated with ROS mediated activation of NF-κB and p38 MAPK signaling pathway.


Asunto(s)
Procedimientos Quirúrgicos del Sistema Digestivo/efectos adversos , Ileus/inmunología , Complicaciones Posoperatorias/inmunología , Células Th2/inmunología , Animales , Comunicación Celular/inmunología , Modelos Animales de Enfermedad , Femenino , Humanos , Ileus/sangre , Sistema de Señalización de MAP Quinasas/inmunología , Masculino , Mastocitos/inmunología , Mastocitos/metabolismo , Ratones , FN-kappa B/metabolismo , Complicaciones Posoperatorias/sangre , Especies Reactivas de Oxígeno/metabolismo , Células TH1/inmunología , Células Th2/metabolismo
4.
Inflammation ; 44(3): 1145-1159, 2021 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-33398542

RESUMEN

Inflammation theory has suggested that the pathogenesis of postoperative ileus (POI) involves the steroid receptor coactivator-3 (SRC-3). Therefore, we investigated the role of SRC-3 in the muscles of the small intestine using a mouse POI model. Here, we reported that intestinal manipulation (IM) significantly reduced the extent of phenol red migration in the entire gastrointestinal tract, and the calculated geometric center (GC) value in wild-type (WT) mice at 24 h after surgery was higher than that in the knockout (KO) mice and in the sham-operated control group. The expression of SRC-3 was upregulated in the mouse intestinal muscularis at 24 h after surgical manipulation, and the mRNA and protein levels of inflammatory cytokines were upregulated compared with those in the control group. At 24 h after IM, the number of neutrophils in the experimental group was significantly higher than that in the control group; in the IM group, the number of neutrophils in the SRC-3-/- mice was markedly higher than that in the WT mice. At 24 h after IM, the myeloperoxidase (MPO) activity in the experimental group was significantly higher than that in the control group. In the IM group, the MPO activity of the SRC-3-/- mice was markedly higher than that of the WT mice. In summary, proinflammatory cytokines, the number of neutrophils, and the MPO activity were significantly increased in the muscularis of the jejunum and ileum of KO mice after IM compared with those of the WT mice, indicating that SRC-3 might play a protective role in POI.


Asunto(s)
Citocinas/metabolismo , Motilidad Gastrointestinal , Ileus/metabolismo , Mediadores de Inflamación/metabolismo , Intestino Delgado/metabolismo , Músculo Liso/metabolismo , Coactivador 3 de Receptor Nuclear/metabolismo , Complicaciones Posoperatorias/metabolismo , Animales , Modelos Animales de Enfermedad , Femenino , Ileus/etiología , Ileus/inmunología , Ileus/fisiopatología , Intestino Delgado/inmunología , Intestino Delgado/fisiopatología , Yeyuno/inmunología , Yeyuno/metabolismo , Yeyuno/fisiopatología , Ratones de la Cepa 129 , Ratones Endogámicos C57BL , Ratones Noqueados , Músculo Liso/inmunología , Músculo Liso/fisiopatología , Infiltración Neutrófila , Coactivador 3 de Receptor Nuclear/genética , Peroxidasa/metabolismo , Complicaciones Posoperatorias/etiología , Complicaciones Posoperatorias/inmunología , Complicaciones Posoperatorias/fisiopatología , Técnicas de Cultivo de Tejidos
5.
Neurogastroenterol Motil ; 33(5): e14046, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-33252179

RESUMEN

BACKGROUND: Postoperative ileus is common and is a major clinical problem. It has been widely studied in patients and in experimental models in laboratory animals. A wide variety of treatments have been tested to prevent or modify the course of this disorder. PURPOSE: This review draws together information on animal studies of ileus with studies on human patients. It summarizes some of the conceptual advances made in understanding the mechanisms that underlie paralytic ileus. The treatments that have been tested in human subjects (both pharmacological and non-pharmacological) and their efficacy are summarized and graded consistent with current clinical guidelines. The review is not intended to provide a comprehensive overview of ileus, but rather a general understanding of the major clinical problems associated with it, how animal models have been useful to elucidate key mechanisms and, finally, some perspectives from both scientists and clinicians as to how we may move forward with this debilitating yet common condition.


Asunto(s)
Sistema Nervioso Entérico/fisiopatología , Motilidad Gastrointestinal/fisiología , Ileus/fisiopatología , Complicaciones Posoperatorias/fisiopatología , Sistema Nervioso Simpático/fisiopatología , Anestesia Epidural , Animales , Benzofuranos/uso terapéutico , Goma de Mascar , Colinérgicos/uso terapéutico , Medios de Contraste/uso terapéutico , Inhibidores de la Ciclooxigenasa/uso terapéutico , Diatrizoato de Meglumina/uso terapéutico , Procedimientos Quirúrgicos del Sistema Digestivo/métodos , Recuperación Mejorada Después de la Cirugía , Nutrición Enteral , Fluidoterapia , Fármacos Gastrointestinales/uso terapéutico , Ghrelina/uso terapéutico , Humanos , Ileus/inmunología , Ileus/prevención & control , Ileus/terapia , Inflamación/inmunología , Seudoobstrucción Intestinal/inmunología , Seudoobstrucción Intestinal/fisiopatología , Seudoobstrucción Intestinal/prevención & control , Seudoobstrucción Intestinal/terapia , Intubación Gastrointestinal , Laparoscopía , Mastocitos/inmunología , Piperidinas/uso terapéutico , Complicaciones Posoperatorias/inmunología , Complicaciones Posoperatorias/prevención & control , Complicaciones Posoperatorias/terapia , Agonistas del Receptor de Serotonina 5-HT4/uso terapéutico , Simpaticolíticos/uso terapéutico
6.
Front Immunol ; 11: 308, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32265899

RESUMEN

The dense innervation of the gastro-intestinal tract with neuronal networks, which are in close proximity to immune cells, implies a pivotal role of neurons in modulating immune functions. Neurons have the ability to directly sense danger signals, adapt immune effector functions and integrate these signals to maintain tissue integrity and host defense strategies. The expression pattern of a large set of immune cells in the intestine characterized by receptors for neurotransmitters and neuropeptides suggest a tight neuronal hierarchical control of immune functions in order to systemically control immune reactions. Compelling evidence implies that targeting neuro-immune interactions is a promising strategy to dampen immune responses in autoimmune diseases such as inflammatory bowel diseases or rheumatoid arthritis. In fact, electric stimulation of vagal fibers has been shown to be an extremely effective treatment strategy against overwhelming immune reactions, even after exhausted conventional treatment strategies. Such findings argue that the nervous system is underestimated coordinator of immune reactions and underline the importance of neuro-immune crosstalk for body homeostasis. Herein, we review neuro-immune interactions with a special focus on disease pathogenesis throughout the gastro-intestinal tract.


Asunto(s)
Homeostasis/inmunología , Inmunidad/inmunología , Neuroinmunomodulación/inmunología , Animales , Artritis Reumatoide/inmunología , Artritis Reumatoide/terapia , Enfermedades Autoinmunes/inmunología , Enfermedades Autoinmunes/terapia , Sistema Nervioso Autónomo/fisiología , Sistema Nervioso Entérico/fisiología , Microbioma Gastrointestinal , Humanos , Sistema Hipotálamo-Hipofisario/fisiología , Ileus/inmunología , Ileus/terapia , Inmunidad Innata , Enfermedades Inflamatorias del Intestino/inmunología , Linfocitos/inmunología , Neuroinmunomodulación/efectos de los fármacos , Neuronas/fisiología , Neuropéptidos/inmunología , Sistemas Neurosecretores/fisiología , Neurotransmisores/agonistas , Neurotransmisores/fisiología , Complicaciones Posoperatorias/inmunología , Complicaciones Posoperatorias/terapia , Receptores de Neurotransmisores/inmunología , Sepsis/inmunología , Sepsis/terapia , Simbiosis , Estimulación del Nervio Vago
7.
Front Immunol ; 11: 581111, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33519804

RESUMEN

Postoperative ileus (POI) is triggered by an innate immune response in the muscularis externa (ME) and is accompanied by bacterial translocation. Bacteria can trigger an innate immune response via toll-like receptor (TLR) activation, but the latter's contribution to POI has been disproved for several TLRs, including TLR2 and TLR4. Herein we investigated the role of double-stranded RNA detection via TLR3 and TIR-domain-containing adapter-inducing interferon-ß (TRIF) signaling pathway in POI. POI was induced by small bowel intestinal manipulation in wt, TRIF-/-, TLR3-/-, type I interferon receptor-/- and interferon-ß reporter mice, all on C57BL/6 background, and POI severity was quantified by gene expression analysis, gastrointestinal transit and leukocyte extravasation into the ME. TRIF/TLR3 deficiency reduced postoperative ME inflammation and prevented POI. With bone marrow transplantation, RNA-sequencing, flow cytometry and immunohistochemistry we revealed a distinct TLR3-expressing radio-resistant MHCIIhiCX3CR1- IBA-1+ resident macrophage population within the deep myenteric plexus. TLR3 deficiency in these cells, but not in MHCIIhiCX3CR1+ macrophages, reduced cytokine expression in POI. While this might not be an exclusive macrophage-privileged pathway, the TLR3/TRIF axis contributes to proinflammatory cytokine production in MHCIIhiCX3CR1- IBA-1+ macrophages during POI. Deficiency in TLR3/TRIF protects mice from POI. These data suggest that TLR3 antagonism may prevent POI in humans.


Asunto(s)
Ileus/etiología , Macrófagos/inmunología , Complicaciones Posoperatorias/etiología , Receptor Toll-Like 3/inmunología , Proteínas Adaptadoras del Transporte Vesicular/deficiencia , Proteínas Adaptadoras del Transporte Vesicular/genética , Proteínas Adaptadoras del Transporte Vesicular/inmunología , Animales , Receptor 1 de Quimiocinas CX3C/genética , Receptor 1 de Quimiocinas CX3C/inmunología , Modelos Animales de Enfermedad , Femenino , Expresión Génica , Ileus/inmunología , Ileus/patología , Inmunidad Innata , Macrófagos/clasificación , Macrófagos/efectos de la radiación , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Plexo Mientérico/inmunología , Complicaciones Posoperatorias/inmunología , Complicaciones Posoperatorias/patología , Tolerancia a Radiación/inmunología , Receptor de Interferón alfa y beta/deficiencia , Receptor de Interferón alfa y beta/genética , Receptor de Interferón alfa y beta/inmunología , Transducción de Señal/inmunología , Receptor Toll-Like 3/deficiencia , Receptor Toll-Like 3/genética , Quimera por Trasplante/inmunología
8.
Sci Rep ; 9(1): 10602, 2019 07 22.
Artículo en Inglés | MEDLINE | ID: mdl-31332247

RESUMEN

Postoperative ileus (POI) is an intestinal dysmotility frequently occurring after abdominal surgery. An orchestrated neuroimmune response within the muscularis externa (ME) involves activation of resident macrophages, enteric glia and infiltration of blood-derived leukocytes. Interleukin-1 receptor type-I (IL1R1) signalling on enteric glia has been shown to be involved in POI development. Herein we investigated the distinct role of the IL1R1 ligands interleukin (IL) -1α and IL-1ß and focused on the mechanism of IL-1ß production. IL-1α and IL-1ß deficient mice were protected from POI. Bone-marrow transplantation studies indicated that IL-1α originated from radio-resistant cells while IL-1ß was released from the radio-sensitive infiltrating leukocytes. Mouse strains deficient in inflammasome formation identified the absent in melanoma 2 (AIM2) inflammasome to be crucial for IL-1ß production in POI. Mechanistically, antibiotic-treated mice revealed a prominent role of the microbiome in IL-1ß production. Our study provides new insights into distinct roles of IL-1α and IL-1ß signalling during POI. While IL-1α release is most likely an immediate passive response to the surgical trauma, IL-1ß production depends on AIM2 inflammasome formation and the microbiome. Selective interaction in this pathway might be a promising target to prevent POI in surgical patients.


Asunto(s)
Proteínas de Unión al ADN/metabolismo , Ileus/etiología , Inflamasomas/metabolismo , Interleucina-1beta/metabolismo , Complicaciones Posoperatorias/etiología , Animales , Microbioma Gastrointestinal , Ileus/inmunología , Ileus/metabolismo , Inmunidad Innata , Interleucina-1alfa/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Complicaciones Posoperatorias/inmunología , Complicaciones Posoperatorias/metabolismo
9.
Gut ; 68(8): 1406-1416, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-30472681

RESUMEN

OBJECTIVES: Vagus nerve stimulation (VNS), most likely via enteric neurons, prevents postoperative ileus (POI) by reducing activation of alpha7 nicotinic receptor (α7nAChR) positive muscularis macrophages (mMφ) and dampening surgery-induced intestinal inflammation. Here, we evaluated if 5-HT4 receptor (5-HT4R) agonist prucalopride can mimic this effect in mice and human. DESIGN: Using Ca2+ imaging, the effect of electrical field stimulation (EFS) and prucalopride was evaluated in situ on mMφ activation evoked by ATP in jejunal muscularis tissue. Next, preoperative and postoperative administration of prucalopride (1-5 mg/kg) was compared with that of preoperative VNS in a model of POI in wild-type and α7nAChR knockout mice. Finally, in a pilot study, patients undergoing a Whipple procedure were preoperatively treated with prucalopride (n=10), abdominal VNS (n=10) or sham/placebo (n=10) to evaluate the effect on intestinal inflammation and clinical recovery of POI. RESULTS: EFS reduced the ATP-induced Ca2+ response of mMφ, an effect that was dampened by neurotoxins tetrodotoxin and ω-conotoxin and mimicked by prucalopride. In vivo, prucalopride administered before, but not after abdominal surgery reduced intestinal inflammation and prevented POI in wild-type, but not in α7nAChR knockout mice. In humans, preoperative administration of prucalopride, but not of VNS, decreased Il6 and Il8 expression in the muscularis externa and improved clinical recovery. CONCLUSION: Enteric neurons dampen mMφ activation, an effect mimicked by prucalopride. Preoperative, but not postoperative treatment with prucalopride prevents intestinal inflammation and shortens POI in both mice and human, indicating that preoperative administration of 5-HT4R agonists should be further evaluated as a treatment of POI. TRIAL REGISTRATION NUMBER: NCT02425774.


Asunto(s)
Benzofuranos , Ileus , Intestino Delgado , Músculo Liso , Pancreaticoduodenectomía/efectos adversos , Complicaciones Posoperatorias , Adulto , Animales , Benzofuranos/administración & dosificación , Benzofuranos/farmacología , Modelos Animales de Enfermedad , Femenino , Motilidad Gastrointestinal/efectos de los fármacos , Humanos , Ileus/etiología , Ileus/inmunología , Ileus/fisiopatología , Ileus/prevención & control , Inflamación/inmunología , Inflamación/prevención & control , Intestino Delgado/inmunología , Intestino Delgado/inervación , Intestino Delgado/patología , Intestino Delgado/fisiopatología , Macrófagos/inmunología , Macrófagos/patología , Masculino , Ratones , Músculo Liso/efectos de los fármacos , Músculo Liso/patología , Músculo Liso/fisiopatología , Pancreaticoduodenectomía/métodos , Proyectos Piloto , Complicaciones Posoperatorias/inmunología , Complicaciones Posoperatorias/fisiopatología , Complicaciones Posoperatorias/prevención & control , Agonistas del Receptor de Serotonina 5-HT4/administración & dosificación , Agonistas del Receptor de Serotonina 5-HT4/farmacología , Resultado del Tratamiento , Receptor Nicotínico de Acetilcolina alfa 7/metabolismo
10.
Front Immunol ; 9: 2599, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30581430

RESUMEN

Objective: Postoperative ileus (POI) is an inflammation-mediated complication of abdominal surgery, characterized by intestinal dysmotility and leukocyte infiltration into the muscularis externa (ME). Previous studies indicated that interleukin (IL)-10 is crucial for the resolution of a variety of inflammation-driven diseases. Herein, we investigated how IL-10 affects the postoperative ME inflammation and found an unforeseen role of IL-10 in POI. Design: POI was induced by a standardized intestinal manipulation (IM) in C57BL/6 and multiple transgenic mouse strain including C-C motif chemokine receptor 2-/-, IL-10-/-, and LysMcre/IL-10fl/fl mice. Leukocyte infiltration, gene and protein expression of cytokines, chemokines, and macrophage differentiation markers as well as intestinal motility were analyzed. IL-10 serum levels in surgical patients were determined by ELISA. Results: IL-10 serum levels were increased in patient after abdominal surgery. In mice, a complete or leucocyte-restricted IL-10 deficiency ameliorated POI and reduced the postoperative ME neutrophil infiltration. Infiltrating monocytes were identified as main IL-10 producers and undergo IL-10-dependent M2 polarization. Interestingly, M2 polarization is not crucial to POI development as abrogation of monocyte infiltration did not prevent POI due to a compensation of the IL-10 loss by resident macrophages and neutrophils. Organ culture studies demonstrated that IL-10 deficiency impeded neutrophil migration toward the surgically traumatized ME. This mechanism is mediated by reduction of neutrophil attracting chemokines. Conclusion: Monocyte-derived macrophages are the major IL-10 source during POI. An IL-10 deficiency decreases the postoperative expression of neutrophil-recruiting chemokines, consequently reduces the neutrophil extravasation into the postsurgical bowel wall, and finally protects mice from POI.


Asunto(s)
Ileus/inmunología , Interleucina-10/inmunología , Intestinos/inmunología , Leucocitos/inmunología , Complicaciones Posoperatorias/inmunología , Animales , Modelos Animales de Enfermedad , Motilidad Gastrointestinal/inmunología , Humanos , Inflamación/inmunología , Macrófagos/inmunología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos/inmunología , Monocitos/inmunología , Infiltración Neutrófila/inmunología , Neutrófilos/inmunología , Periodo Posoperatorio
11.
Med Sci Monit ; 24: 7231-7237, 2018 Oct 10.
Artículo en Inglés | MEDLINE | ID: mdl-30303179

RESUMEN

BACKGROUND The aim of this study was to determine the effects of laparoscopic surgery within an ERAS program on outcomes and immunological function in patients with a carcinoma in the right colon. MATERIAL AND METHODS Patient data were acquired from a prospectively maintained database, and 176 patients diagnosed with right colon carcinoma with surgery were selected from the database. These patients were divided into a laparoscopic group (Lap group, n=86) and an open operation group (Open group, n=90). All patients received treatment according to a standardized ERAS protocol. We collected data on CRP levels, CD4+/CD8+ ratios, and Treg values in peripheral blood, baseline and surgical characteristics, postoperative complications, and postoperative ileus (POI). RESULTS Circulating CD4+/CD8+ ratios and Treg values were decreased and CRP levels were increased in both groups after the operation. However, the values in the Lap group patients recovered much more quickly than those of patients in the Open group (P<0.05). Patients undergoing laparoscopic surgery had significantly less preoperative bleeding (P<0.01), reduced ratio of overall POI (mainly early ileus), and shorter postoperative hospital stay (P=0.03). Multivariate logistic regression analysis showed that POD1 Treg value was an independent predicator for postoperative ileus in patients with right colon carcinoma resection. CONCLUSIONS In patients with a carcinoma in the right colon, laparoscopic surgery within an ERAS protocol leads to better immunity preservation after surgery, and POD1 Treg value may be an independent predicator for postoperative ileus, which could, at least in part, explain the shorter hospital stay after surgery.


Asunto(s)
Neoplasias del Colon/cirugía , Procedimientos Quirúrgicos del Sistema Digestivo/métodos , Ileus/prevención & control , Laparoscopía/métodos , Linfocitos T Reguladores/inmunología , Anciano , Anciano de 80 o más Años , Neoplasias del Colon/sangre , Neoplasias del Colon/inmunología , Neoplasias del Colon/patología , Procedimientos Quirúrgicos del Sistema Digestivo/efectos adversos , Femenino , Humanos , Ileus/sangre , Ileus/etiología , Ileus/inmunología , Laparoscopía/efectos adversos , Tiempo de Internación , Masculino , Persona de Mediana Edad , Estadificación de Neoplasias , Atención Perioperativa/métodos , Cuidados Posoperatorios/métodos , Complicaciones Posoperatorias/sangre , Complicaciones Posoperatorias/etiología , Complicaciones Posoperatorias/inmunología , Complicaciones Posoperatorias/prevención & control , Recuperación de la Función , Estudios Retrospectivos , Linfocitos T Reguladores/patología , Resultado del Tratamiento
12.
J Pharmacol Sci ; 137(4): 379-386, 2018 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-30145033

RESUMEN

Zingiberis processum rhizoma (ZPR) is a major active component of daikenchuto (DKT), which induces anti-inflammatory action by inhibiting macrophage infiltration. However, it is unclear whether ZPR is related to DKT-induced anti-inflammatory action via a reduction of neutrophil infiltration against postoperative ileus (POI). In this study, we orally administered individual herbal components of DKT to mice four times before and after intestinal manipulation (IM). The anti-inflammatory action of each crude drug was evaluated by histochemical analysis of relevant molecules. The results showed that treatment with all herbal components of DKT significantly inhibits neutrophil infiltration. This inhibition of neutrophil infiltration by ZPR was significantly reduced in 5-hydroxytryptamine receptor 4 (5-HT4R) knockout (KO) mice but not in alpha-7 nicotinic acetylcholine receptor (α7nAChR) KO mice. Also, transient receptor potential ankyrin 1 (TRPA1) and muscarinic acetylcholine receptor (mAChR) antagonists partly and significantly inhibited the amelioration of neutrophil infiltration by ZPR. Therefore, DKT-induced anti-inflammatory action, mediated by inhibition of neutrophil infiltration in POI, depends, in part, on the effects of ZPR. ZPR activates TRPA1 channels, possibly in enterochromaffin (EC) cells, to release 5-HT. This 5-HT stimulates 5-HT4R in the myenteric plexus neurons to release acetylcholine, which, in turn, activates mAChR to inhibit inflammation in POI.


Asunto(s)
Antiinflamatorios , Ileus/inmunología , Infiltración Neutrófila/efectos de los fármacos , Extractos Vegetales/farmacología , Complicaciones Posoperatorias/inmunología , Receptores Muscarínicos/metabolismo , Zingiberaceae/química , Acetilcolina/metabolismo , Administración Oral , Animales , Modelos Animales de Enfermedad , Células Enterocromafines/metabolismo , Masculino , Ratones Endogámicos C57BL , Plexo Mientérico/metabolismo , Panax , Extractos Vegetales/administración & dosificación , Extractos Vegetales/aislamiento & purificación , Receptores de Serotonina 5-HT4/metabolismo , Serotonina/metabolismo , Canal Catiónico TRPA1/metabolismo , Zanthoxylum
13.
Am J Physiol Gastrointest Liver Physiol ; 315(1): G104-G116, 2018 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-29565641

RESUMEN

In this study, we investigated the role of transient receptor potential melastatin 2 (TRPM2), a nonselective cation channel abundantly expressed in inflammatory cells such as macrophages, in the development of postoperative ileus, a complication of abdominal surgery characterized by gastrointestinal dysmotility. In wild-type mice, we found that intestinal manipulation, a maneuver that elicits symptoms typical of postoperative ileus, delays the transit of fluorescein-labeled dextran, promotes the infiltration of CD68+ macrophages, Ly6B.2+ neutrophils, and MPO+ cells into intestinal muscles, boosts expression of IL-1ß, IL-6, TNF-α, iNOS, and CXCL2 in intestinal muscles and peritoneal macrophages, enhances phosphorylation of ERK and p38 MAPK in intestinal muscles, and amplifies IL-1ß, IL-6, TNF-α, iNOS, and CXCL2 expression in resident and thioglycolate-elicited peritoneal macrophages following exposure to lipopolysaccharide. Remarkably, TRPM2 deficiency completely blocks or diminishes these effects. Indeed, intestinal manipulation appears to activate TRPM2 in resident muscularis macrophages and elicits release of inflammatory cytokines and chemokines, which, in turn, promote infiltration of macrophages and neutrophils into the muscle, ultimately resulting in dysmotility. NEW & NOTEWORTHY Activation of transient receptor potential melastatin 2 (TRPM2) releases inflammatory cytokines and chemokines, which, in turn, promote the infiltration of inflammatory cells and macrophages into intestinal muscles, ultimately resulting in dysmotility. Thus TRPM2 is a promising target in treating dysmotility due to postoperative ileus, a complication of abdominal surgery.


Asunto(s)
Motilidad Gastrointestinal/inmunología , Ileus , Laparotomía/efectos adversos , Complicaciones Posoperatorias/inmunología , Canales Catiónicos TRPM/metabolismo , Animales , Quimiocina CXCL2/metabolismo , Modelos Animales de Enfermedad , Ileus/etiología , Ileus/inmunología , Inflamación/metabolismo , Interleucina-1beta/metabolismo , Macrófagos Peritoneales/metabolismo , Ratones , Músculo Liso/metabolismo , Neutrófilos/metabolismo , Canales Catiónicos TRPC/metabolismo
14.
Am J Physiol Gastrointest Liver Physiol ; 314(1): G75-G80, 2018 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-28912251

RESUMEN

Many essential gastrointestinal functions, including motility, secretion, and blood flow, are regulated by the autonomic nervous system (ANS), both through intrinsic enteric neurons and extrinsic (sympathetic and parasympathetic) innervation. Recently identified neuroimmune mechanisms, in particular the interplay between enteric neurons and muscularis macrophages, are now considered to be essential for fine-tuning peristalsis. These findings shed new light on how intestinal immune cells can support enteric nervous function. In addition, both intrinsic and extrinsic neural mechanisms control intestinal immune homeostasis in different layers of the intestine, mainly by affecting macrophage activation through neurotransmitter release. In this mini-review, we discuss recent insights on immunomodulation by intrinsic enteric neurons and extrinsic innervation, with a particular focus on intestinal macrophages. In addition, we discuss the relevance of these novel mechanisms for intestinal immune homeostasis in physiological and pathological conditions, mainly focusing on motility disorders (gastroparesis and postoperative ileus) and inflammatory disorders (colitis).


Asunto(s)
Sistema Nervioso Entérico/fisiología , Intestinos/inmunología , Intestinos/inervación , Macrófagos/inmunología , Neuroinmunomodulación , Animales , Colitis/inmunología , Colitis/fisiopatología , Gastroparesia/inmunología , Gastroparesia/fisiopatología , Homeostasis , Humanos , Ileus/inmunología , Ileus/fisiopatología , Activación de Macrófagos
15.
Gut ; 66(12): 2098-2109, 2017 12.
Artículo en Inglés | MEDLINE | ID: mdl-28615302

RESUMEN

OBJECTIVE: Postoperative ileus (POI) is assumed to result from myeloid cells infiltrating the intestinal muscularis externa (ME) in patients undergoing abdominal surgery. In the current study, we investigated the role of infiltrating monocytes in a murine model of intestinal manipulation (IM)-induced POI in order to clarify whether monocytes mediate tissue damage and intestinal dysfunction or they are rather involved in the recovery of gastrointestinal (GI) motility. DESIGN: IM was performed in mice with defective monocyte migration to tissues (C-C motif chemokine receptor 2, Ccr2-/ - mice) and wild-type (WT) mice to study the role of monocytes and monocyte-derived macrophages (MΦs) during onset and resolution of ME inflammation. RESULTS: At early time points, IM-induced GI transit delay and inflammation were equal in WT and Ccr2 -/- mice. However, GI transit recovery after IM was significantly delayed in Ccr2 -/- mice compared with WT mice, associated with increased neutrophil-mediated immunopathology and persistent impaired neuromuscular function. During recovery, monocyte-derived MΦs acquire pro-resolving features that aided in the resolution of inflammation. In line, bone marrow reconstitution and treatment with MΦ colony-stimulating factor 1 enhanced monocyte recruitment and MΦ differentiation and ameliorated GI transit in Ccr2 -/- mice. CONCLUSION: Our study reveals a critical role for monocyte-derived MΦs in restoring intestinal homeostasis after surgical trauma. From a therapeutic point of view, our data indicate that inappropriate targeting of monocytes may increase neutrophil-mediated immunopathology and prolong the clinical outcome of POI, while future therapies should be aimed at enhancing MΦ physiological repair functions.


Asunto(s)
Ileus/inmunología , Ileus/patología , Macrófagos/inmunología , Monocitos/inmunología , Complicaciones Posoperatorias/inmunología , Complicaciones Posoperatorias/patología , Receptores CCR2/inmunología , Animales , Diferenciación Celular , Movimiento Celular , Modelos Animales de Enfermedad , Motilidad Gastrointestinal , Tránsito Gastrointestinal , Homeostasis/inmunología , Inflamación/inmunología , Inflamación/patología , Ratones , Músculo Liso/patología
16.
Gut ; 66(12): 2110-2120, 2017 12.
Artículo en Inglés | MEDLINE | ID: mdl-28615301

RESUMEN

OBJECTIVE: Postoperative ileus (POI), the most frequent complication after intestinal surgery, depends on dendritic cells (DCs) and macrophages. Here, we have investigated the mechanism that activates these cells and the contribution of the intestinal microbiota for POI induction. DESIGN: POI was induced by manipulating the intestine of mice, which selectively lack DCs, monocytes or macrophages. The disease severity in the small and large intestine was analysed by determining the distribution of orally applied fluorescein isothiocyanate-dextran and by measuring the excretion time of a retrogradely inserted glass ball. The impact of the microbiota on intestinal peristalsis was evaluated after oral antibiotic treatment. RESULTS: We found that Cd11c-Cre+ Irf4flox/flox mice lack CD103+CD11b+ DCs, a DC subset unique to the intestine whose function is poorly understood. Their absence in the intestinal muscularis reduced pathogenic inducible nitric oxide synthase (iNOS) production by monocytes and macrophages and ameliorated POI. Pathogenic iNOS was produced in the jejunum by resident Ly6C- macrophages and infiltrating chemokine receptor 2-dependent Ly6C+ monocytes, but in the colon only by the latter demonstrating differential tolerance mechanisms along the intestinal tract. Consistently, depletion of both cell subsets reduced small intestinal POI, whereas the depletion of Ly6C+ monocytes alone was sufficient to prevent large intestinal POI. The differential role of monocytes and macrophages in small and large intestinal POI suggested a potential role of the intestinal microbiota. Indeed, antibiotic treatment reduced iNOS levels and ameliorated POI. CONCLUSIONS: Our findings reveal that CD103+CD11b+ DCs and the intestinal microbiome are a prerequisite for the activation of intestinal monocytes and macrophages and for dysregulating intestinal motility in POI.


Asunto(s)
Células Dendríticas/citología , Microbioma Gastrointestinal , Ileus/inmunología , Ileus/microbiología , Activación de Macrófagos , Monocitos/inmunología , Peristaltismo/inmunología , Complicaciones Posoperatorias/inmunología , Complicaciones Posoperatorias/microbiología , Animales , Antígenos CD/inmunología , Antígeno CD11b/inmunología , Modelos Animales de Enfermedad , Tránsito Gastrointestinal , Ileus/fisiopatología , Cadenas alfa de Integrinas/inmunología , Ratones , Ratones Transgénicos , Complicaciones Posoperatorias/fisiopatología
17.
J Neurosci ; 36(28): 7428-40, 2016 07 13.
Artículo en Inglés | MEDLINE | ID: mdl-27413153

RESUMEN

UNLABELLED: Acute brain ischemia induces a local neuroinflammatory reaction and alters peripheral immune homeostasis at the same time. Recent evidence has suggested a key role of the gut microbiota in autoimmune diseases by modulating immune homeostasis. Therefore, we investigated the mechanistic link among acute brain ischemia, microbiota alterations, and the immune response after brain injury. Using two distinct models of acute middle cerebral artery occlusion, we show by next-generation sequencing that large stroke lesions cause gut microbiota dysbiosis, which in turn affects stroke outcome via immune-mediated mechanisms. Reduced species diversity and bacterial overgrowth of bacteroidetes were identified as hallmarks of poststroke dysbiosis, which was associated with intestinal barrier dysfunction and reduced intestinal motility as determined by in vivo intestinal bolus tracking. Recolonizing germ-free mice with dysbiotic poststroke microbiota exacerbates lesion volume and functional deficits after experimental stroke compared with the recolonization with a normal control microbiota. In addition, recolonization of mice with a dysbiotic microbiome induces a proinflammatory T-cell polarization in the intestinal immune compartment and in the ischemic brain. Using in vivo cell-tracking studies, we demonstrate the migration of intestinal lymphocytes to the ischemic brain. Therapeutic transplantation of fecal microbiota normalizes brain lesion-induced dysbiosis and improves stroke outcome. These results support a novel mechanism in which the gut microbiome is a target of stroke-induced systemic alterations and an effector with substantial impact on stroke outcome. SIGNIFICANCE STATEMENT: We have identified a bidirectional communication along the brain-gut microbiota-immune axis and show that the gut microbiota is a central regulator of immune homeostasis. Acute brain lesions induced dysbiosis of the microbiome and, in turn, changes in the gut microbiota affected neuroinflammatory and functional outcome after brain injury. The microbiota impact on immunity and stroke outcome was transmissible by microbiota transplantation. Our findings support an emerging concept in which the gut microbiota is a key regulator in priming the neuroinflammatory response to brain injury. These findings highlight the key role of microbiota as a potential therapeutic target to protect brain function after injury.


Asunto(s)
Disbiosis/etiología , Encefalitis/complicaciones , Encefalitis/etiología , Microbiota/fisiología , Accidente Cerebrovascular/complicaciones , Animales , Infarto Encefálico/etiología , Complejo CD3/metabolismo , Modelos Animales de Enfermedad , Disbiosis/inmunología , Disbiosis/microbiología , Heces/microbiología , Femenino , Enfermedades Gastrointestinales/etiología , Motilidad Gastrointestinal/fisiología , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Ileus/inmunología , Ileus/microbiología , Ileus/patología , Infarto de la Arteria Cerebral Media/complicaciones , Leucocitos/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Microbiota/inmunología , Accidente Cerebrovascular/etiología , Estructuras Linfoides Terciarias/patología
19.
J Leukoc Biol ; 99(2): 231-9, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26292977

RESUMEN

Resolution of inflammation is an active counter-regulatory mechanism involving polyunsaturated fatty acid-derived proresolving lipid mediators. Postoperative intestinal motility disturbances, clinically known as postoperative ileus, occur frequently after abdominal surgery and are mediated by a complex inflammation of the intestinal muscularis externa. Herein, we tested the hypothesis that proresolving lipid mediators are involved in the resolution of postoperative ileus. In a standardized experimental model of postoperative ileus, we detected strong expression of 12/15-lipoxygenase within the postoperative muscularis externa of C57BL/6 mice, predominately located within CX3CR1(+)/Ly6C(+) infiltrating monocytes rather than Ly6G(+) neutrophils. Mass spectrometry analyses demonstrated that a 12/15-lipoxygenase increase was accompanied by production of docosahexaenoic acid-derived lipid mediators, particularly protectin DX and resolvin D2, and their common precursor 17-hydroxy docosahexaenoic acid. Perioperative administration of protectin DX, but not resolvin D2 diminished blood-derived leukocyte infiltration into the surgically manipulated muscularis externa and improved the gastrointestinal motility. Flow cytometry analyses showed impaired Ly6G(+)/Ly6C(+) neutrophil extravasation after protectin DX treatment, whereas Ly6G(-)/Ly6C(+) monocyte numbers were not affected. 12/15-lipoxygenase-deficient mice, lacking endogenous protectin DX synthesis, demonstrated increased postoperative leukocyte levels. Preoperative intravenous administration of a docosahexaenoic acid-rich lipid emulsion reduced postoperative leukocyte infiltration in wild-type mice but failed in 12/15-lipoxygenase-deficient mice mice. Protectin DX application reduced leukocyte influx and rescued 12/15-lipoxygenase-deficient mice mice from postoperative ileus. In conclusion, our results show that 12/15-lipoxygenase mediates postoperative ileus resolution via production of proresolving docosahexaenoic acid-derived protectin DX. Perioperative, parenteral protectin DX or docosahexaenoic acid supplementation, as well as modulation of the 12/15-lipoxygenase pathway, may be instrumental in prevention of postoperative ileus.


Asunto(s)
Araquidonato 12-Lipooxigenasa/fisiología , Araquidonato 15-Lipooxigenasa/fisiología , Quimiotaxis de Leucocito , Ácidos Docosahexaenoicos/fisiología , Ileus/inmunología , Yeyuno/inmunología , Músculo Liso/inmunología , Neutrófilos/inmunología , Complicaciones Posoperatorias/inmunología , Animales , Araquidonato 12-Lipooxigenasa/deficiencia , Araquidonato 15-Lipooxigenasa/deficiencia , Quimiotaxis de Leucocito/fisiología , Ácidos Docosahexaenoicos/biosíntesis , Ácidos Docosahexaenoicos/deficiencia , Ácidos Docosahexaenoicos/uso terapéutico , Evaluación Preclínica de Medicamentos , Emulsiones , Ácidos Grasos Omega-3/administración & dosificación , Ácidos Grasos Omega-3/uso terapéutico , Motilidad Gastrointestinal/efectos de los fármacos , Ileus/enzimología , Ileus/etiología , Ileus/prevención & control , Inflamación , Yeyuno/metabolismo , Yeyuno/patología , Ratones , Ratones Endogámicos C57BL , Modelos Inmunológicos , Músculo Liso/metabolismo , Músculo Liso/patología , Complicaciones Posoperatorias/enzimología , Complicaciones Posoperatorias/prevención & control , Organismos Libres de Patógenos Específicos
20.
Br J Pharmacol ; 172(20): 4864-74, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26227770

RESUMEN

BACKGROUND AND PURPOSE: As the pathogenesis of postoperative ileus (POI) involves inflammation and oxidative stress, comparable to ischaemia/reperfusion injury which can be ameliorated with nitrite, we investigated whether nitrite can protect against POI and explored the mechanisms involved. EXPERIMENTAL APPROACH: We used intestinal manipulation (IM) of the small intestine to induce POI in C57BL/6J mice. Sodium nitrite (48 nmol) was administered intravenously just before IM. Intestinal transit was assessed using fluorescent imaging. Bethanechol-stimulated jejunal circular muscle contractions were measured in organ baths. Inflammatory parameters, neutrophil infiltration, inducible NOS (iNOS) activity, reactive oxygen species (ROS) levels, mitochondrial complex I activity and cGMP were measured in the intestinal muscularis. KEY RESULTS: Pre-treatment with nitrite markedly improved the delay in intestinal transit and restored the reduced intestinal contractility observed 24 h following IM. This was accompanied by reduced protein levels of TNF-α, IL-6 and the chemokine CCL2, along with reduced iNOS activity and ROS levels. The associated neutrophil influx at 24 h was not influenced by nitrite. IM reduced mitochondrial complex I activity and cGMP levels; treatment with nitrite increased cGMP levels. Pre-treatment with the NO scavenger carboxy-PTIO or the soluble guanylyl cyclase inhibitor ODQ abolished nitrite-induced protective effects. CONCLUSIONS AND IMPLICATIONS: Exogenous nitrite deserves further investigation as a possible treatment for POI. Nitrite-induced protection of POI in mice was dependent on NO and this effect was not related to inhibition of mitochondrial complex I, but did involve activation of soluble guanylyl cyclase.


Asunto(s)
Ileus/tratamiento farmacológico , Complicaciones Posoperatorias/tratamiento farmacológico , Nitrito de Sodio/uso terapéutico , Animales , Quimiocina CCL2/metabolismo , GMP Cíclico/metabolismo , Tránsito Gastrointestinal/efectos de los fármacos , Ileus/inmunología , Ileus/metabolismo , Ileus/fisiopatología , Interleucina-6/metabolismo , Yeyuno/efectos de los fármacos , Yeyuno/metabolismo , Yeyuno/fisiología , Masculino , Ratones Endogámicos C57BL , Contracción Muscular/efectos de los fármacos , Neutrófilos/fisiología , Óxido Nítrico Sintasa de Tipo II/metabolismo , Complicaciones Posoperatorias/inmunología , Complicaciones Posoperatorias/metabolismo , Complicaciones Posoperatorias/fisiopatología , Especies Reactivas de Oxígeno/metabolismo , Nitrito de Sodio/sangre , Nitrito de Sodio/farmacocinética , Nitrito de Sodio/farmacología , Factor de Necrosis Tumoral alfa/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...