Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
1.
J Biol Chem ; 299(8): 105027, 2023 08.
Article in English | MEDLINE | ID: mdl-37423298

ABSTRACT

Metabolism controls cellular phenotype and fate. In this report, we demonstrate that nicotinamide N-methyltransferase (NNMT), a metabolic enzyme that regulates developmental stem cell transitions and tumor progression, is highly expressed in human idiopathic pulmonary fibrosis (IPF) lungs, and is induced by the pro-fibrotic cytokine, transforming growth factor-ß1 (TGF-ß1) in lung fibroblasts. NNMT silencing reduces the expression of extracellular matrix proteins, both constitutively and in response to TGF-ß1. Furthermore, NNMT controls the phenotypic transition from homeostatic, pro-regenerative lipofibroblasts to pro-fibrotic myofibroblasts. This effect of NNMT is mediated, in part, by the downregulation of lipogenic transcription factors, TCF21 and PPARγ, and the induction of a less proliferative but more differentiated myofibroblast phenotype. NNMT confers an apoptosis-resistant phenotype to myofibroblasts that is associated with the downregulation of pro-apoptotic members of the Bcl-2 family, including Bim and PUMA. Together, these studies indicate a critical role for NNMT in the metabolic reprogramming of fibroblasts to a pro-fibrotic and apoptosis-resistant phenotype and support the concept that targeting this enzyme may promote regenerative responses in chronic fibrotic disorders such as IPF.


Subject(s)
Myofibroblasts , Nicotinamide N-Methyltransferase , Humans , Apoptosis , Basic Helix-Loop-Helix Transcription Factors/metabolism , Fibroblasts/metabolism , Fibrosis , Idiopathic Pulmonary Fibrosis/metabolism , Lung/metabolism , Myofibroblasts/metabolism , Nicotinamide N-Methyltransferase/metabolism , Transforming Growth Factor beta1/metabolism
2.
J Matern Fetal Neonatal Med ; 35(26): 10565-10576, 2022 Dec.
Article in English | MEDLINE | ID: mdl-36261134

ABSTRACT

OBJECTIVE: We sought to determine the clinical and histopathological factors linked with intestinal repair and its correlation with clinical outcomes in preterm infants following surgical necrotizing enterocolitis (NEC). METHODS: A retrospective study has compared clinical and histopathological characteristics between preterm infants with histopathological reparative changes versus non-reparative changes in resected intestinal tissue following surgical treatment of NEC. Reparative changes were defined as microscopic evidence of neovascularization, increased fibroblasts or myofibroblasts, and epithelial regeneration during histopathological examination of the most affected area of resected intestinal tissue. RESULTS: The infants with reparative changes (53/148) had significantly lower median birth weight (725 [650-963] vs. 920 [690-1320]; p = .018), higher likelihood of patent ductus arteriosus (38/53 [71.7%] vs. 48/95 [50.5%]; p = .012), longer TPN days (99 [56-147] vs. 76.5 [39-112.5]; p = .034), higher CRP levels (7.3 [3.2-13] vs. 2.6 [1.1-7.8]; p = .011) at NEC onset, and more short bowel syndrome (27/53 [54.0%] vs. 28/95 [32.2%]; p = .012). Those with reparative changes also received more Penrose drain therapy (21/53 [39.6%] vs. 14/95 [14.7%]; p = .011) and had a longer median time to laparotomy (108 h [28-216] vs. 24 [12-96]; p = .003). Epithelial regeneration observed in 6/53 (11.3%) infants lagged fibroblast proliferation and neovascularization changes in the submucosa/muscularis intestinal layers. On a multivariable logistic regression model which included histopathological and clinical factors, inflammation with a percentage <25% area involvement, time from NEC diagnosis to surgery, and Apgar score < 6 at 5 min were independently and significantly associated with higher odds reparative changes. CONCLUSION: In neonates with surgical NEC, the histopathological findings in the resected bowel are significantly associated with clinical characteristics, other histopathological findings, and outcomes. The presence of reparative changes consistent with healing is significantly associated with Apgar score, Penrose drain therapy, longer time from NEC diagnosis to surgery, and lower burden of inflammation in the resected bowel tissue in multivariable analyses. Routine histopathological grading of resected bowel and optimal use of Penrose drain therapy warrant further investigation in the care of neonates with surgical NEC.


