Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 27
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Mikrochim Acta ; 191(10): 570, 2024 09 02.
Artículo en Inglés | MEDLINE | ID: mdl-39218927

RESUMEN

Loofah sponge-like carbon nanofibers (LF-Co,N/CNFs) were utilized as a carrier for Ru(bpy)32+, and then combined with CdS to create a novel solid-state electrochemiluminescence sensor capable of detecting trace amounts of fenpropathrin. LF-Co,N/CNFs, obtained through the high-temperature pyrolysis of ZIF-67 coaxial electrospinning fibers, were characterized by a loofah-like morphology and exhibited a significant specific surface area and porosity. Apart from serving as a carrier, LF-Co,N/CNFs also functioned as a luminescence accelerator, enhancing the system's luminescence efficiency by facilitating electron transmission and reducing the transmission distance. The inclusion of CdS in the luminescence reaction, in conjunction with Ru(bpy)32+, further boosted the sensor's luminescence signal. The resulting sensor demonstrated a satisfactory signal, with fenpropathrin causing significant quenching of the ECL signal. Under optimized conditions, a linear relationship between the signal quench value and fenpropathrin concentration in the range 1 × 10-12 to 1 × 10-6 M was observed, with a detection limit of 3.3 × 10-13 M (S/N = 3). This developed sensor is characterized by its simplicity, sensitivity, and successful application in detecting fenpropathrin in real samples. The study not only presents a straightforward detection platform for fenpropathrin but also introduces new avenues for the rapid determination of various food contaminants, thereby expanding the utility of carbon fibers in electrochemiluminescence sensors.


Asunto(s)
Carbono , Técnicas Electroquímicas , Límite de Detección , Mediciones Luminiscentes , Nanofibras , Nanofibras/química , Mediciones Luminiscentes/métodos , Carbono/química , Técnicas Electroquímicas/métodos , Técnicas Electroquímicas/instrumentación , Animales , Contaminación de Alimentos/análisis , Compuestos de Cadmio/química , Piretrinas/análisis , Compuestos Organometálicos
2.
Proc Natl Acad Sci U S A ; 116(18): 9125-9134, 2019 04 30.
Artículo en Inglés | MEDLINE | ID: mdl-30979808

RESUMEN

Carbamoyl phosphate synthetase-1 (CPS1) is the major mitochondrial urea cycle enzyme in hepatocytes. It is released into mouse and human blood during acute liver injury, where is has a short half-life. The function of CPS1 in blood and the reason for its short half-life in serum are unknown. We show that CPS1 is released normally into mouse and human bile, and pathologically into blood during acute liver injury. Other cytoplasmic and mitochondrial urea cycle enzymes are also found in normal mouse bile. Serum, bile, and purified CPS1 manifest sedimentation properties that overlap with extracellular vesicles, due to the propensity of CPS1 to aggregate despite being released primarily as a soluble protein. During liver injury, CPS1 in blood is rapidly sequestered by monocytes, leading to monocyte M2-polarization and homing to the liver independent of its enzyme activity. Recombinant CPS1 (rCPS1), but not control r-transferrin, increases hepatic macrophage numbers and phagocytic activity. Notably, rCPS1 does not activate hepatic macrophages directly; rather, it activates bone marrow and circulating monocytes that then home to the liver. rCPS1 administration prevents mouse liver damage induced by Fas ligand or acetaminophen, but this protection is absent in macrophage-deficient mice. Moreover, rCPS1 protects from acetaminophen-induced liver injury even when given therapeutically after injury induction. In summary, CPS1 is normally found in bile but is released by hepatocytes into blood upon liver damage. We demonstrate a nonenzymatic function of CPS1 as an antiinflammatory protective cytokine during acute liver injury.


Asunto(s)
Lesión Pulmonar Aguda/metabolismo , Ácidos y Sales Biliares/metabolismo , Carbamoil-Fosfato Sintasa (Amoniaco)/metabolismo , Acetaminofén/metabolismo , Lesión Pulmonar Aguda/enzimología , Adulto , Animales , Bilis/metabolismo , Citocinas/metabolismo , Proteína Ligando Fas/metabolismo , Femenino , Hepatocitos/metabolismo , Humanos , Hígado/metabolismo , Hepatopatías , Macrófagos/metabolismo , Masculino , Ratones , Mitocondrias/metabolismo
3.
J Pediatr Surg ; 54(3): 511-516, 2019 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-29753524

RESUMEN

BACKGROUND: Although serial transverse enteroplasty (STEP) improves function of dilated short bowel, a significant proportion of patients require repeat surgery. To address underlying reasons for unsuccessful STEP, we compared small intestinal mucosal characteristics between initial and repeat STEP procedures in children with short bowel syndrome (SBS). METHODS: Fifteen SBS children, who underwent 13 first and 7 repeat STEP procedures with full thickness small bowel samples at median age 1.5 years (IQR 0.7-3.7) were included. The specimens were analyzed histologically for mucosal morphology, inflammation and muscular thickness. Mucosal proliferation and apoptosis was analyzed with MIB1 and Tunel immunohistochemistry. RESULTS: Median small bowel length increased 42% by initial STEP and 13% by repeat STEP (p=0.05), while enteral caloric intake increased from 6% to 36% (p=0.07) during 14 (12-42) months between the procedures. Abnormal mucosal inflammation was frequently observed both at initial (69%) and additional STEP (86%, p=0.52) surgery. Villus height, crypt depth, enterocyte proliferation and apoptosis as well as muscular thickness were comparable at first and repeat STEP (p>0.05 for all). Patients, who required repeat STEP tended to be younger (p=0.057) with less apoptotic crypt cells (p=0.031) at first STEP. Absence of ileocecal valve associated with increased intraepithelial leukocyte count and reduced crypt cell proliferation index (p<0.05 for both). CONCLUSIONS: No adaptive mucosal hyperplasia or muscular alterations occurred between first and repeat STEP. Persistent inflammation and lacking mucosal growth may contribute to continuing bowel dysfunction in SBS children, who require repeat STEP procedure, especially after removal of the ileocecal valve. LEVEL OF EVIDENCE: Level IV, retrospective study.


Asunto(s)
Procedimientos Quirúrgicos del Sistema Digestivo/efectos adversos , Mucosa Intestinal/patología , Intestino Delgado/cirugía , Síndrome del Intestino Corto/patología , Apoptosis , Niño , Preescolar , Procedimientos Quirúrgicos del Sistema Digestivo/métodos , Humanos , Inmunohistoquímica , Lactante , Inflamación/patología , Intestino Delgado/patología , Reoperación/efectos adversos , Estudios Retrospectivos , Síndrome del Intestino Corto/cirugía
4.
Cell Mol Gastroenterol Hepatol ; 3(3): 447-468, 2017 May.
Artículo en Inglés | MEDLINE | ID: mdl-28462383

RESUMEN

BACKGROUND & AIMS: Total parenteral nutrition (TPN), a crucial treatment for patients who cannot receive enteral nutrition, is associated with mucosal atrophy, barrier dysfunction, and infectious complications. Glucagon-like peptide-2 (GLP-2) and epidermal growth factor (EGF) improve intestinal epithelial cell (IEC) responses and attenuate mucosal atrophy in several TPN models. However, it remains unclear whether these 2 factors use distinct or overlapping signaling pathways to improve IEC responses. We investigated the interaction of GLP-2 and EGF signaling in a mouse TPN model and in patients deprived of enteral nutrition. METHODS: Adult C57BL/6J, IEC-Egfrknock out (KO) and IEC-pik3r1KO mice receiving TPN or enteral nutrition were treated with EGF or GLP-2 alone or in combination with reciprocal receptor inhibitors, GLP-2(3-33) or gefitinib. Jejunum was collected and mucosal atrophy and IEC responses were assessed by histologic, gene, and protein expression analyses. In patients undergoing planned looped ileostomies, fed and unfed ileum was analyzed. RESULTS: Enteral nutrient deprivation reduced endogenous EGF and GLP-2 signaling in mice and human beings. In the mouse TPN model, exogenous EGF or GLP-2 attenuated mucosal atrophy and restored IEC proliferation. The beneficial effects of EGF and GLP-2 were decreased upon Gefitinib treatment and in TPN-treated IEC-EgfrKO mice, showing epidermal growth factor-receptor dependency for these IEC responses. By contrast, in TPN-treated IEC-pi3kr1KO mice, the beneficial actions of EGF were lost, although GLP-2 still attenuated mucosal atrophy. CONCLUSIONS: Upon enteral nutrient deprivation, exogenous GLP-2 and EGF show strong interdependency for improving IEC responses. Understanding the differential requirements for phosphatidylinositol 3-kinase/phosphoAKT (Ser473) signaling may help improve future therapies to prevent mucosal atrophy.

5.
Am J Physiol Gastrointest Liver Physiol ; 311(4): G734-G743, 2016 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-27586649

RESUMEN

Total parenteral nutrition (TPN) leads to a shift in small intestinal microbiota with a characteristic dominance of Proteobacteria This study examined how metabolomic changes within the small bowel support an altered microbial community in enterally deprived mice. C57BL/6 mice were given TPN or enteral chow. Metabolomic analysis of jejunal contents was performed by liquid chromatography/mass spectrometry (LC/MS). In some experiments, leucine in TPN was partly substituted with [13C]leucine. Additionally, jejunal contents from TPN-dependent and enterally fed mice were gavaged into germ-free mice to reveal whether the TPN phenotype was transferrable. Small bowel contents of TPN mice maintained an amino acid composition similar to that of the TPN solution. Mass spectrometry analysis of small bowel contents of TPN-dependent mice showed increased concentration of 13C compared with fed mice receiving saline enriched with [13C]leucine. [13C]leucine added to the serosal side of Ussing chambers showed rapid permeation across TPN-dependent jejunum, suggesting increased transmucosal passage. Single-cell analysis by fluorescence in situ hybridization (FISH)-NanoSIMS demonstrated uptake of [13C]leucine by TPN-associated bacteria, with preferential uptake by Enterobacteriaceae Gavage of small bowel effluent from TPN mice into germ-free, fed mice resulted in a trend toward the proinflammatory TPN phenotype with loss of epithelial barrier function. TPN dependence leads to increased permeation of TPN-derived nutrients into the small intestinal lumen, where they are predominately utilized by Enterobacteriaceae The altered metabolomic composition of the intestinal lumen during TPN promotes dysbiosis.


Asunto(s)
Microbioma Gastrointestinal/fisiología , Mucosa Intestinal/metabolismo , Yeyuno/metabolismo , Nutrición Parenteral Total , Sepsis/metabolismo , Animales , Modelos Animales de Enfermedad , Mucosa Intestinal/microbiología , Yeyuno/microbiología , Masculino , Metaboloma , Ratones , Ratones Endogámicos C57BL , Sepsis/microbiología
6.
Sci Rep ; 6: 27634, 2016 06 15.
Artículo en Inglés | MEDLINE | ID: mdl-27302484

RESUMEN

Intestinal resident macrophages (Mϕs) regulate gastrointestinal homeostasis via production of an anti-inflammatory cytokine interleukin (IL)-10. Although a constant replenishment by circulating monocytes is required to maintain the pool of resident Mϕs in the colonic mucosa, the homeostatic regulation of Mϕ in the small intestine (SI) remains unclear. Here, we demonstrate that direct stimulation by dietary amino acids regulates the homeostasis of intestinal Mϕs in the SI. Mice that received total parenteral nutrition (TPN), which deprives the animals of enteral nutrients, displayed a significant decrease of IL-10-producing Mϕs in the SI, whereas the IL-10-producing CD4(+) T cells remained intact. Likewise, enteral nutrient deprivation selectively affected the monocyte-derived F4/80(+) Mϕ population, but not non-monocytic precursor-derived CD103(+) dendritic cells. Notably, in contrast to colonic Mϕs, the replenishment of SI Mϕs and their IL-10 production were not regulated by the gut microbiota. Rather, SI Mϕs were directly regulated by dietary amino acids. Collectively, our study highlights the diet-dependent, microbiota-independent regulation of IL-10-producing resident Mϕs in the SI.


Asunto(s)
Interleucina-10/metabolismo , Intestino Delgado/metabolismo , Macrófagos/metabolismo , Membrana Mucosa/metabolismo , Aminoácidos/metabolismo , Alimentación Animal , Animales , Antígenos CD/metabolismo , Antígeno CD11b/metabolismo , Linfocitos T CD4-Positivos/citología , Quimiocina CCL2/metabolismo , Citocinas/metabolismo , Microbioma Gastrointestinal , Homeostasis , Cadenas alfa de Integrinas/metabolismo , Mucosa Intestinal/metabolismo , Intestino Delgado/microbiología , Macrófagos/microbiología , Masculino , Ratones , Ratones Endogámicos C57BL , Monocitos/metabolismo , Membrana Mucosa/microbiología , Receptores CCR2/metabolismo
7.
Am J Physiol Gastrointest Liver Physiol ; 310(4): G273-84, 2016 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-26635320

RESUMEN

Feeding strategies to care for patients who transition from enteral nutrient deprivation while on total parenteral nutrition (TPN) to enteral feedings generally proceed to full enteral nutrition once the gastrointestinal tract recovers; however, an increasing body of literature suggests that a subgroup of patients may actually develop an increased incidence of adverse events, including death. To examine this further, we studied the effects of acute refeeding in a mouse model of TPN. Interestingly, refeeding led to some beneficial effects, including prevention in the decline in intestinal epithelial cell (IEC) proliferation. However, refeeding led to a significant increase in mucosal expression of proinflammatory cytokines, including tumor necrosis factor-α (TNF-α), as well as an upregulation in Toll-like receptor 4 (TLR-4). Refeeding also failed to prevent TPN-associated increases in IEC apoptosis, loss of epithelial barrier function, and failure of the leucine-rich repeat-containing G protein-coupled receptor 5-positive stem cell expression. Transitioning from TPN to enteral feedings led to a partial restoration of the small bowel microbial population. In conclusion, while acute refeeding led to some restoration of normal gastrointestinal physiology, enteral refeeding led to a significant increase in mucosal inflammatory markers and may suggest alternative strategies to enteral refeeding should be considered.


Asunto(s)
Nutrición Enteral/efectos adversos , Homeostasis , Mucosa Intestinal/efectos de los fármacos , Intestino Delgado/efectos de los fármacos , Nutrición Parenteral Total/efectos adversos , Animales , Apoptosis , Proliferación Celular , Citocinas/biosíntesis , Citocinas/metabolismo , Microbioma Gastrointestinal , Mucosa Intestinal/metabolismo , Intestino Delgado/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Receptores Acoplados a Proteínas G/biosíntesis , Receptores Acoplados a Proteínas G/genética , Células Madre , Proteínas de Uniones Estrechas/biosíntesis , Proteínas de Uniones Estrechas/genética , Receptor Toll-Like 4/biosíntesis , Receptor Toll-Like 4/genética , Factor de Necrosis Tumoral alfa/biosíntesis
8.
Dig Dis Sci ; 61(6): 1524-33, 2016 06.
Artículo en Inglés | MEDLINE | ID: mdl-26685910

RESUMEN

BACKGROUND: Total parenteral nutrition (TPN), a necessary treatment for patients who cannot receive enteral nutrition, is associated with infectious complications due in part to a loss of intestinal epithelial barrier function (EBF). Using a mouse model of TPN, with enteral nutrient deprivation, we previously demonstrated an increase in mucosal interferon-γ and tumor necrosis factor-α; these cytokine changes are a major mediator driving a reduction in epithelial tight junction (TJ) protein expression. However, the exact ultrastructural changes to the intestinal epithelial barrier have not been previously described. AIM: We hypothesized that TPN dependence results in ultrastructural changes in the intestinal epithelial TJ meshwork. METHODS: C57BL/6 mice underwent internal jugular venous cannulation and were given enteral nutrition or TPN with enteral nutrient deprivation for 7 days. Freeze-fracture electron microscopy was performed on ileal tissue to characterize changes in TJ ultrastructure. EBF was measured using transepithelial resistance and tracer permeability, while TJ expression was measured via Western immunoblotting and immunofluorescence staining. RESULTS: While strand density, linearity, and appearance were unchanged, TPN dependence led to a mean reduction in one horizontal strand out of the TJ compact meshwork to a more basal region, resulting in a reduction in meshwork depth. These findings were correlated with the loss of TJ localization of claudin-4 and tricellulin, reduced expression of claudin-5 and claudin-8, and reduced ex vivo EBF. CONCLUSION: Tight junction ultrastructural changes may contribute to reduced EBF in the setting of TPN dependence.


Asunto(s)
Mucosa Intestinal/citología , Nutrición Parenteral Total/efectos adversos , Uniones Estrechas/ultraestructura , Animales , Técnica de Fractura por Congelación , Mucosa Intestinal/efectos de los fármacos , Masculino , Ratones , Ratones Endogámicos C57BL , Microscopía Electrónica/métodos , Uniones Estrechas/efectos de los fármacos
9.
Mol Cell Biol ; 35(21): 3604-21, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-26283731

RESUMEN

Total parenteral nutrition (TPN) is commonly used clinically to sustain patients; however, TPN is associated with profound mucosal atrophy, which may adversely affect clinical outcomes. Using a mouse TPN model, removing enteral nutrition leads to decreased crypt proliferation, increased intestinal epithelial cell (IEC) apoptosis and increased mucosal tumor necrosis factor alpha (TNF-α) expression that ultimately produces mucosal atrophy. Upregulation of TNF-α signaling plays a central role in mediating TPN-induced mucosal atrophy without intact epidermal growth factor receptor (EGFR) signaling. Currently, the mechanism and the tissue-specific contributions of TNF-α signaling to TPN-induced mucosal atrophy remain unclear. ADAM17 is an ectodomain sheddase that can modulate the signaling activity of several cytokine/growth factor receptor families, including the TNF-α/TNF receptor and ErbB ligand/EGFR pathways. Using TPN-treated IEC-specific ADAM17-deficient mice, the present study demonstrates that a loss of soluble TNF-α signaling from IECs attenuates TPN-induced mucosal atrophy. Importantly, this response remains dependent on the maintenance of functional EGFR signaling in IECs. TNF-α blockade in wild-type mice receiving TPN confirmed that soluble TNF-α signaling is responsible for downregulation of EGFR signaling in IECs. These results demonstrate that ADAM17-mediated TNF-α signaling from IECs has a significant role in the development of the proinflammatory state and mucosal atrophy observed in TPN-treated mice.


Asunto(s)
Proteínas ADAM/genética , Mucosa Intestinal/patología , Nutrición Parenteral Total/efectos adversos , Transducción de Señal , Factor de Necrosis Tumoral alfa/inmunología , Proteínas ADAM/inmunología , Proteína ADAM17 , Animales , Apoptosis , Atrofia/inmunología , Atrofia/patología , Proliferación Celular , Citocinas/inmunología , Receptores ErbB/inmunología , Femenino , Técnicas de Inactivación de Genes , Humanos , Mucosa Intestinal/citología , Mucosa Intestinal/inmunología , Mucosa Intestinal/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Factor de Transcripción STAT3/inmunología
10.
FASEB J ; 29(7): 2943-58, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-25782989

RESUMEN

Recent studies suggest a close interaction between epidermal growth factor (EGF) and TLR signaling in the modulation of intestinal epithelial cell (IEC) proliferation; however, how these signaling pathways adjust IEC proliferation is poorly understood. We utilized a model of total parenteral nutrition (TPN), or enteral nutrient deprivation, to study this interaction as TPN results in mucosal atrophy due to decreased IEC proliferation and increased apoptosis. We identified the novel finding of decreased mucosal atrophy in TLR4 knockout (TLR4KO) mice receiving TPN. We hypothesized that EGF signaling is preserved in TLR4KO-TPN mice and prevents mucosal atrophy. C57Bl/6 and strain-matched TLR4KO mice were provided either enteral feeding or TPN. IEC proliferation and apoptosis were measured. Cytokine and growth factor abundances were detected in both groups. To examine interdependence of these pathways, ErbB1 pharmacologic blockade was used. The marked decline in IEC proliferation with TPN was nearly prevented in TLR4KO mice, and intestinal length was partially preserved. EGF was significantly increased, and TNF-α decreased in TLR4KO-TPN versus wild-type (WT)-TPN mice. Apoptotic positive crypt cells were 15-fold higher in WT-TPN versus TLR4KO-TPN mice. Bcl-2 was significantly increased in TLR4KO-TPN mice, while Bax decreased 10-fold. ErbB1 blockade prevented this otherwise protective effect in TLR4KO-sTPN mice. TLR4 blockade significantly prevented TPN-associated atrophy by preserving proliferation and preventing apoptosis. This is driven by a reduction in TNF-α abundance and increased EGF. Potential manipulation of this regulatory pathway may have significant clinical potential to prevent TPN-associated atrophy.


Asunto(s)
Factor de Crecimiento Epidérmico/metabolismo , Mucosa Intestinal/metabolismo , Mucosa Intestinal/patología , Nutrición Parenteral Total/efectos adversos , Receptor Toll-Like 4/metabolismo , Animales , Apoptosis , Atrofia , Proliferación Celular , Factor de Crecimiento Epidérmico/antagonistas & inhibidores , Factor de Crecimiento Epidérmico/genética , Receptores ErbB/antagonistas & inhibidores , Gefitinib , Interferón gamma/metabolismo , Mucosa Intestinal/efectos de los fármacos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Quinazolinas/farmacología , ARN Mensajero/genética , ARN Mensajero/metabolismo , Transducción de Señal , Receptor Toll-Like 4/deficiencia , Receptor Toll-Like 4/genética , Factor de Necrosis Tumoral alfa/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...