Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 33
Filtrar
1.
Evid. actual. práct. ambul ; 23(1): e002045, 2020. tab
Artigo em Espanhol | LILACS | ID: biblio-1103172

RESUMO

Una mujer de 36 años, diagnosticada con síndrome de intestino irritable a predominio de diarrea (SII-D) acude a la consulta médica. Ella pregunta si el uso de probióticos sería útil para controlar los episodios de diarrea, ya que los fármacos con los que está siendo tratada no le resultan eficaces. Se realizó una búsqueda bibliográfica con el objetivo de en contrar evidencia en respuesta a su consulta, tras la cual se seleccionaron dos ensayos clínicos y una revisión sistemática. Se evidenciaron diversos resultados en cuanto al uso de probióticos en el SII-D y se discutieron los riesgos y beneficios del tratamiento, así como las implicancias en la vida de la paciente. (AU)


A 36-year-old woman diagnosed with diarrhea predominant irritable bowel syndrome (D-IBS) goes to meet the doctor. She raises whether the use of probiotics would be useful for controlling diarrhea episodes, since the drugs which she is being treated with, are not effective. A bibliographic search was conducted with the objective of finding evidence in response toher query. Two clinical trials and a systematic review were found. Variable results were found regarding the use of probioticsin D-IBS. The risks and benefits of the treatment were discussed, as well as the implications in the patient's lifestyle. (AU)


Assuntos
Humanos , Feminino , Adulto , Probióticos/uso terapêutico , Síndrome do Intestino Irritável/terapia , Diarreia/terapia , Parassimpatolíticos/uso terapêutico , Qualidade de Vida , Literatura de Revisão como Assunto , Dor Abdominal/terapia , Resina de Colestiramina/uso terapêutico , Ensaios Clínicos como Assunto , Probióticos/administração & dosagem , Probióticos/efeitos adversos , Síndrome do Intestino Irritável/diagnóstico , Síndrome do Intestino Irritável/etiologia , Diarreia/complicações , Duração da Terapia , Motilidade Gastrointestinal/imunologia , Mucosa Intestinal/imunologia , Loperamida/uso terapêutico , Antidepressivos/uso terapêutico
2.
Surgery ; 166(6): 1048-1054, 2019 12.
Artigo em Inglês | MEDLINE | ID: mdl-31543322

RESUMO

BACKGROUND: Intestinal inflammation is the predominant contributor to the genesis of postoperative ileus. Janus kinase 1 plays an important role during inflammation. Here, we investigated the role of Janus kinase 1 in postoperative ileus and whether inhibition of Janus kinase 1 could mitigate postoperative ileus. METHODS: A mouse model of postoperative ileus was induced by intestinal manipulation. Janus kinase 1 inhibitor GLPG0634 or placebo was administered orally before intestinal manipulation. At the indicated time points post operation, neutrophil infiltration was assessed by immunohistochemistry and enzyme-linked immunosorbent assay; proinflammatory gene expression was quantified by quantitative reverse-transcriptase polymerase chain reaction and enzyme-linked immunosorbent assay; and Janus kinase 1 activation was detected by Western blot. Functional studies were conducted to evaluate intestinal motility. RESULTS: We found that intestinal manipulation led to marked activation of Janus kinase 1, with increased proinflammatory gene expression and upregulated myeloperoxidase level. Moreover, intestinal manipulation resulted in an impairment of intestinal transit in vivo and inhibition of smooth muscle contractility in vitro. Preoperative administration of GLPG0634 markedly lowered the expression of proinflammatory cytokines, the myeloperoxidase level in the muscularis layer after bowel manipulation, and significantly ameliorated smooth muscle contractile function and intestinal transit ability. CONCLUSION: Our data showed that Janus kinase 1 activation mediated intestinal manipulation-induced resident macrophage activation after intestinal manipulation, and subsequent complex inflammatory cascade and gut dysmotility. Janus kinase 1 inhibition appears to be a prospective and convenient approach for the prevention of postoperative ileus.


Assuntos
Íleus/prevenção & controle , Janus Quinase 1/antagonistas & inibidores , Jejuno/cirurgia , Complicações Pós-Operatórias/prevenção & controle , Piridinas/administração & dosagem , Triazóis/administração & dosagem , Animais , Modelos Animais de Doenças , Motilidade Gastrointestinal/efeitos dos fármacos , Motilidade Gastrointestinal/imunologia , Humanos , Íleus/etiologia , Mediadores da Inflamação/metabolismo , Mucosa Intestinal/efeitos dos fármacos , Mucosa Intestinal/imunologia , Mucosa Intestinal/cirurgia , Janus Quinase 1/metabolismo , Jejuno/efeitos dos fármacos , Jejuno/imunologia , Masculino , Camundongos , Contração Muscular/efeitos dos fármacos , Contração Muscular/imunologia , Músculo Liso/efeitos dos fármacos , Músculo Liso/imunologia , Peroxidase/metabolismo , Complicações Pós-Operatórias/etiologia , Cuidados Pré-Operatórios/métodos , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/imunologia , Regulação para Cima/efeitos dos fármacos
3.
World J Gastroenterol ; 25(29): 3956-3971, 2019 Aug 07.
Artigo em Inglês | MEDLINE | ID: mdl-31413530

RESUMO

BACKGROUND: Irritable bowel syndrome (IBS) is a common chronic non-organic disease of the digestive system. Berberine (BBR) has been used to treat patients with IBS, but the underlying therapeutic mechanism is little understood. We believe that BBR achieves its therapeutic effect on IBS by preventing stress intestinal inflammation and visceral hypersensitivity and reducing bowel motility. AIM: To test the hypothesis that BBR achieves its therapeutic effect on IBS by preventing subclinical inflammation of the intestinal mucosa and reducing visceral hypersensitivity and intestinal motility. METHODS: IBS was induced in rats via water avoidance stress (WAS). qRT-PCR and histological analyses were used to evaluate the levels of cytokines and mucosal inflammation, respectively. Modified ELISA and qRT-PCR were used to evaluate the nuclear factor kappa-B (NF-κB) signal transduction pathway. Colorectal distention test, gastrointestinal transit measurement, Western blot, and qRT-PCR were used to analyze visceral sensitivity, intestinal motility, the expression of C-kit (marker of Cajal mesenchymal cells), and the expression of brain derived neurotrophic factor (BDNF) and its receptor TrkB. RESULTS: WAS led to mucosal inflammation, visceral hyperalgesia, and high intestinal motility. Oral administration of BBR inhibited the NF-κB signal transduction pathway, reduced the expression of pro-inflammatory cytokines [interleukin (IL)-1ß, IL-6, interferon-γ, and tumor necrosis factor-α], promoted the expression of anti-inflammatory cytokines (IL-10 and transforming growth factor-ß), and improved the terminal ileum tissue inflammation. BBR inhibited the expression of BDNF, TrkB, and C-kit in IBS rats, leading to the reduction of intestinal motility and visceral hypersensitivity. The therapeutic effect of BBR at a high dose (100 mg/kg) was superior to than that of the low-dose (25 mg/kg) group. CONCLUSION: BBR reduces intestinal mucosal inflammation by inhibiting the intestinal NF-κB signal pathway in the IBS rats. BBR reduces the expression of BDNF, its receptor TrkB, and C-kit. BBR also reduces intestinal motility and visceral sensitivity to achieve its therapeutic effect on IBS.


Assuntos
Berberina/farmacologia , Sistema Nervoso Entérico/efeitos dos fármacos , Hiperalgesia/tratamento farmacológico , Síndrome do Intestino Irritável/tratamento farmacológico , Estresse Psicológico/complicações , Animais , Berberina/uso terapêutico , Citocinas/imunologia , Citocinas/metabolismo , Modelos Animais de Doenças , Sistema Nervoso Entérico/fisiopatologia , Motilidade Gastrointestinal/efeitos dos fármacos , Motilidade Gastrointestinal/imunologia , Humanos , Hiperalgesia/fisiopatologia , Hiperalgesia/psicologia , Mucosa Intestinal/efeitos dos fármacos , Mucosa Intestinal/imunologia , Mucosa Intestinal/inervação , Síndrome do Intestino Irritável/imunologia , Síndrome do Intestino Irritável/psicologia , Masculino , Ratos , Estresse Psicológico/psicologia , Resultado do Tratamento
4.
World J Gastroenterol ; 25(30): 4092-4104, 2019 Aug 14.
Artigo em Inglês | MEDLINE | ID: mdl-31435166

RESUMO

Estrogens play important roles in the development and progression of multiple tumor types. Accumulating evidence points to the significance of estrogen action not only in tumors of hormonally regulated tissues such as the breast, endometrium and ovary, but also in the development of colorectal cancer (CRC). The effects of estrogens in physiological and pathophysiological conditions are mediated by the nuclear estrogen receptors α and ß, as well as the membrane-bound G protein-coupled estrogen receptor (GPER). The roles of GPER in CRC development and progression, however, remain poorly understood. Studies on the functions of GPER in the colon have shown that this estrogen receptor regulates colonic motility as well as immune responses in CRC-associated diseases, such as Crohn's disease and ulcerative colitis. GPER is also involved in cell cycle regulation, endoplasmic reticulum stress, proliferation, apoptosis, vascularization, cell migration, and the regulation of fatty acid and estrogen metabolism in CRC cells. Thus, multiple lines of evidence suggest that GPER may play an important role in colorectal carcinogenesis. In this review, we present the current state of knowledge regarding the contribution of GPER to colon function and CRC.