Subject(s)
Enterocolitis, Necrotizing , Fetal Diseases , Infant, Newborn, Diseases , Infant , Female , Infant, Newborn , Humans , Infant, Premature , Enterocolitis, Necrotizing/surgery , Enterocolitis, Necrotizing/complications , Retrospective Studies , Birth Weight , Inflammation/complications
3.
Nat Aging ; 1(2): 205-217, 2021 02.
Article in English | MEDLINE | ID: mdl-34386777

ABSTRACT

Aging is a risk factor for progressive fibrotic disorders involving diverse organ systems, including the lung. Idiopathic pulmonary fibrosis, an age-associated degenerative lung disorder, is characterized by persistence of apoptosis-resistant myofibroblasts. In this report, we demonstrate that sirtuin-3 (SIRT3), a mitochondrial deacetylase, is downregulated in lungs of IPF human subjects and in mice subjected to lung injury. Over-expression of the SIRT3 cDNA via airway delivery restored capacity for fibrosis resolution in aged mice, in association with activation of the forkhead box transcription factor, FoxO3a, in fibroblasts, upregulation of pro-apoptotic members of the Bcl-2 family, and recovery of apoptosis susceptibility. While transforming growth factor-ß1 reduced levels of SIRT3 and FoxO3a in lung fibroblasts, cell non-autonomous effects involving macrophage secreted products were necessary for SIRT3-mediated activation of FoxO3a. Together, these findings reveal a novel role of SIRT3 in pro-resolution macrophage functions that restore susceptibility to apoptosis in fibroblasts via a FoxO3a-dependent mechanism.


Subject(s)
Idiopathic Pulmonary Fibrosis , Sirtuin 3 , Humans , Animals , Mice , Sirtuin 3/genetics , Lung/metabolism , Fibrosis , Idiopathic Pulmonary Fibrosis/metabolism , Gene Expression
4.
Respir Med ; 161: 105821, 2020 01.
Article in English | MEDLINE | ID: mdl-31765873

ABSTRACT

INTRODUCTION: Cellular senescence has been linked to the pathogenesis of idiopathic pulmonary fibrosis (IPF). CCN1 is a matricellular protein that has been shown to induce cellular senescence and contribute to lung fibrosis in pre-clinical models. In this report, we determined plasma CCN1 levels in patients with IPF and its potential role in clinical outcomes. METHODS AND RESULTS: We evaluated 88 patients diagnosed with IPF at the University of Alabama at Birmingham. CCN1 levels were measured in plasma specimens by ELISA. The primary outcome measure was transplant-free survival (TFS) duration. High-CCN1 levels were associated with a lower transplant-free survival independent of %FVC and %DLCO compared to patients with low plasma CCN1 (HR = 2.15; 95%CI 1.04-4.45, p = 0.04). CONCLUSION: This study demonstrates that plasma levels of CCN1 may be predictive of survival in IPF. Given the plausible role of CCN1 in cellular senescence and pathobiology of IPF, the predictive value of CCN1 in disease progression among patients with IPF warrants further investigation.


