Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
Más filtros











Intervalo de año de publicación
1.
Cell Death Differ ; 23(8): 1331-46, 2016 08.
Artículo en Inglés | MEDLINE | ID: mdl-26891693

RESUMEN

Nod-like receptor, pyrin containing 3 (NLRP3) is characterized primarily as a canonical caspase-1 activating inflammasome in macrophages. NLRP3 is also expressed in the epithelium of the kidney and gut; however, its function remains largely undefined. Primary mouse tubular epithelial cells (TEC) lacking Nlrp3 displayed reduced apoptosis downstream of the tumor necrosis factor (TNF) receptor and CD95. TECs were identified as type II apoptotic cells that activated caspase-8, tBid and mitochondrial apoptosis via caspase-9, responses that were reduced in Nlrp3-/- cells. The activation of caspase-8 during extrinsic apoptosis induced by TNFα/cycloheximide (TNFα/CHX) was dependent on adaptor protein apoptosis-associated speck-like protein containing a CARD (ASC) and completely independent of caspase-1 or caspase-11. TECs and primary human proximal tubular epithelial cells (HPTC) did not activate a canonical inflammasome, caspase-1, or IL-1ß secretion in response to TNFα/CHX or NLRP3-dependent triggers, such as ATP or nigericin. In cell fractionation studies and by confocal microscopy, NLRP3 colocalized with ASC and caspase-8 in speck-like complexes at the mitochondria during apoptosis. The formation of NLRP3/ASC/caspase-8 specks in response to TNFα/CHX was downstream of TNFR signaling and dependent on potassium efflux. Epithelial ASC specks were present in enteroids undergoing apoptosis and in the injured tubules of wild-type but not Nlrp3-/- or ASC-/- mice following ureteric unilateral obstruction in vivo. These data show that NLRP3 and ASC form a conserved non-canonical platform for caspase-8 activation, independent of the inflammasome that regulates apoptosis within epithelial cells.


Asunto(s)
Apoptosis , Caspasa 8/metabolismo , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Adenosina Trifosfato/farmacología , Animales , Apoptosis/efectos de los fármacos , Caspasa 1/genética , Caspasa 1/metabolismo , Caspasas/genética , Caspasas/metabolismo , Células Cultivadas , Cicloheximida/toxicidad , Células Epiteliales/citología , Células Epiteliales/efectos de los fármacos , Células Epiteliales/metabolismo , Humanos , Inflamasomas/metabolismo , Interleucina-1beta/análisis , Interleucina-1beta/metabolismo , Túbulos Renales Proximales/citología , Macrófagos/citología , Macrófagos/efectos de los fármacos , Macrófagos/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteína con Dominio Pirina 3 de la Familia NLR/deficiencia , Proteína con Dominio Pirina 3 de la Familia NLR/genética , Nigericina/farmacología , Factor de Necrosis Tumoral alfa/toxicidad
2.
Am J Physiol Gastrointest Liver Physiol ; 308(5): G389-402, 2015 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-25501546

RESUMEN

We previously showed that vasoactive intestinal peptide (VIP) protects against bacterial pathogen-induced epithelial barrier disruption and colitis, although the mechanisms remain poorly defined. The aim of the current study was to identify cellular pathways of VIP-mediated protection with use of pharmacological inhibitors during enteropathogenic Escherichia coli (EPEC) infection of Caco-2 cell monolayers and during Citrobacter rodentium-induced colitis. EPEC-induced epithelial barrier disruption involved the PKC pathway but was independent of functional cAMP, Rho, and NF-κB pathways. VIP mediated its protective effects by inhibiting EPEC-induced PKC activity and increasing expression of the junctional protein claudin-4. Short-term treatment with TPA, which is known to activate PKC, was inhibited by VIP pretreatment, while PKC degradation via long-term treatment with TPA mimicked the protective actions of VIP. Immunostaining for specific PKC isotypes showed upregulated expression of PKCθ and PKCε during EPEC infection. Treatment with specific inhibitors revealed a critical role for PKCε in EPEC-induced barrier disruption. Furthermore, activation of PKCε and loss of barrier integrity correlated with claudin-4 degradation. In contrast, inhibition of PKCε by VIP pretreatment or the PKCε inhibitor maintained membrane-bound claudin-4 levels, along with barrier function. Finally, in vivo treatment with the PKCε inhibitor protected mice from C. rodentium-induced colitis. In conclusion, EPEC infection increases intracellular PKCε levels, leading to decreased claudin-4 levels and compromising epithelial barrier integrity. VIP inhibits PKCε activation, thereby attenuating EPEC-induced barrier disruption.


Asunto(s)
Escherichia coli Enteropatógena/patogenicidad , Infecciones por Escherichia coli/metabolismo , Proteína Quinasa C-epsilon/metabolismo , Péptido Intestinal Vasoactivo/farmacología , Adulto , Anciano , Animales , Células CACO-2 , Células Cultivadas , Citrobacter rodentium/patogenicidad , Claudina-4/genética , Claudina-4/metabolismo , Colitis/tratamiento farmacológico , Colitis/metabolismo , AMP Cíclico/metabolismo , Infecciones por Escherichia coli/tratamiento farmacológico , Femenino , Células HT29 , Humanos , Mucosa Intestinal/efectos de los fármacos , Mucosa Intestinal/metabolismo , Mucosa Intestinal/microbiología , Masculino , Ratones , Ratones Endogámicos C57BL , FN-kappa B/metabolismo , Proteína Quinasa C-epsilon/antagonistas & inhibidores , Péptido Intestinal Vasoactivo/uso terapéutico , Quinasas Asociadas a rho/metabolismo
3.
Am J Physiol Gastrointest Liver Physiol ; 297(4): G735-50, 2009 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-19661153

RESUMEN

Attaching and effacing bacterial pathogens attach to the apical surface of epithelial cells and disrupt epithelial barrier function, increasing permeability and allowing luminal contents access to the underlying milieu. Previous in vitro studies demonstrated that the neuropeptide vasoactive intestinal peptide (VIP) regulates epithelial paracellular permeability, and the high concentrations and close proximity of VIP-containing nerve fibers to intestinal epithelial cells would support such a function in vivo. The aim of this study was to examine whether VIP treatment modulated Citrobacter rodentium-induced disruption of intestinal barrier integrity and to identify potential mechanisms of action. Administration of VIP had no effect on bacterial attachment although histopathological scoring demonstrated a VIP-induced amelioration of colitis-induced epithelial damage compared with controls. VIP treatment prevented the infection-induced increase in mannitol flux a measure of paracellular permeability, resulting in levels similar to control mice, and immunohistochemical studies demonstrated that VIP prevented the translocation of tight junction proteins: zonula occludens-1, occludin, and claudin-3. Enteropathogenic Escherichia coli (EPEC) infection of Caco-2 monolayers confirmed a protective role for VIP on epithelial barrier function. VIP prevented EPEC-induced increase in long myosin light chain kinase (MLCK) expression and myosin light chain phosphorylation (p-MLC). Furthermore, MLCK inhibition significantly attenuated bacterial-induced epithelial damage both in vivo and in vitro. In conclusion, our results indicate that VIP protects the colonic epithelial barrier by minimizing bacterial-induced redistribution of tight junction proteins in part through actions on MLCK and MLC phosphorylation.


Asunto(s)
Antiinflamatorios/administración & dosificación , Traslocación Bacteriana/efectos de los fármacos , Citrobacter rodentium/patogenicidad , Colitis/prevención & control , Colon/efectos de los fármacos , Infecciones por Enterobacteriaceae/tratamiento farmacológico , Mucosa Intestinal/efectos de los fármacos , Péptido Intestinal Vasoactivo/administración & dosificación , Animales , Azepinas/farmacología , Adhesión Bacteriana , Células CACO-2 , Claudina-3 , Colitis/metabolismo , Colitis/microbiología , Colitis/patología , Colon/metabolismo , Colon/microbiología , Colon/patología , Modelos Animales de Enfermedad , Infecciones por Enterobacteriaceae/complicaciones , Infecciones por Enterobacteriaceae/metabolismo , Infecciones por Enterobacteriaceae/microbiología , Humanos , Inyecciones Intraperitoneales , Mucosa Intestinal/metabolismo , Mucosa Intestinal/microbiología , Manitol/metabolismo , Proteínas de la Membrana/metabolismo , Ratones , Ratones Endogámicos C57BL , Cadenas Ligeras de Miosina/metabolismo , Quinasa de Cadena Ligera de Miosina/antagonistas & inhibidores , Quinasa de Cadena Ligera de Miosina/metabolismo , Naftalenos/farmacología , Ocludina , Permeabilidad , Fosfoproteínas/metabolismo , Fosforilación , Inhibidores de Proteínas Quinasas/farmacología , Uniones Estrechas/efectos de los fármacos , Uniones Estrechas/metabolismo , Uniones Estrechas/microbiología , Factores de Tiempo , Proteína de la Zonula Occludens-1
4.
Cell Microbiol ; 10(3): 618-31, 2008 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-17979981

RESUMEN

Myeloid differentiation factor (MyD)88, an adaptor protein shared by the Toll-interleukin 1 receptor superfamily, plays a critical role in host defence during many systemic bacterial infections by inducing protective inflammatory responses that limit bacterial growth. However, the role of innate responses during gastrointestinal (GI) infections is less clear, in part because the GI tract is tolerant to commensal antigens. The current study investigated the role of MyD88 following infection by the murine bacterial pathogen, Citrobacter rodentium. MyD88-deficient mice suffered a lethal colitis coincident with colonic mucosal ulcerations and bleeding. Their susceptibility was associated with an overwhelming bacterial burden and selectively impaired immune responses in colonic tissues, which included delayed inflammatory cell recruitment, reduced iNOS and abrogated production of TNF-alpha and IL-6 from MyD88-deficient macrophages and colons cultured ex vivo. Immunostaining for Ki67 and BrDU revealed that MyD88 signalling mediated epithelial hyper-proliferation in response to C. rodentium infection. Thus, MyD88-deficient mice could not promote epithelial cell turnover and repair, leading to deep bacterial invasion of colonic crypts, intestinal barrier dysfunction and, ultimately, widespread mucosal ulcerations. In conclusion, MyD88 signalling within the GI tract plays a critical role in mediating host defence against an enteric bacterial pathogen, by controlling bacterial numbers and promoting intestinal epithelial homeostasis.


Asunto(s)
Citrobacter rodentium/inmunología , Colitis/inmunología , Células Epiteliales/microbiología , Factor 88 de Diferenciación Mieloide/deficiencia , Factor 88 de Diferenciación Mieloide/fisiología , Animales , Médula Ósea/microbiología , Colon/química , Colon/microbiología , Colon/patología , Recuento de Colonia Microbiana , Ensayo de Inmunoadsorción Enzimática , Interleucina-6/análisis , Antígeno Ki-67/análisis , Macrófagos/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Óxido Nítrico Sintasa de Tipo II/análisis , Técnicas de Cultivo de Órganos , Análisis de Supervivencia , Factor de Necrosis Tumoral alfa/análisis
5.
Infect Immun ; 69(10): 6217-24, 2001 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-11553563

RESUMEN

Enteropathogenic Escherichia coli (EPEC) is an extracellular bacterial pathogen that infects the human intestinal epithelium and is a major cause of infantile diarrhea in developing countries. EPEC belongs to the group of attaching and effacing (A/E) pathogens. It uses a type III secretion system to deliver proteins into the host cell that mediate signal transduction events in host cells. We used gene array technology to study epithelial cell responses to EPEC infection at the level of gene expression. We found that EPEC induces the expression of several genes in infected HeLa cells by a lipopolysaccharide (LPS)-independent mechanism, including cytokines and early growth response factor 1 (Egr-1). The transcription factor Egr-1 is an immediate-early-induced gene that is activated in most cell types in response to stress. EPEC-induced upregulation of egr-1 is mediated by the activation of the MEK/extracellular signal-regulated kinase signal transduction pathway and is dependent on the type III secretion system. egr-1 is also induced during infection of mice by the A/E pathogen Citrobacter rodentium, suggesting that both Egr-1 and the activation of this mitogen-activated protein kinase signal transduction pathway may play a role in disease.


Asunto(s)
Proteínas de Unión al ADN/genética , Infecciones por Escherichia coli/metabolismo , Proteínas Inmediatas-Precoces/genética , Sistema de Señalización de MAP Quinasas , Factores de Transcripción/genética , Animales , Northern Blotting/métodos , Citrobacter freundii , Proteína 1 de la Respuesta de Crecimiento Precoz , Infecciones por Enterobacteriaceae/metabolismo , Activación Enzimática , Células Epiteliales , Expresión Génica , Células HeLa , Humanos , Interleucina-8/genética , Lipopolisacáridos/farmacología , Ratones , Ratones Endogámicos C57BL , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Fosforilación , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
6.
Am J Physiol Gastrointest Liver Physiol ; 281(1): G102-10, 2001 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-11408260

RESUMEN

Immune responses elicited by nematode parasite infections are characterized by T helper 2 (Th2) cell induction. The immunologic basis for changes in intestinal physiology accompanying nematode infection is poorly understood. This study examined whether worm expulsion and associated goblet cell hyperplasia and muscle contractility share a similar immune basis by shifting the response from Th2 to Th1 using interleukin-12 (IL-12) overexpression. We used a single administration of recombinant adenovirus vector expressing IL-12 (Ad5IL-12) in Trichinella spiralis-infected mice. Ad5IL-12 administered 1 day after infection prolonged worm survival and inhibited infection-induced muscle hypercontractility and goblet cell hyperplasia. This was correlated with upregulated interferon-gamma (IFN-gamma) expression and downregulated IL-13 expression in the muscularis externa layer. We also observed increased IFN-gamma production and decreased IL-4 and IL-13 production from in vitro stimulated spleen and mesenteric lymph node cells of infected Ad5IL-12-treated mice. These results indicate that transfer and overexpression of the IL-12 gene during Th2-based nematode infection shifts the immune response toward Th1 and delays worm expulsion. Moreover, the immune response shift abrogated the physiological responses to infection, attenuating both muscle hypercontractility and goblet cell hyperplasia. These findings strongly indicate that worm expulsion, muscle hypercontractility, and goblet cell hyperplasia share a common immunologic basis and may be causally linked.


Asunto(s)
Células Caliciformes/inmunología , Células Caliciformes/parasitología , Interleucina-12/genética , Trichinella spiralis , Triquinelosis/inmunología , Adenoviridae/genética , Animales , Expresión Génica/inmunología , Técnicas de Transferencia de Gen , Células Caliciformes/patología , Interacciones Huésped-Parásitos/inmunología , Hiperplasia , Técnicas In Vitro , Interferón gamma/genética , Interferón gamma/inmunología , Interleucina-12/sangre , Interleucina-12/inmunología , Interleucina-13/genética , Interleucina-13/inmunología , Interleucina-4/genética , Interleucina-4/inmunología , Ganglios Linfáticos/citología , Masculino , Ratones , Músculo Liso/inmunología , Músculo Liso/parasitología , Peroxidasa/metabolismo , Bazo/citología
7.
Am J Physiol Gastrointest Liver Physiol ; 281(1): G151-8, 2001 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-11408267

RESUMEN

Intestinal inflammation due to nematode infection impairs enteric cholinergic nerve function and induces hypercontractility of intestinal muscle. Macrophages have been implicated in the neural changes, but the subpopulation and mechanism involved are unknown. We examined whether macrophages alter nerves by virtue of their ability to activate lymphocytes via major histocompatibility complex (MHC) II-restricted antigen presentation. We also attempted to evaluate the role of macrophage subsets using op/op mice deficient in macrophage colony-stimulating factor (M-CSF). ACh release from the myenteric plexus was measured in MHC II- and M-CSF-deficient (op/op) mice infected with Trichinella spiralis. F4/80-positive macrophages and interleukin-1 beta were constitutively present in op/op and op/? mice but increased only in op/? mice postinfection. After infection, a marked suppression of ACh release occurred only in infected MHC II-deficient and op/? mice. Muscle hypercontractility remained evident in infected op/? mice. Treatment with M-CSF restored macrophage number, and this was accompanied by suppression of cholinergic nerve function during infection. Thus M-CSF plays a critical role in this model by recruiting a subset of macrophages that selectively suppresses enteric neural function.


Asunto(s)
Fibras Colinérgicas/inmunología , Fibras Colinérgicas/parasitología , Factor Estimulante de Colonias de Macrófagos/inmunología , Macrófagos/parasitología , Trichinella , Triquinelosis/inmunología , Acetilcolina/metabolismo , Animales , Fibras Colinérgicas/metabolismo , Expresión Génica/inmunología , Antígenos de Histocompatibilidad Clase II/genética , Antígenos de Histocompatibilidad Clase II/inmunología , Enfermedades Inflamatorias del Intestino/inmunología , Enfermedades Inflamatorias del Intestino/parasitología , Interleucina-1/genética , Interleucina-1/inmunología , Interleucina-4/genética , Interleucina-4/inmunología , Yeyuno/inmunología , Yeyuno/inervación , Yeyuno/parasitología , Factor Estimulante de Colonias de Macrófagos/genética , Macrófagos/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Mutantes , Contracción Muscular/fisiología , Músculo Liso/fisiología , Mutación/inmunología , Plexo Mientérico/citología , Plexo Mientérico/inmunología , Plexo Mientérico/parasitología , ARN Mensajero , Organismos Libres de Patógenos Específicos , Tritio
8.
Am J Physiol Gastrointest Liver Physiol ; 280(4): G640-8, 2001 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-11254490

RESUMEN

In response to nematode infection, the host presumably attempts to create an unfavorable environment to prevent larval penetration of the host and to expedite parasite expulsion from the gut. In this study, we have used W/W(V) mice with or without mast cells after bone marrow reconstitution (BMR-W/W(V)) to examine the role of mast cells in the host response. W/W(V), BMR-W/W(V), and wild-type (+/+) mice were infected with Trichinella spiralis. Infected W/W(V) mice exhibited less tissue damage and experienced a delay in worm expulsion and a greater degree of larval penetration of the gut leading to encystment in skeletal muscle. Tissue injury was greater and worm expulsion was normalized in BMR-W/W(V) mice, but larval penetration remained unchanged. Spontaneous contractile activity of jejunal muscle was disrupted in W/W(V) mice, as was the contractile response to carbachol. These abnormalities were also present in BMR-W/W(V) mice. These results indicate that mast cells mediate tissue damage and contribute to the timely expulsion of nematodes from the gut during primary infection.


Asunto(s)
Inmunidad Celular/fisiología , Mastocitos/fisiología , Músculo Esquelético/fisiopatología , Trichinella spiralis , Triquinelosis/inmunología , Triquinelosis/fisiopatología , Animales , Antígenos Helmínticos/inmunología , Peso Corporal/fisiología , Trasplante de Médula Ósea/fisiología , Recuento de Células , Células Madre Hematopoyéticas/fisiología , Mucosa Intestinal/parasitología , Mucosa Intestinal/patología , Ratones , Ratones Congénicos , Ratones Endogámicos , Contracción Muscular/fisiología , Músculo Esquelético/parasitología , Proteínas Proto-Oncogénicas c-kit/metabolismo , Triquinelosis/parasitología
9.
Infect Immun ; 69(2): 838-44, 2001 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-11159976

RESUMEN

Intestinal nematode infections in rats or mice are accompanied by intestinal muscle hyper contractility that may contribute to parasite expulsion from the gut. Previous studies demonstrated that both the expulsion of nematode parasites and the associated muscle hyper contractility are dependent on CD4(+) T helper cells. Nevertheless, the precise immunological mechanism underlying changes in intestinal muscle function remains to be determined. In this study, we investigated the role of interleukin 4 (IL-4) and signal transducer and activator of transcription factor 6 (STAT6) in the development of intestinal muscle hypercontractility and worm expulsion by infecting IL-4 and STAT6-deficient mice with Trichinella spiralis. Worm expulsion was almost normal in IL-4-deficient mice but substantially delayed in STAT6-deficient mice. Consistent with delayed worm expulsion, we also observed a marked attenuation of carbachol-induced muscle contraction in STAT6-deficient mice but only a moderate decrease in muscle hypercontractility in IL-4-deficient mice. In addition, we also observed severe impairment of T helper type 2 cytokine responses and intestinal mucosal mastocytosis in STAT6-deficient mice, although some degree of intestinal tissue eosinophilia was evident in these animals. These results are consistent with the hypothesis that STAT6-dependent changes in intestinal muscle function contribute to host protection in nematode infection.


Asunto(s)
Intestinos/fisiopatología , Contracción Muscular , Transactivadores/fisiología , Trichinella spiralis , Triquinelosis/inmunología , Animales , Eosinofilia/etiología , Interleucina-13/fisiología , Interleucina-4/fisiología , Ganglios Linfáticos/inmunología , Ratones , Ratones Endogámicos C57BL , Factor de Transcripción STAT6 , Bazo/inmunología , Triquinelosis/fisiopatología
10.
Proc Natl Acad Sci U S A ; 97(16): 8799-806, 2000 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-10922038

RESUMEN

Microbial pathogens have evolved many ingenious ways to infect their hosts and cause disease, including the subversion and exploitation of target host cells. One such subversive microbe is enteropathogenic Escherichia coli (EPEC). A major cause of infantile diarrhea in developing countries, EPEC poses a significant health threat to children worldwide. Central to EPEC-mediated disease is its colonization of the intestinal epithelium. After initial adherence, EPEC causes the localized effacement of microvilli and intimately attaches to the host cell surface, forming characteristic attaching and effacing (A/E) lesions. Considered the prototype for a family of A/E lesion-causing bacteria, recent in vitro studies of EPEC have revolutionized our understanding of how these pathogens infect their hosts and cause disease. Intimate attachment requires the type III-mediated secretion of bacterial proteins, several of which are translocated directly into the infected cell, including the bacteria's own receptor (Tir). Binding to this membrane-bound, pathogen-derived protein permits EPEC to intimately attach to mammalian cells. The translocated EPEC proteins also activate signaling pathways within the underlying cell, causing the reorganization of the host actin cytoskeleton and the formation of pedestal-like structures beneath the adherent bacteria. This review explores what is known about EPEC's subversion of mammalian cell functions and how this knowledge has provided novel insights into bacterial pathogenesis and microbe-host interactions. Future studies of A/E pathogens in animal models should provide further insights into how EPEC exploits not only epithelial cells but other host cells, including those of the immune system, to cause diarrheal disease.


Asunto(s)
Adhesión Bacteriana , Escherichia coli O157/patogenicidad , Proteínas de Escherichia coli , Animales , Proteínas Bacterianas/fisiología , Diarrea/microbiología , Modelos Animales de Enfermedad , Células Epiteliales/microbiología , Infecciones por Escherichia coli/microbiología , Escherichia coli O157/fisiología , Humanos , Receptores de Superficie Celular/fisiología , Transducción de Señal , Virulencia
11.
Am J Physiol Gastrointest Liver Physiol ; 278(2): G259-65, 2000 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-10666050

RESUMEN

Trichinella spiralis infection in rodents is associated with suppression of ACh release from myenteric plexus that can be mimicked by macrophage-derived cytokines. We verified the presence of a macrophage infiltrate in the intestine during T. spiralis infection and determined the extent to which this cell type is responsible for the neural changes. C57BL/6 mice were infected with 375 T. spiralis larvae by gavage, and the presence of macrophages (F4/80 positive) in the jejunum was determined immunohistochemically. In another experiment, infected mice were treated intravenously with liposomes containing dichloromethylene diphosphonate (clodronate, Cl(2)MDP), which causes apoptosis of macrophages, and killed at postinfection day 6, and jejunal tissues were evaluated for the presence of F4/80-positive cells and for [(3)H]ACh release from the myenteric plexus. Infection caused an infiltration of F4/80-positive cells into the intestinal mucosa, muscle layers, and myenteric plexus region and a significant suppression of ACh release (50%). Depletion of F4/80-positive macrophages using Cl(2)MDP-containing liposomes prevented the suppression in [(3)H]ACh release, identifying macrophages as the cell type involved in the functional impairment of enteric cholinergic nerves.


Asunto(s)
Acetilcolina/metabolismo , Citocinas/fisiología , Parasitosis Intestinales/fisiopatología , Macrófagos/fisiología , Plexo Mientérico/fisiopatología , Trichinella spiralis , Triquinelosis/fisiopatología , Animales , Parasitosis Intestinales/patología , Mucosa Intestinal/patología , Yeyuno/patología , Cinética , Liposomas/administración & dosificación , Masculino , Ratones , Ratones Endogámicos C57BL , Peroxidasa/metabolismo , Triquinelosis/patología
12.
Infect Immun ; 67(11): 6090-7, 1999 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-10531271

RESUMEN

Expulsion of intestinal nematode parasites and the associated increased contraction by intestinal muscle are T cell dependent, since both are attenuated in athymic rodents. The CD4 T-cell subset has been strongly associated with worm expulsion; however, the relationship between these cells, antigen presentation, and worm expulsion is not definitive and the role of these factors in intestinal muscle hypercontractility has not been defined. We infected C57BL/6, athymic, CD4-deficient, CD8alpha-deficient, and major histocompatibility complex class II (MHC II)-deficient (C2d) mice with Trichinella spiralis larvae. We examined intestinal worm numbers, longitudinal muscle contraction, and MHC II expression. Numerous MHC II-positive cells were identified within the muscularis externa of infected but not uninfected C57BL/6 mice. C57BL/6 and CD8alpha-deficient mice developed large increases in muscle contraction, expelling the parasite by day 21. Athymic and C2d mice exhibited much smaller increases in muscle contraction and delayed parasite expulsion. CD4-deficient mice exhibited intermediate levels of muscle contraction and delayed parasite expulsion. To further examine the role of MHC II and CD4 T cells, we irradiated C2d mice and reconstituted them with C57BL/6 bone marrow alone or with C57BL/6 CD4 T cells. C57BL/6 bone marrow alone did not affect muscle function or worm expulsion in recipient C2d mice. Partial CD4 T-cell reconstitution was sufficient to restore increased muscle contraction but not worm expulsion. Thus, hematopoietic MHC II expression alone is insufficient for the development of muscle hypercontractility and worm expulsion, but the addition of even small numbers of CD4 T cells was sufficient to induce intestinal muscle pathophysiology.


Asunto(s)
Linfocitos T CD4-Positivos/fisiología , Antígenos de Histocompatibilidad Clase II/fisiología , Contracción Muscular , Trichinella spiralis/inmunología , Triquinelosis/inmunología , Animales , Antígenos de Histocompatibilidad Clase II/análisis , Inmunohistoquímica , Masculino , Ratones , Ratones Endogámicos C57BL , Triquinelosis/parasitología , Triquinelosis/fisiopatología
13.
Am J Physiol ; 276(3): R715-23, 1999 03.
Artículo en Inglés | MEDLINE | ID: mdl-10070131

RESUMEN

Specific in vivo T cell activation initiated by treatment with anti-CD3 antibodies leads to diarrhea and structural damage of the intestinal mucosa. In this study, the effect of T cell-induced mucosal damage on jejunal epithelial ion transport, muscle contractility, and neuronal ACh release was assessed in Ussing chambers, organ baths, and a specialized perfusion apparatus, respectively. Time-matched control mice received hamster serum containing irrelevant antibodies. Jejunal segments from anti-CD3-treated mice displayed a significantly elevated epithelial baseline short-circuit current (which indicates increased ion transport) and a concomitant reduction in responsiveness to prosecretory stimuli (nerve stimulation, carbachol, and forskolin). Longitudinal smooth muscle displayed altered spontaneous contractile activity, length-tension relationships, and carbachol-stimulated contraction in tissues excised from mice 20 and 40 h posttreatment. Anti-CD3 treatment did not affect stimulated ACh release from myenteric plexus neurons. We conclude that specific T cell activation via anti-CD3 antibody results in dramatic alterations in jejunal epithelial and smooth muscle function. Such T cell-induced changes in intestinal function may contribute to the symptomatology of T cell-mediated enteropathies, including graft-versus-host disease, celiac disease, and idiopathic inflammatory bowel disease.


Asunto(s)
Anticuerpos/farmacología , Complejo CD3/inmunología , Yeyuno/efectos de los fármacos , Yeyuno/fisiopatología , Activación de Linfocitos/fisiología , Linfocitos T/efectos de los fármacos , Linfocitos T/fisiología , Acetilcolina/metabolismo , Animales , Bromodesoxiuridina/metabolismo , Cricetinae , Femenino , Técnicas In Vitro , Mucosa Intestinal/efectos de los fármacos , Mucosa Intestinal/inervación , Mucosa Intestinal/patología , Mucosa Intestinal/fisiopatología , Yeyuno/inervación , Yeyuno/patología , Linfocitos/metabolismo , Ratones , Ratones Endogámicos BALB C , Músculo Liso/fisiopatología , Neuronas/metabolismo , Factor de Necrosis Tumoral alfa/biosíntesis
14.
J Clin Invest ; 100(11): 2766-76, 1997 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-9389741

RESUMEN

Inflammatory bowel disease (IBD) is characterized by altered immunoregulation and augmented intestinal synthesis of nitric oxide. The purpose of this study was to determine the effects of exogenous IL-4, introduced by a recombinant human type 5 adenovirus (Ad5) vector, on the tissue injury associated with an experimental model of colonic immune activation and inflammation. Colitis was induced in rats by the intrarectal administration of trinitrobenzene sulfonic acid (TNB) dissolved in 50% ethanol, and control rats received saline via the same route. 1 h later, all rats were randomized into two groups. The first group was injected intraperitoneally (ip) with 3.0 x 10(6) plaque forming units (PFUs) of Ad5 transfected with murine interleukin-4 (Ad5IL-4) and the second group was injected ip with the same amount of Ad5 expressing the Escherichia coli Lac Z gene (Ad5LacZ). One-half of the colitic and control rats were injected again with 3.0 x 10(6) PFUs of Ad5IL-4 or Ad5LacZ on day 3 of the 6-d study. When introduced once or twice via the peritoneal route into control rats, Ad5LacZ was localized to the serosal lining of the peritoneal cavity, the diaphragm and the liver on day 6. One or two injections of Ad5IL-4 into rats also produced measurable levels of circulating IL-4. TNB-colitis in both Ad5LacZ-treated groups was associated with pronounced elevations in serum IFN-gamma, and mucosal ulceration of the distal colon. Myeloperoxidase and inducible nitric oxide synthase II (NOS II) synthetic activity were also increased by 30- and fivefold, respectively, above control levels in the distal colon. However, two injections of Ad5IL-4 into colitic rats caused the overexpression of IL-4, and significantly inhibited tissue damage, serum and colon IFN-gamma levels and myeloperoxidase activity in the distal colon. In addition, NOS II gene expression and NOS II nitric oxide synthesis was significantly inhibited. No therapeutic effect was observed in rats injected once with Ad5IL-4. Thus, IL-4, introduced by Ad5, is therapeutic during acute inflammation in the rat colon. The therapeutic effect of IL-4 was associated with an inhibition of inducible nitric oxide expression and a reduction in nitric oxide synthesis.


Asunto(s)
Técnicas de Transferencia de Gen , Enfermedades Inflamatorias del Intestino/terapia , Interleucina-4/genética , Interleucina-4/uso terapéutico , Adenovirus Humanos/genética , Animales , Colon/metabolismo , Colon/patología , Modelos Animales de Enfermedad , Vectores Genéticos , Granulocitos/citología , Humanos , Enfermedades Inflamatorias del Intestino/inducido químicamente , Enfermedades Inflamatorias del Intestino/metabolismo , Inyecciones Intraperitoneales , Interferón gamma/metabolismo , Interleucina-4/metabolismo , Operón Lac , Masculino , Ratones , Óxido Nítrico Sintasa/biosíntesis , Óxido Nítrico Sintasa/genética , Peroxidasa/metabolismo , Ratas , Ratas Sprague-Dawley , beta-Galactosidasa/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA