Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
Add more filters










Publication year range
1.
J Clin Invest ; 126(2): 495-508, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26690704

ABSTRACT

The nature and role of the intestinal leukocytes in necrotizing enterocolitis (NEC), a severe disease affecting premature infants, remain unknown. We now show that the intestine in mouse and human NEC is rich in lymphocytes that are required for NEC development, as recombination activating gene 1­deficient (Rag1­/­) mice were protected from NEC and transfer of intestinal lymphocytes from NEC mice into naive mice induced intestinal inflammation. The intestinal expression of the lipopolysaccharide receptor TLR4, which is higher in the premature compared with full-term human and mouse intestine, is required for lymphocyte influx through TLR4-mediated upregulation of CCR9/CCL25 signaling. TLR4 also mediates a STAT3-dependent polarization toward increased proinflammatory CD3+CD4+IL-17+ and reduced tolerogenic Foxp3+ Treg lymphocytes (Tregs). Th17 lymphocytes were required for NEC development, as inhibition of STAT3 or IL-17 receptor signaling attenuated NEC in mice, while IL-17 release impaired enterocyte tight junctions, increased enterocyte apoptosis, and reduced enterocyte proliferation, leading to NEC. Importantly, TLR4-dependent Th17 polarization could be reversed by the enteral administration of retinoic acid, which induced Tregs and decreased NEC severity. These findings identify an important role for proinflammatory lymphocytes in NEC development via intestinal epithelial TLR4 that could be reversed through dietary modification.


Subject(s)
Enterocolitis, Necrotizing/immunology , Enterocytes/immunology , Infant, Newborn, Diseases/immunology , T-Lymphocytes, Regulatory/immunology , Th17 Cells/immunology , Toll-Like Receptor 4/immunology , Animals , Enterocolitis, Necrotizing/diet therapy , Enterocolitis, Necrotizing/genetics , Enterocolitis, Necrotizing/pathology , Enterocytes/pathology , Humans , Infant, Newborn , Infant, Newborn, Diseases/diet therapy , Infant, Newborn, Diseases/genetics , Infant, Newborn, Diseases/pathology , Mice , Mice, Knockout , T-Lymphocytes, Regulatory/pathology , Th17 Cells/pathology , Tight Junctions/genetics , Tight Junctions/immunology , Toll-Like Receptor 4/genetics
2.
Am J Physiol Gastrointest Liver Physiol ; 306(11): G917-28, 2014 Jun 01.
Article in English | MEDLINE | ID: mdl-24763555

ABSTRACT

Necrotizing enterocolitis is the leading cause of morbidity and mortality from gastrointestinal disease in premature infants and is characterized by initial feeding intolerance and abdominal distention followed by the rapid progression to coagulation necrosis of the intestine and death in many cases. Although the risk factors for NEC development remain well accepted, namely premature birth and formula feeding, the underlying mechanisms remain incompletely understood. Current thinking indicates that NEC develops in response to an abnormal interaction between the mucosal immune system of the premature host and an abnormal indigenous microflora, leading to an exaggerated mucosal inflammatory response and impaired mesenteric perfusion. In seeking to understand the molecular and cellular events leading to NEC, various animal models have been developed. However, the large number and variability between the available animal models and the unique characteristics of each has raised important questions regarding the validity of particular models for NEC research. In an attempt to provide some guidance to the growing community of NEC researchers, we now seek to review the key features of the major NEC models that have been developed in mammalian and nonmammalian species and to assess the advantages, disadvantage, challenges and major scientific discoveries yielded by each. A strategy for model validation is proposed, the principal models are compared, and future directions and challenges within the field of NEC research are explored.


Subject(s)
Enterocolitis, Necrotizing/physiopathology , Intestinal Mucosa/immunology , Animals , Disease Models, Animal , Humans , Immunity, Innate , Infant, Newborn , Infant, Premature , Reproducibility of Results , Research Design
3.
J Biol Chem ; 289(14): 9584-99, 2014 Apr 04.
Article in English | MEDLINE | ID: mdl-24519940

ABSTRACT

The cellular cues that regulate the apoptosis of intestinal stem cells (ISCs) remain incompletely understood, yet may play a role in diseases characterized by ISC loss including necrotizing enterocolitis (NEC). Toll-like receptor-4 (TLR4) was recently found to be expressed on ISCs, where its activation leads to ISC apoptosis through mechanisms that remain incompletely explained. We now hypothesize that TLR4 induces endoplasmic reticulum (ER) stress within ISCs, leading to their apoptosis in NEC pathogenesis, and that high ER stress within the premature intestine predisposes to NEC development. Using transgenic mice and cultured enteroids, we now demonstrate that TLR4 induces ER stress within Lgr5 (leucine-rich repeat-containing G-protein-coupled receptor 5)-positive ISCs, resulting in crypt apoptosis. TLR4 signaling within crypts was required, because crypt ER stress and apoptosis occurred in TLR4(ΔIEC-OVER) mice expressing TLR4 only within intestinal crypts and epithelium, but not TLR4(ΔIEC) mice lacking intestinal TLR4. TLR4-mediated ER stress and apoptosis of ISCs required PERK (protein kinase-related PKR-like ER kinase), CHOP (C/EBP homologous protein), and MyD88 (myeloid differentiation primary response gene 88), but not ATF6 (activating transcription factor 6) or XBP1 (X-box-binding protein 1). Human and mouse NEC showed high crypt ER stress and apoptosis, whereas genetic inhibition of PERK or CHOP attenuated ER stress, crypt apoptosis, and NEC severity. Strikingly, using intragastric delivery into fetal mouse intestine, prevention of ER stress reduced TLR4-mediated ISC apoptosis and mucosal disruption. These findings identify a novel link between TLR4-induced ER stress and ISC apoptosis in NEC pathogenesis and suggest that increased ER stress within the premature bowel predisposes to NEC development.


Subject(s)
Endoplasmic Reticulum Stress , Enterocolitis, Necrotizing/metabolism , Intestinal Mucosa/metabolism , Stem Cells/metabolism , Toll-Like Receptor 4/metabolism , Activating Transcription Factor 6/genetics , Activating Transcription Factor 6/metabolism , Animals , Apoptosis/genetics , Enterocolitis, Necrotizing/genetics , Enterocolitis, Necrotizing/pathology , HEK293 Cells , Humans , Intestinal Mucosa/pathology , Mice , Mice, Knockout , Stem Cells/pathology , Toll-Like Receptor 4/genetics , Transcription Factor CHOP/genetics , Transcription Factor CHOP/metabolism , eIF-2 Kinase/genetics , eIF-2 Kinase/metabolism
4.
Proc Natl Acad Sci U S A ; 110(23): 9451-6, 2013 Jun 04.
Article in English | MEDLINE | ID: mdl-23650378

ABSTRACT

Necrotizing enterocolitis (NEC) is a devastating disease of premature infants characterized by severe intestinal necrosis and for which breast milk represents the most effective protective strategy. Previous studies have revealed a critical role for the lipopolysaccharide receptor toll-like receptor 4 (TLR4) in NEC development through its induction of mucosal injury, yet the reasons for which intestinal ischemia in NEC occurs in the first place remain unknown. We hypothesize that TLR4 signaling within the endothelium plays an essential role in NEC development by regulating perfusion to the small intestine via the vasodilatory molecule endothelial nitric oxide synthase (eNOS). Using a unique mouse system in which we selectively deleted TLR4 from the endothelium, we now show that endothelial TLR4 activation is required for NEC development and that endothelial TLR4 activation impairs intestinal perfusion without effects on other organs and reduces eNOS expression via activation of myeloid differentiation primary response gene 88. NEC severity was significantly increased in eNOS(-/-) mice and decreased upon administration of the phosphodiesterase inhibitor sildenafil, which augments eNOS function. Strikingly, compared with formula, human and mouse breast milk were enriched in sodium nitrate--a precursor for enteral generation of nitrite and nitric oxide--and repletion of formula with sodium nitrate/nitrite restored intestinal perfusion, reversed the deleterious effects of endothelial TLR4 signaling, and reduced NEC severity. These data identify that endothelial TLR4 critically regulates intestinal perfusion leading to NEC and reveal that the protective properties of breast milk involve enhanced intestinal microcirculatory integrity via augmentation of nitrate-nitrite-NO signaling.


Subject(s)
Enterocolitis, Necrotizing/etiology , Intestinal Mucosa/blood supply , Microcirculation/physiology , Signal Transduction/physiology , Toll-Like Receptor 4/metabolism , Analysis of Variance , Animals , Animals, Newborn , Enterocolitis, Necrotizing/drug therapy , Enterocolitis, Necrotizing/metabolism , Infant Formula/chemistry , Infant Formula/pharmacology , Mice , Mice, Knockout , Microcirculation/drug effects , Microscopy, Confocal , Milk, Human/chemistry , Nitrates/analysis , Nitrates/metabolism , Nitric Oxide/metabolism , Nitric Oxide Synthase Type III/genetics , Nitric Oxide Synthase Type III/metabolism , Nitrites/metabolism , Piperazines/pharmacology , Piperazines/therapeutic use , Purines/pharmacology , Purines/therapeutic use , Signal Transduction/drug effects , Sildenafil Citrate , Sulfones/pharmacology , Sulfones/therapeutic use , Toll-Like Receptor 4/deficiency
5.
J Immunol ; 190(7): 3541-51, 2013 Apr 01.
Article in English | MEDLINE | ID: mdl-23455503

ABSTRACT

Necrotizing enterocolitis (NEC) develops in response to elevated TLR4 signaling in the newborn intestinal epithelium and is characterized by TLR4-mediated inhibition of enterocyte migration and reduced mucosal healing. The downstream processes by which TLR4 impairs mucosal healing remain incompletely understood. In other systems, TLR4 induces autophagy, an adaptive response to cellular stress. We now hypothesize that TLR4 induces autophagy in enterocytes and that TLR4-induced autophagy plays a critical role in NEC development. Using mice selectively lacking TLR4 in enterocytes (TLR4(ΔIEC)) and in TLR4-deficient cultured enterocytes, we now show that TLR4 activation induces autophagy in enterocytes. Immature mouse and human intestine showed increased expression of autophagy genes compared with full-term controls, and NEC development in both mouse and human was associated with increased enterocyte autophagy. Importantly, using mice in which we selectively deleted the autophagy gene ATG7 from the intestinal epithelium (ATG7(ΔIEC)), the induction of autophagy was determined to be required for and not merely a consequence of NEC, because ATG7(ΔIEC) mice were protected from NEC development. In defining the mechanisms involved, TLR4-induced autophagy led to impaired enterocyte migration both in vitro and in vivo, which in cultured enterocytes required the induction of RhoA-mediated stress fibers. These findings depart from current dogma in the field by identifying a unique effect of TLR4-induced autophagy within the intestinal epithelium in the pathogenesis of NEC and identify that the negative consequences of autophagy on enterocyte migration play an essential role in its development.


Subject(s)
Autophagy , Cell Movement , Enterocolitis, Necrotizing/etiology , Enterocytes/metabolism , Toll-Like Receptor 4/metabolism , Animals , Autophagy/genetics , Cell Line , Cell Movement/genetics , Disease Models, Animal , Enterocolitis, Necrotizing/pathology , Humans , Intestinal Mucosa/metabolism , Mice , Mice, Transgenic , Toll-Like Receptor 4/genetics , rho GTP-Binding Proteins/metabolism
6.
J Biol Chem ; 287(44): 37296-308, 2012 Oct 26.
Article in English | MEDLINE | ID: mdl-22955282

ABSTRACT

Factors regulating the proliferation and apoptosis of intestinal stem cells (ISCs) remain incompletely understood. Because ISCs exist among microbial ligands, immune receptors such as toll-like receptor 4 (TLR4) could play a role. We now hypothesize that ISCs express TLR4 and that the activation of TLR4 directly on the intestinal stem cells regulates their ability to proliferate or to undergo apoptosis. Using flow cytometry and fluorescent in situ hybridization for the intestinal stem cell marker Lgr5, we demonstrate that TLR4 is expressed on the Lgr5-positive intestinal stem cells. TLR4 activation reduced proliferation and increased apoptosis in ISCs both in vivo and in ISC organoids, a finding not observed in mice lacking TLR4 in the Lgr5-positive ISCs, confirming the in vivo significance of this effect. To define molecular mechanisms involved, TLR4 inhibited ISC proliferation and increased apoptosis via the p53-up-regulated modulator of apoptosis (PUMA), as TLR4 did not affect crypt proliferation or apoptosis in organoids or mice lacking PUMA. In vivo effects of TLR4 on ISCs required TIR-domain-containing adapter-inducing interferon-ß (TRIF) but were independent of myeloid-differentiation primary response-gene 88 (MYD88) and TNFα. Physiological relevance was suggested, as TLR4 activation in necrotizing enterocolitis led to reduced proliferation and increased apoptosis of the intestinal crypts in a manner that could be reversed by inhibition of PUMA, both globally or restricted to the intestinal epithelium. These findings illustrate that TLR4 is expressed on ISCs where it regulates their proliferation and apoptosis through activation of PUMA and that TLR4 regulation of ISCs contributes to the pathogenesis of necrotizing enterocolitis.


Subject(s)
Apoptosis Regulatory Proteins/metabolism , Apoptosis , Cell Proliferation , Intestinal Mucosa/pathology , Stem Cells/metabolism , Toll-Like Receptor 4/metabolism , Tumor Suppressor Proteins/metabolism , Adaptor Proteins, Vesicular Transport/metabolism , Animals , Apoptosis Regulatory Proteins/genetics , Apoptosis Regulatory Proteins/physiology , Enterocolitis, Necrotizing/metabolism , Enterocolitis, Necrotizing/pathology , Gene Knockout Techniques , Ileum/pathology , Intestinal Mucosa/immunology , Intestinal Mucosa/metabolism , Lipopolysaccharides/pharmacology , Mice , Mice, Inbred C57BL , Mice, Knockout , Myeloid Differentiation Factor 88/metabolism , Rats , Receptors, G-Protein-Coupled/metabolism , Receptors, Tumor Necrosis Factor, Type I/genetics , Receptors, Tumor Necrosis Factor, Type I/metabolism , Signal Transduction , Stem Cells/immunology , Stem Cells/physiology , Toll-Like Receptor 4/genetics , Transcriptional Activation , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/metabolism , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/physiology
7.
Gastroenterology ; 143(3): 708-718.e5, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22796522

ABSTRACT

BACKGROUND & AIMS: Little is known about factors that regulate intestinal epithelial differentiation; microbial recognition receptors such as Toll-like receptor (TLR)4 might be involved. We investigated whether intestinal TLR4 regulates epithelial differentiation and is involved in development of necrotizing enterocolitis (NEC) of the immature intestine. METHODS: Mice with conditional disruption of TLR4 in the intestinal epithelium and TLR4 knockout (TLR4(-/-)) mice were generated by breeding TLR4(loxp/loxp) mice with villin-cre and Ella-cre, respectively. Enterocytes that did not express or overexpressed TLR4 were created by lentiviral or adenoviral transduction. Intestinal organoids were cultured on tissue matrices. Bile acids were measured by colorimetric assays, and microbial composition was determined by 16S pyrosequencing. NEC was induced in 7- to 10-day-old mice by induction of hypoxia twice daily for 4 days. RESULTS: TLR4(-/-) mice and mice with enterocyte-specific deletion of TLR4 were protected from NEC; epithelial differentiation into goblet cells was increased via suppressed Notch signaling in the small intestinal epithelium. TLR4 also regulates differentiation of goblet cells in intestinal organoid and enterocyte cell cultures; differentiation was increased on deletion of TLR4 and restored when TLR4 was expressed ectopically. TLR4 signaling via Notch was increased in intestinal tissue samples from patients with NEC, and numbers of goblet cells were reduced. 16S pyrosequencing revealed that wild-type and TLR4-deficient mice had similar microbial profiles; increased numbers of goblet cells were observed in mice given antibiotics. TLR4 deficiency reduced levels of luminal bile acids in vivo, and addition of bile acids to TLR4-deficient cell cultures prevented differentiation of goblet cells. CONCLUSIONS: TLR4 signaling and Notch are increased in intestinal tissues of patients with NEC and required for induction of NEC in mice. TLR4 prevents goblet cell differentiation, independently of the microbiota. Bile acids might initiate goblet cell development.


Subject(s)
Cell Differentiation , Enterocolitis, Necrotizing/metabolism , Goblet Cells/metabolism , Intestine, Small/metabolism , Toll-Like Receptor 4/metabolism , Animals , Animals, Newborn , Bile Acids and Salts/metabolism , Cell Line , Disease Models, Animal , Enterocolitis, Necrotizing/etiology , Enterocolitis, Necrotizing/genetics , Enterocolitis, Necrotizing/microbiology , Enterocolitis, Necrotizing/pathology , Enterocolitis, Necrotizing/prevention & control , Goblet Cells/microbiology , Goblet Cells/pathology , Humans , Hypoxia/complications , Infant Formula , Infant, Newborn , Intestine, Small/microbiology , Intestine, Small/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Organoids , RNA Interference , Rats , Receptors, Notch/metabolism , Signal Transduction , Tissue Culture Techniques , Toll-Like Receptor 4/deficiency , Toll-Like Receptor 4/genetics , Transfection
8.
Proc Natl Acad Sci U S A ; 109(28): 11330-5, 2012 Jul 10.
Article in English | MEDLINE | ID: mdl-22733781

ABSTRACT

The fetal intestinal mucosa is characterized by elevated Toll-like receptor 4 (TLR4) expression, which can lead to the development of necrotizing enterocolitis (NEC)--a devastating inflammatory disease of the premature intestine--upon exposure to microbes. To define endogenous strategies that could reduce TLR4 signaling, we hypothesized that amniotic fluid can inhibit TLR4 signaling within the fetal intestine and attenuate experimental NEC, and we sought to determine the mechanisms involved. We show here that microinjection of amniotic fluid into the fetal (embryonic day 18.5) gastrointestinal tract reduced LPS-mediated signaling within the fetal intestinal mucosa. Amniotic fluid is abundant in EGF, which we show is required for its inhibitory effects on TLR4 signaling via peroxisome proliferator-activated receptor, because inhibition of EGF receptor (EGFR) with cetuximab or EGF-depleted amniotic fluid blocked the inhibitory effects of amniotic fluid on TLR4, whereas amniotic fluid did not prevent TLR4 signaling in EGFR- or peroxisome proliferator-activated receptor γ-deficient enterocytes or in mice deficient in intestinal epithelial EGFR, and purified EGF attenuated the exaggerated intestinal mucosal TLR4 signaling in wild-type mice. Moreover, amniotic fluid-mediated TLR4 inhibition reduced the severity of NEC in mice through EGFR activation. Strikingly, NEC development in both mice and humans was associated with reduced EGFR expression that was restored upon the administration of amniotic fluid in mice or recovery from NEC in humans, suggesting that a lack of amniotic fluid-mediated EGFR signaling could predispose to NEC. These findings may explain the unique susceptibility of premature infants to the development of NEC and offer therapeutic approaches to this devastating disease.


Subject(s)
Amniotic Fluid/metabolism , Gene Expression Regulation, Developmental , Intestinal Mucosa/metabolism , Toll-Like Receptor 4/metabolism , Animals , Cell Line , Enterocolitis, Necrotizing/metabolism , Enterocytes/metabolism , ErbB Receptors/metabolism , Humans , Infant, Newborn , Intestinal Mucosa/embryology , Intestines/embryology , Mice , Microscopy, Confocal/methods , Signal Transduction , Time Factors
9.
PLoS One ; 7(5): e37699, 2012.
Article in English | MEDLINE | ID: mdl-22629444

ABSTRACT

Although the c-Myc (Myc) oncoprotein controls mitochondrial biogenesis and multiple enzymes involved in oxidative phosphorylation (OXPHOS), the coordination of these events and the mechanistic underpinnings of their regulation remain largely unexplored. We show here that re-expression of Myc in myc-/- fibroblasts is accompanied by a gradual accumulation of mitochondrial biomass and by increases in membrane polarization and mitochondrial fusion. A correction of OXPHOS deficiency is also seen, although structural abnormalities in electron transport chain complexes (ETC) are not entirely normalized. Conversely, the down-regulation of Myc leads to a gradual decrease in mitochondrial mass and a more rapid loss of fusion and membrane potential. Increases in the levels of proteins specifically involved in mitochondrial fission and fusion support the idea that Myc affects mitochondrial mass by influencing both of these processes, albeit favoring the latter. The ETC defects that persist following Myc restoration may represent metabolic adaptations, as mitochondrial function is re-directed away from producing ATP to providing a source of metabolic precursors demanded by the transformed cell.


Subject(s)
DNA, Mitochondrial/metabolism , Fibroblasts/metabolism , Mitochondria/metabolism , Proto-Oncogene Proteins c-myc/metabolism , Animals , Cell Line , DNA, Mitochondrial/genetics , Down-Regulation , Mitochondria/genetics , Oxidative Phosphorylation , Proto-Oncogene Proteins c-myc/genetics , Rats
10.
J Immunol ; 188(9): 4543-57, 2012 May 01.
Article in English | MEDLINE | ID: mdl-22461698

ABSTRACT

Necrotizing enterocolitis (NEC) is the leading cause of gastrointestinal-related mortality in premature infants, and it develops under conditions of exaggerated TLR4 signaling in the newborn intestinal epithelium. Because NEC does not develop spontaneously, despite the presence of seemingly tonic stimulation of intestinal TLR4, we hypothesized that mechanisms must exist to constrain TLR4 signaling that become diminished during NEC pathogenesis and focused on the intracellular stress response protein and chaperone heat shock protein-70 (Hsp70). We demonstrate that the induction of intracellular Hsp70 in enterocytes dramatically reduced TLR4 signaling, as assessed by LPS-induced NF-κB translocation, cytokine expression, and apoptosis. These findings were confirmed in vivo, using mice that either globally lacked Hsp70 or overexpressed Hsp70 within the intestinal epithelium. TLR4 activation itself significantly increased Hsp70 expression in enterocytes, which provided a mechanism of autoinhibition of TLR4 signaling in enterocytes. In seeking to define the mechanisms involved, intracellular Hsp70-mediated inhibition of TLR4 signaling required both its substrate-binding EEVD domain and association with the cochaperone CHIP, resulting in ubiquitination and proteasomal degradation of TLR4. The expression of Hsp70 in the intestinal epithelium was significantly decreased in murine and human NEC compared with healthy controls, suggesting that loss of Hsp70 protection from TLR4 could lead to NEC. In support of this, intestinal Hsp70 overexpression in mice and pharmacologic upregulation of Hsp70 reversed TLR4-induced cytokines and enterocyte apoptosis, as well as prevented and treated experimental NEC. Thus, a novel TLR4 regulatory pathway exists within the newborn gut involving Hsp70 that may be pharmacologically activated to limit NEC severity.


Subject(s)
HSP70 Heat-Shock Proteins/immunology , Intestinal Mucosa/immunology , Signal Transduction/immunology , Toll-Like Receptor 4/immunology , Animals , Apoptosis/drug effects , Apoptosis/immunology , Enterocolitis, Necrotizing/immunology , Enterocolitis, Necrotizing/metabolism , Enterocolitis, Necrotizing/pathology , Female , HSP70 Heat-Shock Proteins/metabolism , Humans , Infant, Newborn , Intestinal Mucosa/metabolism , Intestinal Mucosa/pathology , Lipopolysaccharides/pharmacology , Male , Mice , NF-kappa B/immunology , NF-kappa B/metabolism , Proteasome Endopeptidase Complex/immunology , Proteasome Endopeptidase Complex/metabolism , Protein Structure, Tertiary , Proteolysis/drug effects , Signal Transduction/drug effects , Toll-Like Receptor 4/metabolism , Ubiquitin-Protein Ligases/immunology , Ubiquitin-Protein Ligases/metabolism , Ubiquitination/drug effects , Ubiquitination/immunology
11.
Biophys J ; 100(3): 535-543, 2011 Feb 02.
Article in English | MEDLINE | ID: mdl-21281567

ABSTRACT

Collective cell migration plays an important role during wound healing and embryo development. Although the exact mechanisms that coordinate such migration are still unknown, experimental studies of moving cell layers have shown that the primary interactions governing the motion of the layer are the force of lamellipodia, the adhesion of cells to the substrate, and the adhesion of cells to each other. Here, we derive a two-dimensional continuum mechanical model of cell-layer migration that is based on a novel assumption of elastic deformation of the layer and incorporates basic mechanical interactions of cells as well as cell proliferation and apoptosis. The evolution equations are solved numerically using a level set method. The model successfully reproduces data from two types of experiments: 1), the contraction of an enterocyte cell layer during wound healing; and 2), the expansion of a radially symmetric colony of MDCK cells, both in the edge migration velocity and in cell-layer density. In accord with experimental observations, and in contrast to reaction-diffusion models, this model predicts a partial wound closure if lamellipod formation is inhibited at the wound edge and gives implications of the effect of spatially restricted proliferation.


Subject(s)
Cell Movement , Models, Biological , Wound Healing , Animals , Cell Count , Cell Line , Cell Proliferation , Dogs , Enterocytes/cytology , Pseudopodia/metabolism
12.
J Immunol ; 182(1): 636-46, 2009 Jan 01.
Article in English | MEDLINE | ID: mdl-19109197

ABSTRACT

Necrotizing enterocolitis (NEC) is a common and often fatal inflammatory disorder affecting preterm infants that develops upon interaction of indigenous bacteria with the premature intestine. We now demonstrate that the developing mouse intestine shows reciprocal patterns of expression of TLR4 and TLR9, the receptor for bacterial DNA (CpG-DNA). Using a novel ultrasound-guided in utero injection system, we administered LPS directly into the stomachs of early and late gestation fetuses to induce TLR4 signaling and demonstrated that TLR4-mediated signaling within the developing intestine follows its expression pattern. Murine and human NEC were associated with increased intestinal TLR4 and decreased TLR9 expression, suggesting that reciprocal TLR4 and TLR9 signaling may occur in the pathogenesis of NEC. Enteral administration of adenovirus expressing mutant TLR4 to neonatal mice reduced the severity of NEC and increased TLR9 expression within the intestine. Activation of TLR9 with CpG-DNA inhibited LPS-mediated TLR4 signaling in enterocytes in a mechanism dependent upon the inhibitory molecule IRAK-M. Strikingly, TLR9 activation with CpG-DNA significantly reduced NEC severity, whereas TLR9-deficient mice exhibited increased NEC severity. Thus, the reciprocal nature of TLR4 and TLR9 signaling within the neonatal intestine plays a role in the development of NEC and provides novel therapeutic approaches to this disease.


Subject(s)
Enterocolitis, Necrotizing/immunology , Enterocolitis, Necrotizing/therapy , Gene Expression Regulation, Developmental/immunology , Signal Transduction/immunology , Toll-Like Receptor 4/biosynthesis , Toll-Like Receptor 4/physiology , Toll-Like Receptor 9/biosynthesis , Toll-Like Receptor 9/physiology , Animals , Cell Line , Down-Regulation/immunology , Enterocolitis, Necrotizing/embryology , Enterocolitis, Necrotizing/metabolism , Enterocytes/immunology , Enterocytes/metabolism , Genetic Therapy , Humans , Infant, Newborn , Intestinal Mucosa/embryology , Intestinal Mucosa/growth & development , Intestinal Mucosa/immunology , Intestinal Mucosa/metabolism , Mice , Mice, Inbred C3H , Mice, Inbred C57BL , Mice, Mutant Strains , Rats , Toll-Like Receptor 4/genetics , Toll-Like Receptor 4/therapeutic use , Toll-Like Receptor 9/deficiency , Toll-Like Receptor 9/genetics , Up-Regulation/immunology
13.
J Immunol ; 181(12): 8534-8543, 2008 Dec 15.
Article in English | MEDLINE | ID: mdl-19050272

ABSTRACT

The pathways that lead to the internalization of pathogens via phagocytosis remain incompletely understood. We now demonstrate a previously unrecognized role for the gap junction protein connexin43 (Cx43) in the regulation of phagocytosis by macrophages and in the host response to bacterial infection of the peritoneal cavity. Primary and cultured macrophages were found to express Cx43, which localized to the phagosome upon the internalization of IgG-opsonized particles. The inhibition of Cx43 using small interfering RNA or by obtaining macrophages from Cx43 heterozygous or knockout mice resulted in significantly impaired phagocytosis, while transfection of Cx43 into Fc-receptor expressing HeLa cells, which do not express endogenous Cx43, conferred the ability of these cells to undergo phagocytosis. Infection of macrophages with adenoviruses expressing wild-type Cx43 restored phagocytic ability in macrophages from Cx43 heterozygous or deficient mice, while infection with viruses that expressed mutant Cx43 had no effect. In understanding the mechanisms involved, Cx43 was required for RhoA-dependent actin cup formation under adherent particles, and transfection with constitutively active RhoA restored a phagocytic phenotype after Cx43 inactivation. Remarkably, mortality was significantly increased in a mouse model of bacterial peritonitis after Cx43 inhibition and in Cx43 heterozygous mice compared with untreated and wild-type counterparts. These findings reveal a novel role for Cx43 in the regulation of phagocytosis and rearrangement of the F-actin cytoskeleton, and they implicate Cx43 in the regulation of the host response to microbial infection.


Subject(s)
Connexin 43/physiology , Escherichia coli Infections/immunology , Escherichia coli Infections/mortality , Macrophages/immunology , Macrophages/microbiology , Peritonitis/immunology , Peritonitis/mortality , Animals , Cell Line , Connexin 43/biosynthesis , Connexin 43/deficiency , Connexin 43/genetics , Escherichia coli Infections/pathology , Female , HeLa Cells , Humans , Liver/cytology , Liver/embryology , Liver/immunology , Macrophages/pathology , Macrophages, Peritoneal/immunology , Macrophages, Peritoneal/microbiology , Macrophages, Peritoneal/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Peritonitis/pathology , Phagosomes/immunology , Phagosomes/metabolism , Phagosomes/microbiology , Survival Analysis
14.
J Pediatr Surg ; 43(6): 1175-81, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18558203

ABSTRACT

BACKGROUND: The early signaling events in the development of necrotizing enterocolitis (NEC) remain undefined. We have recently shown that the endotoxin (lipopolysaccharide [LPS]) receptor toll-like receptor 4 (TLR4) on enterocytes is critical in the pathogenesis of experimental NEC. Given that the membrane receptor CD14 is known to facilitate the activation of TLR4, we now hypothesize that endotoxemia induces an early upregulation of CD14 in enterocytes and that this participates in the early intestinal inflammatory response in the development of NEC. METHODS: IEC-6 enterocytes were treated with LPS (50 microg/mL), and the subcellular localization of CD14 and TLR4 was assessed by confocal microscopy. C57/Bl6 or CD14-/- mice were treated with LPS (5 mg/kg), whereas experimental NEC was induced using a combination of gavage formula feeding and intermittent hypoxia. CD14 expression was determined by sodium dodecyl sulfate-polyacrylamide gel electrophoresis and reverse transcriptase-polymerase chain reaction, and interleukin 6 was quantified by enzyme-linked immunosorbent assay and reverse transcriptase-polymerase chain reaction. RESULTS: Exposure of IEC-6 enterocytes to LPS led to an initial, transient increase in CD14 expression. The early increase in CD14 expression was associated with internalization of CD14 to a perinuclear compartment where increased colocalization with TLR4 was noted. The in vivo significance of these findings is suggested as treatment of mice with LPS led to an early increase in CD14 expression in the intestinal mucosa, whereas the persistent endotoxemia of experimental NEC was associated with decreased CD14 expression within enterocytes. CONCLUSIONS: LPS signaling in the enterocyte is marked by an early, transient increase in expression of CD14 and redistribution of the receptor. This process may contribute to the early activation of the intestinal inflammatory response that is observed in the development of NEC.


Subject(s)
Endotoxemia/physiopathology , Enterocolitis, Necrotizing/genetics , Lipopolysaccharide Receptors/metabolism , Lipopolysaccharides/pharmacology , Toll-Like Receptor 4/metabolism , Animals , Cells, Cultured , Disease Models, Animal , Endotoxins , Enterocolitis, Necrotizing/physiopathology , Enterocytes/cytology , Enterocytes/metabolism , Enzyme-Linked Immunosorbent Assay , Gene Expression Regulation , Immunohistochemistry , Intestinal Mucosa/metabolism , Intestinal Mucosa/physiopathology , Lipopolysaccharide Receptors/genetics , Mice , Mice, Inbred C57BL , Microscopy, Confocal , Probability , Random Allocation , Reference Values , Reverse Transcriptase Polymerase Chain Reaction , Sensitivity and Specificity , Signal Transduction , Toll-Like Receptor 4/genetics , Up-Regulation
15.
J Immunol ; 179(7): 4808-20, 2007 Oct 01.
Article in English | MEDLINE | ID: mdl-17878380

ABSTRACT

Necrotizing enterocolitis (NEC) is the leading cause of death from gastrointestinal disease in preterm infants and is characterized by translocation of LPS across the inflamed intestine. We hypothesized that the LPS receptor (TLR4) plays a critical role in NEC development, and we sought to determine the mechanisms involved. We now demonstrate that NEC in mice and humans is associated with increased expression of TLR4 in the intestinal mucosa and that physiological stressors associated with NEC development, namely, exposure to LPS and hypoxia, sensitize the murine intestinal epithelium to LPS through up-regulation of TLR4. In support of a critical role for TLR4 in NEC development, TLR4-mutant C3H/HeJ mice were protected from the development of NEC compared with wild-type C3H/HeOUJ littermates. TLR4 activation in vitro led to increased enterocyte apoptosis and reduced enterocyte migration and proliferation, suggesting a role for TLR4 in intestinal repair. In support of this possibility, increased NEC severity in C3H/HeOUJ mice resulted from increased enterocyte apoptosis and reduced enterocyte restitution and proliferation after mucosal injury compared with mutant mice. TLR4 signaling also led to increased serine phosphorylation of intestinal focal adhesion kinase (FAK). Remarkably, TLR4 coimmunoprecipitated with FAK, and small interfering RNA-mediated FAK inhibition restored enterocyte migration after TLR4 activation, demonstrating that the FAK-TLR4 association regulates intestinal healing. These findings demonstrate a critical role for TLR4 in the development of NEC through effects on enterocyte injury and repair, identify a novel TLR4-FAK association in regulating enterocyte migration, and suggest TLR4/FAK as a therapeutic target in this disease.


Subject(s)
Enterocolitis, Necrotizing/metabolism , Enterocolitis, Necrotizing/pathology , Intestinal Mucosa/metabolism , Intestines/pathology , Toll-Like Receptor 4/metabolism , Animals , Apoptosis , Cell Hypoxia/drug effects , Cell Line , Cell Movement , Endotoxins/pharmacology , Enterocolitis, Necrotizing/chemically induced , Enterocolitis, Necrotizing/genetics , Focal Adhesion Protein-Tyrosine Kinases/genetics , Focal Adhesion Protein-Tyrosine Kinases/metabolism , Humans , Intestines/injuries , Kinetics , Lipopolysaccharide Receptors/metabolism , Mice , Mutation/genetics , Signal Transduction , Toll-Like Receptor 4/genetics , Up-Regulation/drug effects
16.
J Leukoc Biol ; 82(5): 1257-65, 2007 Nov.
Article in English | MEDLINE | ID: mdl-17675562

ABSTRACT

Phagocytosis is the process by which microbial pathogens are engulfed by macrophages and neutrophils and represents the first line of defense against bacterial infection. The importance of phagocytosis for bacterial clearance is of particular relevance to systemic inflammatory diseases, which are associated with the development of hypoxia, yet the precise effects of hypoxia on phagocytosis remain largely unexplored. We now hypothesize that hypoxia inhibits phagocytosis in macrophages and sought to determine the mechanisms involved. Despite our initial prediction, hypoxia significantly increased the phagocytosis rate of particles in vitro by RAW264.7 and primary peritoneal macrophages and increased phagocytosis of labeled bacteria in vivo by hypoxic mice compared with normoxic controls. In understanding the mechanisms involved, hypoxia caused no changes in RhoA-GTPase signaling but increased the phosphorylation of p38-MAPK significantly. Inhibition of p38 reversed the effects of hypoxia on phagocytosis, suggesting a role for p38 in the hypoxic regulation of phagocytosis. Hypoxia also significantly increased the expression of hypoxia-inducible factor-1alpha (HIF-1alpha) in macrophages, which was reversed after p38 inhibition, suggesting a link between p38 activation and HIF-1alpha expression. It is striking that small interfering RNA knockdown of HIF-1alpha reversed the effects of hypoxia on phagocytosis, and overexpression of HIF-1alpha caused a surprising increase in phagocytosis compared with nontransfected controls, demonstrating a specific role for HIF-1alpha in the regulation of phagocytosis. These data indicate that hypoxia enhances phagocytosis in macrophages in a HIF-1alpha-dependent manner and shed light on an important role for HIF-1alpha in host defense.


Subject(s)
Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Hypoxia , Macrophages, Peritoneal/metabolism , Phagocytosis , Animals , Apoptosis , Cell Differentiation , Cells, Cultured , Escherichia coli/drug effects , Gene Expression Regulation , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/antagonists & inhibitors , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Macrophages, Peritoneal/cytology , Macrophages, Peritoneal/drug effects , Mice , Mice, Inbred C3H , Phosphorylation , RNA, Small Interfering/pharmacology , Tumor Necrosis Factor-alpha/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism , rhoA GTP-Binding Protein/metabolism
17.
J Biol Chem ; 282(35): 25852-63, 2007 Aug 31.
Article in English | MEDLINE | ID: mdl-17597062

ABSTRACT

Recent studies have shown that myostatin, first identified as a negative regulator of skeletal muscle growth, may also be involved in the formation of fibrosis within skeletal muscle. In this study, we further explored the potential role of myostatin in skeletal muscle fibrosis, as well as its interaction with both transforming growth factor-beta1 and decorin. We discovered that myostatin stimulated fibroblast proliferation in vitro and induced its differentiation into myofibroblasts. We further found that transforming growth factor-beta1 stimulated myostatin expression, and conversely, myostatin stimulated transforming growth factor-beta1 secretion in C2C12 myoblasts. Decorin, a small leucine-rich proteoglycan, was found to neutralize the effects of myostatin in both fibroblasts and myoblasts. Moreover, decorin up-regulated the expression of follistatin, an antagonist of myostatin. The results of in vivo experiments showed that myostatin knock-out mice developed significantly less fibrosis and displayed better skeletal muscle regeneration when compared with wild-type mice at 2 and 4 weeks following gastrocnemius muscle laceration injury. In wild-type mice, we found that transforming growth factor-beta1 and myostatin co-localize in myofibers in the early stages of injury. Recombinant myostatin protein stimulated myofibers to express transforming growth factor-beta1 in skeletal muscles at early time points following injection. In summary, these findings define a fibrogenic property of myostatin and suggest the existence of co-regulatory relationships between transforming growth factor-beta1, myostatin, and decorin.


Subject(s)
Cell Differentiation , Extracellular Matrix Proteins/metabolism , Fibroblasts/metabolism , Muscle, Skeletal/injuries , Muscle, Skeletal/metabolism , Muscular Diseases/metabolism , Myoblasts/metabolism , Proteoglycans/metabolism , Transforming Growth Factor beta1/metabolism , Transforming Growth Factor beta/metabolism , Animals , Cell Differentiation/drug effects , Cell Proliferation/drug effects , Decorin , Extracellular Matrix Proteins/pharmacology , Female , Fibroblasts/pathology , Fibrosis , Follistatin/biosynthesis , Mice , Muscle Fibers, Skeletal/metabolism , Muscle Fibers, Skeletal/pathology , Muscle, Skeletal/pathology , Muscular Diseases/pathology , Myoblasts/pathology , Myostatin , NIH 3T3 Cells , Proteoglycans/pharmacology , Recombinant Proteins/metabolism , Recombinant Proteins/pharmacology , Transforming Growth Factor beta/antagonists & inhibitors , Transforming Growth Factor beta/deficiency , Transforming Growth Factor beta/pharmacology , Transforming Growth Factor beta1/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...