Subject(s)
Cellular Senescence , Cysteine-Rich Protein 61/blood , Idiopathic Pulmonary Fibrosis/blood , Idiopathic Pulmonary Fibrosis/etiology , Aged , Disease Progression , Enzyme-Linked Immunosorbent Assay , Female , Humans , Idiopathic Pulmonary Fibrosis/mortality , Male , Middle Aged , Outcome Assessment, Health Care , Predictive Value of Tests , Survival Rate
5.
JCI Insight ; 4(1)2019 Jan 10.
Article in English | MEDLINE | ID: mdl-30626741

ABSTRACT

Acute lung injury (ALI) is characterized by exuberant proinflammatory responses and mitochondrial dysfunction. However, the link between mitochondrial dysfunction and inflammation in ALI is not well understood. In this report, we demonstrate a critical role for the mitochondrial NAD+-dependent deacetylase, sirtuin-3 (SIRT3), in regulating macrophage mitochondrial bioenergetics, ROS formation, and proinflammatory responses. We found that SIRT3 expression was significantly diminished in lungs of mice subjected to LPS-induced ALI. SIRT3-deficient mice (SIRT3-/-) develop more severe ALI compared with wild-type controls (SIRT3+/+). Macrophages obtained from SIRT3-/- mice show significant alterations in mitochondrial bioenergetic and redox homeostasis, in association with a proinflammatory phenotype characterized by NLRP3 inflammasome activation. The SIRT3 activator viniferin restored macrophage bioenergetic function in LPS-treated macrophages. Viniferin also reduced NLRP3 activation and the production of proinflammatory cytokines, effects that were absent in SIRT3-/- macrophages. In-vivo administration of viniferin reduced production of inflammatory mediators TNF-α, MIP-2, IL-6, IL-1ß, and HMGB1, and diminished neutrophil influx and severity of endotoxin-mediated ALI; this protective effect of vinferin was abolished in SIRT3-/- mice. Taken together, our results show that the induction/activation of SIRT3 may serve as a new therapeutic strategy in ALI by modulating cellular bioenergetics, controlling inflammatory responses, and reducing the severity of lung injury.

6.
Am J Physiol Lung Cell Mol Physiol ; 312(6): L926-L935, 2017 06 01.
Article in English | MEDLINE | ID: mdl-28360109

ABSTRACT

Alveolar epithelial cell (AEC) injury and apoptosis are prominent pathological features of idiopathic pulmonary fibrosis (IPF). There is evidence of AEC plasticity in lung injury repair response and in IPF. In this report, we explore the role of focal adhesion kinase (FAK) signaling in determining the fate of lung epithelial cells in response to transforming growth factor-ß1 (TGF-ß1). Rat type II alveolar epithelial cells (RLE-6TN) were treated with or without TGF-ß1, and the expressions of mesenchymal markers, phenotype, and function were analyzed. Pharmacological protein kinase inhibitors were utilized to screen for SMAD-dependent and -independent pathways. SMAD and FAK signaling was analyzed using siRNA knockdown, inhibitors, and expression of a mutant construct of FAK. Apoptosis was measured using cleaved caspase-3 and terminal deoxynucleotidyl transferase dUTP nick end labeling (TUNEL) staining. TGF-ß1 induced the acquisition of mesenchymal markers, including α-smooth muscle actin, in RLE-6TN cells and enhanced the contraction of three-dimensional collagen gels. This phenotypical transition or plasticity, epithelial-myofibroblast plasticity (EMP), is dependent on SMAD3 and FAK signaling. FAK activation was found to be dependent on ALK5/SMAD3 signaling. We observed that TGF-ß1 induces both EMP and apoptosis in the same cell culture system but not in the same cell. While blockade of SMAD signaling inhibited EMP, it had a minimal effect on apoptosis; in contrast, inhibition of FAK signaling markedly shifted to an apoptotic fate. The data support that FAK activation determines whether AECs undergo EMP vs. apoptosis in response to TGF-ß1 stimulation. TGF-ß1-induced EMP is FAK- dependent, whereas TGF-ß1-induced apoptosis is favored when FAK signaling is inhibited.


Subject(s)
Epithelial Cells/enzymology , Focal Adhesion Protein-Tyrosine Kinases/metabolism , Lung/cytology , Signal Transduction/drug effects , