Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
J Cancer Res Clin Oncol ; 149(15): 14315-14327, 2023 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-37572121

RESUMO

BACKGROUND: Pancreatic ductal adenocarcinoma (PDAC) is one of the most aggressive types of cancer, characterized by the spreading of highly metastatic cancer cells, including invasion into surrounding nerves and perineural spaces. Nerves, in turn, can invade the tumor tissue and, through the secretion of neurotrophic factors, chemokines, and cytokines, contribute to PDAC progression. However, the contribution of the nerve-associated glial cells to PDAC progression is not well characterized. METHODS: Two murine PDAC cell lines were cultured with the conditioned media (CM) of primary enteric glial cells or IMS32 Schwann cells (SCs). Different properties of PDAC cells, such as invasiveness, migratory capacity, and resistance to gemcitabine, were measured by RT-qPCR, microscopy, and MTT assays. Using a neuronal cell line, the observed effects were confirmed to be specific to the glial lineage. RESULTS: Compared to the control medium, PDAC cells in the glial cell-conditioned medium showed increased invasiveness and migratory capacity. These cells showed reduced E-cadherin and increased N-cadherin and Vimentin levels, all markers of epithelial-mesenchymal transition (EMT). Primary enteric glial cell CM inhibited the proliferation of PDAC cells but preserved their viability, upregulated transcription factor Snail, and increased their resistance to gemcitabine. The conditioned medium generated from the IMS32 SCs produced comparable results. CONCLUSION: Our data suggest that glial cells can increase the metastatic potential of PDAC cells by increasing their migratory capacity and inducing epithelial-to-mesenchymal transition, a re-programming that many solid tumors use to undergo metastasis. Glial cell-conditioned medium also increased the chemoresistance of PDAC cells. These findings may have implications for future therapeutic strategies, such as targeting glial cell-derived factor signaling in PDAC.

2.
J Biosci ; 482023.
Artigo em Inglês | MEDLINE | ID: mdl-36785487

RESUMO

Enteric glial cells (EGCs) were shown to maintain the barrier integrity and immune homeostasis of the bowel. Postnatally, EGCs develop from progenitor cells located in the myenteric plexus and are continuously replenished through adulthood. Both, murine EGC development and replenishment were shown to depend on the microbiome. The underlying mechanisms are still unknown, and we hypothesized that the myeloid differentiation primary response protein 88 (Myd88) or toll-like receptor signaling pathways may be involved. Adult and neonatal C57BL/6 wild-type (wt) and Myd88-/- mice were housed under specific pathogen-free (SPF) or germ-free (GF) conditions. GF mice were further conventionalized by gavaging stools from, and cohousing with, SPF mice having intact microbiomes. The small bowels were harvested at various time points, and immunohistochemistry and qPCR analysis of EGC markers in the muscularis externa and mucosa were performed. In wt mice, after conventionalization, the glial cell-specific markers, glial fibrillary acidic protein (GFAP) and S100 calcium-binding protein ß (S100ß), were upregulated in the mucosa and muscularis externa. In Myd88-/- mice, this upregulation did not occur. Importantly, GFAP (only in the mucosa) and S100ß (in both the mucosa and muscularis externa) were significantly reduced in conventionalized Myd88-/- mice compared with the conventionalized wt mice. In neonatal mice, the gene expressions of GFAP and S100ß increased between the day of birth (P0) and postnatal day 15 (P15) in the mucosa and muscularis externa of both wt and Myd88-/- mice. Notably, in the mucosa but not the muscularis externa, at P15, the gene expressions of GFAP and S100ß were significantly reduced in Myd88-/-. Our data demonstrated that postnatal development and replenishment of EGCs require intestinal microbiota and depend on Myd88. The specific upstream mechanisms may involve toll-like-receptor recognition of the microbiota and will be the subject of further research.


Assuntos
Microbiota , Fator 88 de Diferenciação Mieloide , Camundongos , Animais , Fator 88 de Diferenciação Mieloide/genética , Fator 88 de Diferenciação Mieloide/metabolismo , Camundongos Endogâmicos C57BL , Neuroglia/metabolismo , Diferenciação Celular/genética
3.
Cells ; 11(16)2022 08 21.
Artigo em Inglês | MEDLINE | ID: mdl-36010681

RESUMO

Intestinal mucosal cells, such as resident macrophages and epithelial cells, express adrenergic receptors and are receptive to norepinephrine, the primary neurotransmitter of the sympathetic nervous system (SNS). It has been suggested that the SNS affects intestinal immune activity in conditions, such as inflammatory bowel disease; however, the underlying mechanisms remain ambiguous. Here, we investigated the effect of SNS on mucosal immune and epithelial cell functions. We employed 6-OHDA-induced sympathetic denervation (cSTX) to characterize muscularis-free mucosal transcriptomes by RNA-seq and qPCR, and quantified mucosal immune cells by flow cytometry. The role of norepinephrine and cytokines on epithelial functions was studied using small intestinal organoids. cSTX increased the presence of activated CD68+CD86+ macrophages and monocytes in the mucosa. In addition, through transcriptional profiling, the proinflammatory cytokines IL-1ß, TNF-α, and IFN-γ were induced, while Arg-1 and CD163 expression was reduced. Further, cSTX increased intestinal permeability in vivo and induced genes involved in barrier integrity and antimicrobial defense. In intestinal organoids, similar alterations were observed after treatment with proinflammatory cytokines, but not norepinephrine. We conclude that a loss in sympathetic input induces a proinflammatory mucosal state, leading to reduced epithelial barrier functioning and enhanced antimicrobial defense. This implies that the SNS might be required to maintain intestinal immune functions during homeostasis.


Assuntos
Doenças Inflamatórias Intestinais , Mucosa Intestinal , Citocinas/metabolismo , Células Epiteliais , Homeostase , Humanos , Mucosa Intestinal/metabolismo , Macrófagos/metabolismo
4.
Eur Surg Res ; 63(4): 224-231, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35184063

RESUMO

INTRODUCTION: Previous work of our group showed that lipoxygenase (LOX) pathways become activated upon surgical manipulation of the bowel wall and revealed a beneficial immune modulating role of the LOX-derived anti-inflammatory mediator protectin DX in postoperative ileus (POI). While we found a particular role of 12/15-LOX in the anti-inflammatory LOX action during POI, the role of 5-LOX, which produces the pro-inflammatory leukotriene B4 (LTB4), remained unknown. The purpose of this study was to investigate the role of 5-LOX within the pathogenesis of POI in a mouse model. METHODS: POI was induced by intestinal manipulation (IM) of the small bowel in C57BL/6, 5-LOX-/-, and CX3CR1GFP/+. Mice were either treated with a vehicle or with the synthetic 5-LOX antagonist zileuton or were left untreated. Cellular localization of 5-LOX and LTB4 release were visualized by immunofluorescence or ELISA, respectively. POI severity was quantified by gastrointestinal transit (GIT) and leukocyte extravasation into the muscularis externa (ME) by immunohistochemistry. RESULTS: 5-LOX expression was detected 24 h after IM within infiltrating leukocytes in the ME. LTB4 levels increased during POI in wild type but not in 5-LOX-/- after IM. POI was ameliorated in 5-LOX-/- as shown by decreased leukocyte numbers and normalized GIT. Zileuton normalized the postoperative GIT and reduced the numbers of infiltrating leukocytes into the ME. DISCUSSION/CONCLUSION: Our data demonstrate that 5-LOX and its metabolite LTB4 play a crucial role in POI. Genetic deficiency of 5-LOX and pharmacological antagonism by zileuton protected mice from POI. 5-LOX antagonism might be a promising target for prevention of POI in surgical patients.


Assuntos
Araquidonato 5-Lipoxigenase , Íleus , Camundongos , Animais , Leucotrieno B4 , Camundongos Endogâmicos C57BL , Íleus/tratamento farmacológico , Íleus/etiologia , Íleus/prevenção & controle , Complicações Pós-Operatórias/prevenção & controle
5.
Int J Mol Sci ; 22(13)2021 Jun 26.
Artigo em Inglês | MEDLINE | ID: mdl-34206766

RESUMO

Interactions between the peripheral nervous system and resident macrophages (MMs) modulate intestinal homeostatic functions. Activation of ß2-adrenergic receptors on MMs has been shown to reduce bacterial challenges. These MMs are also crucial for the development of bowel inflammation in postoperative ileus (POI), an iatrogenic, noninfectious inflammation-based motility disorder. However, the role of the sympathetic nervous system (SNS) in the immune modulation of these MMs during POI or other noninfectious diseases is largely unknown. By employing 6-OHDA-induced denervation, we investigated the changes in the muscularis externa by RNA-seq, quantitative PCR, and flow cytometry. Further, we performed transcriptional phenotyping of sorted CX3CR1+ MMs and ex vivo LPS/M-CSF stimulation on these MMs. By combining denervation with a mouse POI model, we explored distinct changes on CX3CR1+ MMs as well as in the muscularis externa and their functional outcome during POI. Our results identify SNS as an important mediator in noninfectious postoperative inflammation. Upon denervation, MMs anti-inflammatory genes were reduced, and the muscularis externa profile is shaped toward a proinflammatory status. Further, denervation reduced MMs anti-inflammatory genes also in the early phase of POI. Finally, reduced leukocyte infiltration into the muscularis led to a quicker recovery of bowel motility in the late phase of POI.


Assuntos
Pseudo-Obstrução Intestinal/imunologia , Macrófagos/imunologia , Sistema Nervoso Simpático/fisiopatologia , Animais , Receptor 1 de Quimiocina CX3C/metabolismo , Denervação/efeitos adversos , Pseudo-Obstrução Intestinal/etiologia , Leucócitos/imunologia , Fator Estimulador de Colônias de Macrófagos/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Músculo Liso/citologia
6.
EMBO Mol Med ; 13(1): e12724, 2021 01 11.
Artigo em Inglês | MEDLINE | ID: mdl-33332729

RESUMO

Enteric glial cells (EGC) modulate motility, maintain gut homeostasis, and contribute to neuroinflammation in intestinal diseases and motility disorders. Damage induces a reactive glial phenotype known as "gliosis", but the molecular identity of the inducing mechanism and triggers of "enteric gliosis" are poorly understood. We tested the hypothesis that surgical trauma during intestinal surgery triggers ATP release that drives enteric gliosis and inflammation leading to impaired motility in postoperative ileus (POI). ATP activation of a p38-dependent MAPK pathway triggers cytokine release and a gliosis phenotype in murine (and human) EGCs. Receptor antagonism and genetic depletion studies revealed P2X2 as the relevant ATP receptor and pharmacological screenings identified ambroxol as a novel P2X2 antagonist. Ambroxol prevented ATP-induced enteric gliosis, inflammation, and protected against dysmotility, while abrogating enteric gliosis in human intestine exposed to surgical trauma. We identified a novel pathogenic P2X2-dependent pathway of ATP-induced enteric gliosis, inflammation and dysmotility in humans and mice. Interventions that block enteric glial P2X2 receptors during trauma may represent a novel therapy in treating POI and immune-driven intestinal motility disorders.


Assuntos
Gliose , Neuroglia , Antagonistas do Receptor Purinérgico P2X/farmacologia , Animais , Citocinas , Inflamação , Intestino Delgado/fisiopatologia , Camundongos
7.
Sci Rep ; 10(1): 3457, 2020 Feb 21.
Artigo em Inglês | MEDLINE | ID: mdl-32081974

RESUMO

An amendment to this paper has been published and can be accessed via a link at the top of the paper.

8.
Front Immunol ; 11: 581111, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33519804

RESUMO

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.


Assuntos
Íleus/etiologia , Macrófagos/imunologia , Complicações Pós-Operatórias/etiologia , Receptor 3 Toll-Like/imunologia , Proteínas Adaptadoras de Transporte Vesicular/deficiência , Proteínas Adaptadoras de Transporte Vesicular/genética , Proteínas Adaptadoras de Transporte Vesicular/imunologia , Animais , Receptor 1 de Quimiocina CX3C/genética , Receptor 1 de Quimiocina CX3C/imunologia , Modelos Animais de Doenças , Feminino , Expressão Gênica , Íleus/imunologia , Íleus/patologia , Imunidade Inata , Macrófagos/classificação , Macrófagos/efeitos da radiação , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Plexo Mientérico/imunologia , Complicações Pós-Operatórias/imunologia , Complicações Pós-Operatórias/patologia , Tolerância a Radiação/imunologia , Receptor de Interferon alfa e beta/deficiência , Receptor de Interferon alfa e beta/genética , Receptor de Interferon alfa e beta/imunologia , Transdução de Sinais/imunologia , Receptor 3 Toll-Like/deficiência , Receptor 3 Toll-Like/genética , Quimeras de Transplante/imunologia
9.
Sci Rep ; 9(1): 10602, 2019 07 22.
Artigo em Inglês | MEDLINE | ID: mdl-31332247

RESUMO

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.


Assuntos
Proteínas de Ligação a DNA/metabolismo , Íleus/etiologia , Inflamassomos/metabolismo , Interleucina-1beta/metabolismo , Complicações Pós-Operatórias/etiologia , Animais , Microbioma Gastrointestinal , Íleus/imunologia , Íleus/metabolismo , Imunidade Inata , Interleucina-1alfa/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Complicações Pós-Operatórias/imunologia , Complicações Pós-Operatórias/metabolismo
10.
Neurogastroenterol Motil ; 31(3): e13501, 2019 03.
Artigo em Inglês | MEDLINE | ID: mdl-30406957

RESUMO

BACKGROUND: The cholinergic anti-inflammatory pathway comprises the perception of peripheral inflammation by afferent sensory neurons and reflex activation of efferent vagus nerve activity to regulate inflammation. Activation of this pathway was shown to reduce the inflammatory response and improve outcome of postoperative ileus (POI) and sepsis in rodents. Herein, we tested if a non-invasive auricular electrical transcutaneous vagus nerve stimulation (tVNS) affects inflammation in models of POI or endotoxemia. METHODS: Mice underwent tVNS or sham stimulation before and after induction of either POI by intestinal manipulation (IM) or endotoxemia by lipopolysaccharide administration. Some animals underwent a preoperative right cervical vagotomy. Neuronal activation of the solitary tract nucleus (NTS) and the dorsal motor nucleus of the vagus nerve (DMV) were analyzed by immunohistological detection of c-fos+ cells. Gene and protein expression of IL-6, MCP-1, IL-1ß as well as leukocyte infiltration and gastrointestinal transit were analyzed at different time points after IM. IL-6, TNFα, and IL-1ß serum levels were analyzed 3 hours after lipopolysaccharide administration. RESULTS: tVNS activated the NTS and DMV and reduced intestinal cytokine expression, reduced leukocyte recruitment to the manipulated intestine segment, and improved gastrointestinal transit after IM. Endotoxemia-induced IL-6 and TNF-α release was also reduced by tVNS. The protective effects of tVNS on POI and endotoxemia were abrogated by vagotomy. CONCLUSION: tVNS prevents intestinal and systemic inflammation. Activation of the DMV indicates an afferent to efferent central circuitry of the tVNS stimulation and the beneficial effects of tVNS depend on an intact vagus nerve. tVNS may become a non-invasive approach for treatment of POI.


Assuntos
Endotoxemia/prevenção & controle , Íleus/prevenção & controle , Complicações Pós-Operatórias/prevenção & controle , Estimulação Elétrica Nervosa Transcutânea/métodos , Estimulação do Nervo Vago/métodos , Animais , Citocinas/metabolismo , Endotoxemia/etiologia , Trânsito Gastrointestinal , Regulação da Expressão Gênica , Íleus/etiologia , Lipopolissacarídeos/toxicidade , Núcleo Mediodorsal do Tálamo/efeitos dos fármacos , Camundongos , Camundongos Endogâmicos C57BL , Núcleo Solitário/efeitos dos fármacos , Vagotomia
11.
PLoS One ; 13(4): e0195516, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29652914

RESUMO

AIM: To explore the effects of abdominal surgery and interleukin-1 signaling on antimicrobial defense in a model of postoperative ileus. METHODS: C57BL/6 and Interleukin-1 receptor type I (IL-1R1) deficient mice underwent intestinal manipulation to induce POI. Expression of mucosal IL-1α, IL-1ß and IL-1R1 and several antimicrobial peptides and enzymes were measured by quantitative PCR or ELISA, western blotting or immunohistochemistry. Bacterial overgrowth was determined by fluorescent in-situ hybridization and counting of jejunal luminal bacteria. Translocation of aerobic and anaerobic bacteria into the intestinal wall, mesenteric lymph nodes, liver and spleen was determined by counting bacterial colonies on agar plates 48h after plating of tissue homogenates. Antimicrobial activity against E. coli and B. vulgatus was analyzed in total and cationic fractions of small bowel mucosal tissue homogenates by a flow cytometry-based bacterial depolarization assay. RESULTS: Jejunal bacterial overgrowth was detected 24h after surgery. At the same time point, but not in the early phase 3h after surgery, bacterial translocation into the liver and mesenteric lymph nodes was observed. Increased antimicrobial activity against E. coli was induced within early phase of POI. Basal antimicrobial peptide and enzyme gene expression was higher in the ileal compared to the jejunal mucosa. The expression of lysozyme 1, cryptdin 1, cryptdin 4 and mucin 2 were reduced 24h after surgery in the ileal mucosa and mucin 2 was also reduced in the jejunum. Postoperative IL-1α and IL-1ß were increased in the postoperative mucosa. Deficiency of IL-1R1 affected the expression of antimicrobial peptides during homeostasis and POI. CONCLUSION: Small bowel antimicrobial capacity is disturbed during POI which is accompanied by bacterial overgrowth and translocation. IL-1R1 is partially involved in the gene expression of mucosal antimicrobial peptides. Altered small bowel antimicrobial activity may contribute also to POI development and manifestation in patients undergoing abdominal surgery.


Assuntos
Íleus/microbiologia , Mucosa Intestinal/microbiologia , Complicações Pós-Operatórias/microbiologia , Animais , Modelos Animais de Doenças , Íleus/metabolismo , Íleus/patologia , Mucosa Intestinal/metabolismo , Mucosa Intestinal/patologia , Masculino , Camundongos , Complicações Pós-Operatórias/metabolismo , Complicações Pós-Operatórias/patologia , Receptores Tipo I de Interleucina-1/metabolismo , Transdução de Sinais
12.
J Leukoc Biol ; 99(2): 231-9, 2016 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-26292977

RESUMO

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.


Assuntos
Araquidonato 12-Lipoxigenase/fisiologia , Araquidonato 15-Lipoxigenase/fisiologia , Quimiotaxia de Leucócito , Ácidos Docosa-Hexaenoicos/fisiologia , Íleus/imunologia , Jejuno/imunologia , Músculo Liso/imunologia , Neutrófilos/imunologia , Complicações Pós-Operatórias/imunologia , Animais , Araquidonato 12-Lipoxigenase/deficiência , Araquidonato 15-Lipoxigenase/deficiência , Quimiotaxia de Leucócito/fisiologia , Ácidos Docosa-Hexaenoicos/biossíntese , Ácidos Docosa-Hexaenoicos/deficiência , Ácidos Docosa-Hexaenoicos/uso terapêutico , Avaliação Pré-Clínica de Medicamentos , Emulsões , Ácidos Graxos Ômega-3/administração & dosagem , Ácidos Graxos Ômega-3/uso terapêutico , Motilidade Gastrointestinal/efeitos dos fármacos , Íleus/enzimologia , Íleus/etiologia , Íleus/prevenção & controle , Inflamação , Jejuno/metabolismo , Jejuno/patologia , Camundongos , Camundongos Endogâmicos C57BL , Modelos Imunológicos , Músculo Liso/metabolismo , Músculo Liso/patologia , Complicações Pós-Operatórias/enzimologia , Complicações Pós-Operatórias/prevenção & controle , Organismos Livres de Patógenos Específicos
13.
Methods Mol Biol ; 885: 213-32, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22565998

RESUMO

This chapter deals with a technique for isolating intact islets of Langerhans from the pig pancreas based on our experience performing approximately 750 isolations. The procedure we describe involves identification of an optimal donor pancreas, purification and in vitro culture of islets, diabetes induction in recipients, and transplantation of islets and their immunomodulation. Besides the sophistication of the technical equipment employed, the major factors influencing the isolation outcome are the pig breed, the number and morphology of the islets in the donor pancreas, the quality of the collagenase/neutral protease, and the skill of the team members.


Assuntos
Separação Celular/métodos , Transplante das Ilhotas Pancreáticas/métodos , Ilhotas Pancreáticas , Transplante Heterólogo/métodos , Animais , Sobrevivência Celular , Diabetes Mellitus Experimental/terapia , Feminino , Humanos , Ilhotas Pancreáticas/imunologia , Camundongos , Ratos , Suínos , Técnicas de Cultura de Tecidos , Coleta de Tecidos e Órgãos/métodos
14.
Transpl Immunol ; 26(4): 220-9, 2012 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-22430277

RESUMO

BACKGROUND: The phenomenon of T cell stimulation by MHC class II expressing (MHC II(pos)) CD4+ T cells has been intensively investigated for T cell clones but, so far, not for native T cells. The extensive use of T cell clones may explain the inconsistent outcomes of T cell-mediated antigen-presentation. Therefore, we used freshly isolated primed rat CD4+ T cells induced by immunisation with an allogeneic peptide P1, which is involved in allograft rejection. METHODS: MHC II(pos) and MHC II(neg) CD4+ T cells were isolated from popliteal lymph nodes of P1-immunised Lewis rats and were purified by combining depletion and positive selection steps. Purified MHC II(pos) CD4+ T cells and MHC II(neg) CD4+ T cells (105 cells per well each) were autostimulated or restimulated with P1-loaded (33 µg/ml peptide P1) and subsequently irradiated (with 20 Gy) autologous DC. RESULTS: Seven days after immunisation, a small population of MHC II(pos) CD4+ T cells was detectable (approximately 8.0% of total lymph node cells), as well as a large population of MHC II(neg) CD4+ T cells (up to 45%). Antigen-specific proliferation was observed for both T cell populations but only P1-loaded MHC II(pos) CD4+ T cells presented antigen presenting cell (APC) function for P1-primed T cells. Their inability to activate unprimed T cells may be due to impaired surface expression of costimulatory molecules (CD80 and CD86). CONCLUSION: Immunisation with the allogeneic peptide antigen P1 induced antigen-specific MHC II(pos) CD4+ rat T cells demonstrating perfect APC function for primed T cells in vitro.


Assuntos
Linfócitos T CD4-Positivos/imunologia , Células Dendríticas/imunologia , Rejeição de Enxerto/imunologia , Isoantígenos/imunologia , Fragmentos de Peptídeos/imunologia , Animais , Apresentação de Antígeno , Células Cultivadas , Citocinas/metabolismo , Imunização , Isoantígenos/metabolismo , Ativação Linfocitária , Fragmentos de Peptídeos/metabolismo , Ratos , Ratos Endogâmicos Lew
15.
Anesth Analg ; 114(2): 371-4, 2012 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-22025492

RESUMO

Renal failure is a common complication of critically ill patients. Colloids such as hydroxyethyl starch (HES), gelatin, or albumin are regularly used for intravascular volume resuscitation, but there are increasing reports about the nephrotoxic side effects of synthetic colloids in septic patients. Therefore, we investigated the influence of colloids (HES130/0.4 (Voluven®), gelatin (Gelafundin®), human albumin, and the crystalloid Sterofundin® ISO on cell viability of human proximal tubular (HK-2) cells. HK-2 cells were incubated with colloids (0.1%-4%) and with equivalent volumes of the crystalloid solution Sterofundin ISO. After 21 hours, cell viability of HK-2 cells was measured by EZ4U assay (dye XTT). Application of HES130/0.4 decreased cell viability significantly in a concentration-dependent manner (86.80% ± 10.79% by 0.5% HES down to 24.02% ± 4.27% by 4% HES). Human albumin (>1.25%) as well as gelatin (>1%) also showed deleterious effects on HK-2 cells. Interestingly, in lower concentrations, human albumin and the crystalloid solution Sterofundin ISO were cytoprotective in comparison with the NaCl control. In conclusion, synthetic and natural colloids showed a harmful impact on HK-2 cells in higher concentrations without any prior proinflammatory stimulus. HES130/0.4 exhibited the most distinctive harmful impact, whereas the application of crystalloid Sterofundin ISO revealed cytoprotective effects.


Assuntos
Albuminas/toxicidade , Gelatina/toxicidade , Derivados de Hidroxietil Amido/toxicidade , Soluções Isotônicas/toxicidade , Túbulos Renais Proximais/efeitos dos fármacos , Substitutos do Plasma/toxicidade , Linhagem Celular , Sobrevivência Celular/efeitos dos fármacos , Coloides , Soluções Cristaloides , Citoproteção , Relação Dose-Resposta a Droga , Humanos , Túbulos Renais Proximais/patologia , Compostos Orgânicos/toxicidade , Fatores de Tempo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...