Assuntos
Colite Ulcerativa/patologia , Colo/fisiopatologia , Neoplasias Colorretais/patologia , Doença de Crohn/patologia , Receptores de Estrogênio/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Animais , Carcinogênese/imunologia , Carcinogênese/patologia , Colite Ulcerativa/imunologia , Colite Ulcerativa/fisiopatologia , Colo/imunologia , Colo/patologia , Neoplasias Colorretais/imunologia , Doença de Crohn/imunologia , Doença de Crohn/fisiopatologia , Modelos Animais de Doenças , Progressão da Doença , Estrogênios/metabolismo , Motilidade Gastrointestinal/imunologia , Humanos , Mucosa Intestinal/imunologia , Mucosa Intestinal/patologia , Mucosa Intestinal/fisiopatologia
5.
Am J Pathol ; 189(3): 540-551, 2019 03.
Artigo em Inglês | MEDLINE | ID: mdl-30593823

RESUMO

Inflammatory bowel diseases frequently cause gastrointestinal dysmotility, suggesting that they may also affect the enteric nervous system. So far, the precise mechanisms that lead to gastrointestinal dysmotility in inflammatory bowel diseases have not been elucidated. To determine the effect of CD8 T cells on gastrointestinal motility, transgenic mice expressing ovalbumin on enteric neurons were generated. In these mice, adoptive transfer of ovalbumin-specific OT-I CD8 T cells induced severe enteric ganglionitis. CD8 T cells homed to submucosal and myenteric plexus neurons, 60% of which were lost, clinically resulting in severely impaired gastrointestinal transition. Anti-interferon-γ treatment rescued neurons by preventing their up-regulation of major histocompatibility complex class I antigen, thus preserving gut motility. These preclinical murine data translated well into human gastrointestinal dysmotility. In a series of 30 colonic biopsy specimens from patients with gastrointestinal dysmotility, CD8 T cell-mediated ganglionitis was detected that was followed by severe loss of enteric neurons (74.8%). Together, the preclinical and clinical data support the concept that autoimmune CD8 T cells play an important pathogenetic role in gastrointestinal dysmotility and may destroy enteric neurons.


Assuntos
Doenças Autoimunes/imunologia , Linfócitos T CD8-Positivos/imunologia , Motilidade Gastrointestinal/imunologia , Doenças Inflamatórias Intestinais/imunologia , Plexo Mientérico/imunologia , Animais , Doenças Autoimunes/genética , Doenças Autoimunes/patologia , Linfócitos T CD8-Positivos/patologia , Motilidade Gastrointestinal/genética , Humanos , Doenças Inflamatórias Intestinais/genética , Doenças Inflamatórias Intestinais/patologia , Camundongos , Camundongos Transgênicos , Plexo Mientérico/patologia
6.
Front Immunol ; 9: 2599, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30581430

RESUMO

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.


Assuntos
Íleus/imunologia , Interleucina-10/imunologia , Intestinos/imunologia , Leucócitos/imunologia , Complicações Pós-Operatórias/imunologia , Animais , Modelos Animais de Doenças , Motilidade Gastrointestinal/imunologia , Humanos , Inflamação/imunologia , Macrófagos/imunologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos/imunologia , Monócitos/imunologia , Infiltração de Neutrófilos/imunologia , Neutrófilos/imunologia , Período Pós-Operatório
7.
Cell ; 175(2): 400-415.e13, 2018 10 04.
Artigo em Inglês | MEDLINE | ID: mdl-30173915

RESUMO

Macrophages are highly heterogeneous tissue-resident immune cells that perform a variety of tissue-supportive functions. The current paradigm dictates that intestinal macrophages are continuously replaced by incoming monocytes that acquire a pro-inflammatory or tissue-protective signature. Here, we identify a self-maintaining population of macrophages that arise from both embryonic precursors and adult bone marrow-derived monocytes and persists throughout adulthood. Gene expression and imaging studies of self-maintaining macrophages revealed distinct transcriptional profiles that reflect their unique localization (i.e., closely positioned to blood vessels, submucosal and myenteric plexus, Paneth cells, and Peyer's patches). Depletion of self-maintaining macrophages resulted in morphological abnormalities in the submucosal vasculature and loss of enteric neurons, leading to vascular leakage, impaired secretion, and reduced intestinal motility. These results provide critical insights in intestinal macrophage heterogeneity and demonstrate the strategic role of self-maintaining macrophages in gut homeostasis and intestinal physiology.


Assuntos
Intestinos/imunologia , Macrófagos/imunologia , Animais , Padronização Corporal/fisiologia , Diferenciação Celular/genética , Diferenciação Celular/imunologia , Motilidade Gastrointestinal/imunologia , Motilidade Gastrointestinal/fisiologia , Homeostase , Inflamação/imunologia , Mucosa Intestinal/imunologia , Intestino Delgado/metabolismo , Camundongos , Monócitos/metabolismo , Neurônios/metabolismo , Fagócitos/imunologia , Transcriptoma
8.
Ann Oncol ; 29(Suppl 4): iv126-iv142, 2018 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-29931177
9.
Am J Physiol Gastrointest Liver Physiol ; 315(1): G104-G116, 2018 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-29565641

RESUMO

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.


Assuntos
Motilidade Gastrointestinal/imunologia , Íleus , Laparotomia/efeitos adversos , Complicações Pós-Operatórias/imunologia , Canais de Cátion TRPM/metabolismo , Animais , Quimiocina CXCL2/metabolismo , Modelos Animais de Doenças , Íleus/etiologia , Íleus/imunologia , Inflamação/metabolismo , Interleucina-1beta/metabolismo , Macrófagos Peritoneais/metabolismo , Camundongos , Músculo Liso/metabolismo , Neutrófilos/metabolismo , Canais de Cátion TRPC/metabolismo
10.
Cell Immunol ; 330: 142-150, 2018 08.
Artigo em Inglês | MEDLINE | ID: mdl-29291892

RESUMO

Macrophages residing in the muscularis externa of the gastrointestinal tract are highly specialized cells that are essential for tissue homeostasis during steady-state conditions as well as during disease. They are characterized by their unique protective functional phenotype that is undoubtedly a consequence of the reciprocal interaction with their environment, including the enteric nervous system. This muscularis macrophage-neuron interaction dictates intestinal motility and promotes tissue-protection during injury and infection, but can also contribute to tissue damage in gastrointestinal disorders such as post-operative ileus and gastroparesis. Although the importance of muscularis macrophages is clearly recognized, different aspects of these cells remain largely unexplored such their origin, longevity and instructive signals that determine their function and phenotype. In this review, we will discuss the phenotype, functions and origin of muscularis macrophages during steady-state and disease conditions. We will highlight the bidirectional crosstalk with neurons and potential therapeutic strategies that target and manipulate muscularis macrophages to restore their protective signature as a treatment for disease.


Assuntos
Homeostase/imunologia , Enteropatias/imunologia , Intestinos/imunologia , Macrófagos/imunologia , Músculo Liso/imunologia , Animais , Sistema Nervoso Entérico/imunologia , Motilidade Gastrointestinal/imunologia , Humanos , Intestinos/inervação , Músculo Liso/citologia , Músculo Liso/inervação , Neurônios/imunologia
11.
Am J Physiol Gastrointest Liver Physiol ; 313(4): G320-G329, 2017 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-28684460

RESUMO

Inflammation plays a role in abdominal surgery (AS)-induced intestinal ileus that is alleviated by electrical vagal stimulation. Intracisternal injection of RX-77368, the stable thyrotropin-releasing hormone agonist, activates dorsal motor nucleus neurons and gastric vagal efferent discharges. We investigated the gastric inflammation induced by AS and the modulation by intracisternal RX-77368 in rats. RX-77368 (50 ng/rat) or saline was injected followed, 1 h later, by laparotomy and small intestinal/cecal manipulation. The sham group had anesthesia alone. After 6 h, gastric emptying (GE) and the inflammation in gastric corpus were determined. AS inhibited GE by 72% vs. control and doubled the number of M1-like macrophage immunoreactive for major histocompatibility complex class II (MHCII; M1 marker) but not for cluster of differentiation 206 (CD206; M2 marker) (MHCII+/CD206-) while there was no change in M2-like macrophages (MHCII-/CD206+). AS increased mRNA levels of interleukin-1ß (IL-1ß) and tumor necrosis factor α (TNF-α) by 1.7- and 1.5-fold, respectively, in the gastric submucosa plus muscle layers and the infiltration of neutrophils labeled by myeloperoxidase by 9.5-fold in the muscularis externa. RX-77368 inhibited AS-related gastric changes while not altering these parameters in the sham group. There was a significant negative correlation between GE and IL-1ß (r = -0.46), TNF-α (r = -0.44), M1 macrophage (r = -0.82), and neutrophils (r = -0.91). The M2-like macrophages and IL-10 expression were unchanged by AS with intracisternal saline or RX-77368. These data indicate that AS activates gastric M1 macrophages and increases proinflammatory cytokines expression, which are prevented by central vagal activation and may contribute to the correlated dampening of postoperative gastric ileus.NEW & NOTEWORTHY MHCII+/CD206- (M1) and MHCII-/CD206+ (M2) constitute two distinct populations of macrophages that are in close apposition to the cholinergic neurons in the rat gastric myenteric plexus (MP). Abdominal surgery (6 h) activates M1 macrophage leading to inflammation in the gastric MP correlated with the delayed gastric emptying, which was abolished by central vagal stimulation via intracisternal injection of RX-77368. Vagal stimulation linked with the cephalic phase may have potential beneficial effects to curtail postoperative gastric ileus.


Assuntos
Enterostomia/efeitos adversos , Motilidade Gastrointestinal/imunologia , Pseudo-Obstrução Intestinal/imunologia , Pseudo-Obstrução Intestinal/prevenção & controle , Ativação de Macrófagos/imunologia , Plexo Mientérico/fisiopatologia , Nervo Vago/fisiopatologia , Animais , Motilidade Gastrointestinal/efeitos dos fármacos , Pseudo-Obstrução Intestinal/etiologia , Ativação de Macrófagos/efeitos dos fármacos , Masculino , Plexo Mientérico/efeitos dos fármacos , Complexo Mioelétrico Migratório/efeitos dos fármacos , Complexo Mioelétrico Migratório/imunologia , Ácido Pirrolidonocarboxílico/administração & dosagem , Ácido Pirrolidonocarboxílico/análogos & derivados , Ratos , Ratos Sprague-Dawley , Hormônio Liberador de Tireotropina/administração & dosagem , Hormônio Liberador de Tireotropina/análogos & derivados , Resultado do Tratamento , Nervo Vago/efeitos dos fármacos
12.
Neurogastroenterol Motil ; 27(6): 743-9, 2015 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-26011782

RESUMO

The persistent phase of postoperative ileus (POI) is mediated by inflammatory activation of the resident myeloid immune cell population in the gut wall, likely elicited by neurogenic activation. Mast cells are thought to play a critical role in this inflammatory response and involvement of mast cells in POI has been investigated and described thoroughly in experimental studies. Intestinal manipulation (IM) leads to degranulation of mast cells, resulting in an increase in mast cell proteases in peritoneal fluid and gut tissue. The inflammatory infiltrate formed in the intestinal wall thereby impairs gastrointestinal motility. In the clinical study by Berdun et al., the experimentally known association between mast cell degranulation and delayed motility is shown in a clinical setting. These findings are important and open up therapeutic opportunities to reduce or prevent POI. In this mini-review, the role of mast cells in POI is discussed. Furthermore, an update is given on the involvement of the inflammatory response in POI and potential therapeutic strategies.


Assuntos
Degranulação Celular/imunologia , Procedimentos Cirúrgicos do Sistema Digestório , Motilidade Gastrointestinal/imunologia , Íleus/imunologia , Mastócitos/imunologia , Complicações Pós-Operatórias/imunologia , Humanos , Inflamação , Laparoscopia
13.
Gastroenterology ; 146(1): 176-87.e1, 2014 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-24067878

RESUMO

BACKGROUND & AIMS: Postoperative ileus (POI) is a common consequence of abdominal surgery that increases the risk of postoperative complications and morbidity. We investigated the cellular mechanisms and immune responses involved in the pathogenesis of POI. METHODS: We studied a mouse model of POI in which intestinal manipulation leads to inflammation of the muscularis externa and disrupts motility. We used C57BL/6 (control) mice as well as mice deficient in Toll-like receptors (TLRs) and cytokine signaling components (TLR-2(-/-), TLR-4(-/-), TLR-2/4(-/-), MyD88(-/-), MyD88/TLR adaptor molecule 1(-/-), interleukin-1 receptor [IL-1R1](-/-), and interleukin (IL)-18(-/-) mice). Bone marrow transplantation experiments were performed to determine which cytokine receptors and cell types are involved in the pathogenesis of POI. RESULTS: Development of POI did not require TLRs 2, 4, or 9 or MyD88/TLR adaptor molecule 2 but did require MyD88, indicating a role for IL-1R1. IL-1R1(-/-) mice did not develop POI; however, mice deficient in IL-18, which also signals via MyD88, developed POI. Mice given injections of an IL-1 receptor antagonist (anakinra) or antibodies to deplete IL-1α and IL-1ß before intestinal manipulation were protected from POI. Induction of POI activated the inflammasome in muscularis externa tissues of C57BL6 mice, and IL-1α and IL-1ß were released in ex vivo organ bath cultures. In bone marrow transplantation experiments, the development of POI required activation of IL-1 receptor in nonhematopoietic cells. IL-1R1 was expressed by enteric glial cells in the myenteric plexus layer, and cultured primary enteric glia cells expressed IL-6 and the chemokine monocyte chemotactic protein 1 in response to IL-1ß stimulation. Immunohistochemical analysis of human small bowel tissue samples confirmed expression of IL-1R1 in the ganglia of the myenteric plexus. CONCLUSIONS: IL-1 signaling, via IL-1R1 and MyD88, is required for development of POI after intestinal manipulation in mice. Agents that interfere with the IL-1 signaling pathway are likely to be effective in the treatment of POI.


Assuntos
Motilidade Gastrointestinal/imunologia , Íleus/imunologia , Interleucina-1/imunologia , Músculo Liso/imunologia , Fator 88 de Diferenciação Mieloide/imunologia , Plexo Mientérico/imunologia , Neuroglia/imunologia , Complicações Pós-Operatórias/imunologia , Receptores Tipo I de Interleucina-1/imunologia , Animais , Modelos Animais de Doenças , Íleus/metabolismo , Interleucina-1/metabolismo , Interleucina-18/genética , Interleucina-18/imunologia , Interleucina-18/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Músculo Liso/metabolismo , Fator 88 de Diferenciação Mieloide/genética , Fator 88 de Diferenciação Mieloide/metabolismo , Plexo Mientérico/metabolismo , Neuroglia/metabolismo , Complicações Pós-Operatórias/metabolismo , Receptores Tipo I de Interleucina-1/genética , Receptores Tipo I de Interleucina-1/metabolismo , Transdução de Sinais , Receptores Toll-Like/genética , Receptores Toll-Like/imunologia , Receptores Toll-Like/metabolismo
14.
J Vis Exp ; (67)2012 Sep 11.
Artigo em Inglês | MEDLINE | ID: mdl-22990580

RESUMO

Inflammation of the gastrointestinal tract is a common reason for a variety of human diseases. Animal research models are critical in investigating the complex cellular and molecular of intestinal pathology. Although the tunica mucosa is often the organ of interest in many inflammatory diseases, recent works demonstrated that the muscularis externa (ME) is also a highly immunocompetent organ that harbours a dense network of resident immunocytes.(1,2) These works were performed within the standardized model of intestinal manipulation (IM) that leads to inflammation of the bowel wall, mainly limited to the ME. Clinically this inflammation leads to prolonged intestinal dysmotility, known as postoperative ileus (POI) which is a frequent and unavoidable complication after abdominal surgery.(3) The inflammation is characterized by liberation of proinflammatory mediators such as IL-6(4) or IL-1ß or inhibitory neurotransmitters like nitric oxide (NO).(5) Subsequently, tremendous numbers of immunocytes extravasate into the ME, dominated by polymorphonuclear neutrophils (PMN) and monocytes and finally maintain POI.(2) Lasting for days, this intestinal paralysis leads to an increased risk of aspiration, bacterial translocation and infectious complications up to sepsis and multi organ failure and causes a high economic burden.(6) In this manuscript we demonstrate the standardized model of IM and in vivo assessment of gastrointestinal transit (GIT) and colonic transit. Furthermore we demonstrate a method for separation of the ME from the tunica mucosa followed by immunological analysis, which is crucial to distinguish between the inflammatory responses in these both highly immunoactive bowel wall compartments. All analyses are easily transferable to any other research models, affecting gastrointestinal function.


Assuntos
Gastroenterite/fisiopatologia , Motilidade Gastrointestinal/fisiologia , Animais , Gastroenterite/imunologia , Gastroenterite/patologia , Motilidade Gastrointestinal/imunologia , Mucosa Intestinal/imunologia , Mucosa Intestinal/patologia , Mucosa Intestinal/fisiopatologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Modelos Animais
15.
Scand J Gastroenterol ; 47(10): 1165-73, 2012 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-22835010

RESUMO

OBJECTIVE: Many patients, especially women, suffer from severe gastrointestinal pain and dysmotility for several years without being diagnosed. Depletion of gonadotropin-releasing hormone (GnRH) in the enteric nervous system (ENS) has been described in some patients. The aim of this study was to examine the expression of GnRH in ENS and antibodies against GnRH in serum, in a dysmotility patient cohort of southern Sweden. MATERIALS AND METHODS: All consecutive patients (n = 35) referred for laparoscopic full-thickness biopsy because of symptoms or signs of severe dysmotility between 1998 and 2009, or patients with a severe dysmotility disorder having had a bowel resection within the time frame, were considered for inclusion. In 22 cases, representative biopsy material containing ganglia was available, and these patients were included. Medical records were scrutinized. The expression of GnRH was determined by immunohistochemistry in bowel biopsies from these patients and in patients with carcinoma or diverticulosis without ENS histopathology. Antibodies against GnRH in serum were determined by ELISA in patients and controls. RESULTS: 14 patients were diagnosed with enteric dysmotility (ED) and 8 with chronic intestinal pseudo-obstruction due to varying etiology. Immunostained biopsies showed expression of GnRH in the ENS. A reduced expression of GnRH-containing neurons was found in 5 patients, as well as antibodies against GnRH in serum. 3 of these patients had a history of in vitro fertilization (IVF) using GnRH analogs. CONCLUSIONS: A subgroup of patients with severe dysmotility had a reduced expression of GnRH-containing neurons in the ENS and expressed antibodies against GnRH in serum.


Assuntos
Carcinoma/complicações , Divertículo/complicações , Sistema Nervoso Entérico , Motilidade Gastrointestinal/imunologia , Hormônio Liberador de Gonadotropina/imunologia , Neoplasias Intestinais/complicações , Pseudo-Obstrução Intestinal , Adulto , Anticorpos/sangue , Biópsia , Carcinoma/patologia , Doença Crônica , Divertículo/patologia , Sistema Nervoso Entérico/imunologia , Sistema Nervoso Entérico/patologia , Feminino , Fertilização in vitro/efeitos adversos , Humanos , Imuno-Histoquímica , Neoplasias Intestinais/patologia , Pseudo-Obstrução Intestinal/imunologia , Pseudo-Obstrução Intestinal/patologia , Pseudo-Obstrução Intestinal/fisiopatologia , Intestinos/imunologia , Intestinos/inervação , Intestinos/patologia , Masculino , Fatores Desencadeantes , Índice de Gravidade de Doença
16.
Arthritis Rheum ; 63(5): 1426-34, 2011 May.
Artigo em Inglês | MEDLINE | ID: mdl-21312189

RESUMO

OBJECTIVE: In primary Sjögren's syndrome (SS), impairment of the gastrointestinal (GI) tract is common, and includes reduced esophageal motor function, delayed gastric emptying, and abnormalities in colonic motility; the pathogenesis is as yet unknown. We undertook this study to investigate the role of functional antibodies to the type 3 muscarinic receptor (M3R) in GI dysfunction associated with primary SS. METHODS: Muscle strip and whole-organ functional assays were used to determine whether IgG with anti-M3R activity from patients with primary SS disrupted neurotransmission in tissue from throughout the mouse GI tract. Specificity of the autoantibody for the M3R was determined using knockout mice that were deficient in the expression of muscarinic receptor subtypes. RESULTS: Functional antibodies to the M3R inhibited neuronally mediated contraction of smooth muscle from throughout the GI tract and disrupted complex contractile motility patterns in the colon. The autoantibodies were not active on tissue from mice that lacked the M3R, providing compelling evidence of the direct interaction of patient autoantibodies with the M3R. CONCLUSION: Our results indicate that anti-M3R autoantibodies have the potential to mediate multiple dysfunctions of the GI tract in primary SS, ranging from reduced esophageal motor activity to altered colonic motility. We hypothesize that altered GI motility forms part of a broader autonomic dysfunction mediated by pathogenic anti-M3R autoantibodies in primary SS.


Assuntos
Autoanticorpos/imunologia , Motilidade Gastrointestinal/imunologia , Receptor Muscarínico M3/imunologia , Síndrome de Sjogren/imunologia , Transmissão Sináptica/imunologia , Acetilcolina/metabolismo , Análise de Variância , Animais , Protocolos de Quimioterapia Combinada Antineoplásica , Carbacol/farmacologia , Agonistas Colinérgicos/farmacologia , Cisplatino , Motilidade Gastrointestinal/efeitos dos fármacos , Humanos , Ifosfamida , Masculino , Camundongos , Camundongos Knockout , Mitomicina , Contração Muscular/efeitos dos fármacos , Contração Muscular/imunologia , Músculo Liso/efeitos dos fármacos , Músculo Liso/imunologia , Receptor Muscarínico M3/metabolismo , Síndrome de Sjogren/metabolismo , Síndrome de Sjogren/fisiopatologia , Transmissão Sináptica/efeitos dos fármacos
17.
J Immunol ; 183(10): 6744-53, 2009 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-19846874

RESUMO

Endotoxin-mediated ileus is poorly understood. Our objective was to mechanistically investigate the role of cell-specific TLR4 expression/signaling in causing gastrointestinal dysmotility. TLR4 chimeras and CSF-1-dependent macrophage-deficient mice were subjected to i.p. ultrapure (UP)-LPS (5 mg/kg). At 6 h, gastric emptying and gastrointestinal transit assessed in vivo motility, and jejunal circular muscle contractility was measured in vitro. Muscularis infiltration of neutrophils and monocytes were counted, and intestinal muscularis inflammatory mediators were quantified by quantitative PCR. Demonstrating TLR4 dependency, UP-LPS-induced gastric stasis and ileus of TLR4(WT) mice were absent in mutant TLR4(LPS-d) mice. Unexpectedly, engraftment of TLR4-mutant bone marrow into TLR4-competent mice (bmTLR4(LPS-d)/TLR4(WT)) exhibited a significant transit delay to UP-LPS similar to bmTLR4(WT)/TLR4(WT) mice. CSF-1(-/-) mice were not protected from ileus. Contrary, UP-LPS-treated bmTLR4(WT)/TLR4(LPS-d) and bmTLR4(LPS-d)/TLR4(LPS-d) mice had normal transit. No leukocytic infiltration was detected at 6 h. Spontaneous jejunal contractions were markedly suppressed in UP-LPS-treated TLR4-competent mice, but bethanechol-stimulated contractions were not altered by UP-LPS in any group. UP-LPS-induced inflammatory mRNAs in a TLR4-dependent manner, but TLR4 mRNA itself was not significantly altered. In chimera mice, UP-LPS induction of IL-1beta and IL-10 were hemopoietic dependent, and GM-CSF was nonhemopoietic dependent, whereas IL-6 and inducible NO synthase were derived from both cell types. Hemopoietic and nonhemopoietic cells contribute to TLR4-sensitive muscularis inflammatory signaling, but nonhemopoietic TLR4 signaling plays an exclusive primary role in causing functional UP-LPS-induced gastric stasis and ileus. Direct LPS suppression of spontaneous contractility participates in mediating early TLR4-transduced dysmotility.


Assuntos
Íleus/imunologia , Intestinos/imunologia , Fator Estimulador de Colônias de Macrófagos/imunologia , Macrófagos/imunologia , Neutrófilos/imunologia , Receptor 4 Toll-Like/imunologia , Animais , Motilidade Gastrointestinal/efeitos dos fármacos , Motilidade Gastrointestinal/imunologia , Fator Estimulador de Colônias de Granulócitos e Macrófagos/imunologia , Fator Estimulador de Colônias de Granulócitos e Macrófagos/metabolismo , Íleus/metabolismo , Interleucina-10/imunologia , Interleucina-10/metabolismo , Interleucina-1beta/imunologia , Interleucina-1beta/metabolismo , Interleucina-6/imunologia , Interleucina-6/metabolismo , Mucosa Intestinal/metabolismo , Intestinos/efeitos dos fármacos , Jejuno/efeitos dos fármacos , Jejuno/imunologia , Jejuno/metabolismo , Lipopolissacarídeos/farmacologia , Fator Estimulador de Colônias de Macrófagos/genética , Fator Estimulador de Colônias de Macrófagos/metabolismo , Macrófagos/efeitos dos fármacos , Macrófagos/metabolismo , Camundongos , Camundongos Endogâmicos C3H , Camundongos Knockout , Contração Muscular/efeitos dos fármacos , Contração Muscular/imunologia , Neutrófilos/efeitos dos fármacos , Neutrófilos/metabolismo , Óxido Nítrico Sintase Tipo II/imunologia , Óxido Nítrico Sintase Tipo II/metabolismo , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/imunologia , Receptor 4 Toll-Like/genética
18.
Gut ; 58(9): 1300-11, 2009 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-19671558

RESUMO

Postoperative ileus (POI) is a common clinical condition arising after almost every abdominal surgical procedure, leading to increased patient morbidity and prolonged hospitalisation. Recent advances in insight into the underlying pathophysiology have identified intestinal inflammation triggered by handling of the intestine as the main mechanism. Not only does the local inflammatory process compromise the contractile activity of the handled intestine, but it also activates inhibitory neural pathways and possibly triggers inflammation at distant untouched areas, leading to a generalised impairment of gastrointestinal motility. Macrophages residing in the muscularis externa and mast cells are the key players in this inflammatory cascade. Pharmacological interventions preventing the activation of these immune cells reduce the influx of leucocytes into the intestine, an effect associated with a reduction of the duration of POI. New potential therapeutic strategies to shorten POI based on these new insights will undoubtedly enter the clinical arena soon.


Assuntos
Íleus/imunologia , Complicações Pós-Operatórias/imunologia , Animais , Anti-Inflamatórios/uso terapêutico , Sistema Nervoso Entérico/fisiopatologia , Motilidade Gastrointestinal/imunologia , Humanos , Íleus/tratamento farmacológico , Íleus/fisiopatologia , Macrófagos/imunologia , Mastócitos/imunologia
19.
Gastroenterol Clin North Am ; 37(2): 397-410, vi-vii, 2008 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-18499027

RESUMO

Increasing evidence suggests that a subset of gastrointestinal motility disorders is associated with the presence of circulating antibodies. These antibodies are directed against various molecular targets, the best known being anti-neuronal nuclear antibody (ANNA-1 or anti-Hu) associated with paraneoplastic motility disorders. There is also evidence that the presence of distinct autoantibody profiles is associated with non-paraneoplastic motility disorders. This review focuses on the types of antibodies associated with gastrointestinal motility disorders and the significance of these antibodies. Algorithms are suggested for the work-up and treatment of patients with circulating antibodies associated with gastrointestinal motility disorders.


Assuntos
Autoanticorpos/metabolismo , Motilidade Gastrointestinal/imunologia , Neoplasias Intestinais/imunologia , Síndromes Paraneoplásicas/imunologia , Autoanticorpos/imunologia , Motilidade Gastrointestinal/fisiologia , Humanos , Neoplasias Intestinais/complicações
20.
Scand J Gastroenterol ; 42(11): 1289-93, 2007 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-17918010

RESUMO

OBJECTIVE: Enteric neuropathy with mild inflammation (ganglionitis) has been described in several motility disorders including irritable bowel syndrome (IBS), enteric dysmotility (ED), slow-transit constipation (STC) and chronic intestinal pseudo-obstruction (CIPO). The purpose of this study was to test the hypothesis that autoantibodies directed against specific neural antigens including ion channels may be associated with this finding. MATERIAL AND METHODS: Comprehensive routine and immunohistochemical analyses of full-thickness jejunal laparoscopic biopsies were performed on patients fulfilling the international criteria for IBS, ED, STC and CIPO. Patients with ganglionitis had sera screened for specific antibodies to voltage-gated calcium channels (VGCCs) of P/Q- and N-type, voltage-gated potassium channels (VGKCs), glutamic acid decarboxylase (GAD) and neuronal alpha3-AChR by validated immunoprecipitation assays. RESULTS: Thirty-three patients were included in the study. Two of them, both with IBS, were found to have positive antibody titres. One had anti-VGKC antibodies and one had anti-alpha3-AChR antibodies. No antibodies were detected in GAD or VGCCs (case reports presented). CONCLUSIONS: A small proportion of patients with inflammatory enteric neuropathy have antibodies directed towards neuronal ion channels. The pathogenic role of such antibodies requires determination but may represent a possible aetiology of severe functional symptoms in these groups of patients.


Assuntos
Autoanticorpos/sangue , Doenças Autoimunes/imunologia , Enteropatias/imunologia , Doenças do Sistema Nervoso/imunologia , Adulto , Idoso , Biópsia , Canais de Cálcio/imunologia , Constipação Intestinal/imunologia , Feminino , Motilidade Gastrointestinal/imunologia , Glutamato Descarboxilase/imunologia , Humanos , Imunoprecipitação/métodos , Síndrome do Intestino Irritável/imunologia , Jejuno/patologia , Masculino , Pessoa de Meia-Idade , Canais de Potássio de Abertura Dependente da Tensão da Membrana/imunologia , Receptores Colinérgicos/imunologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA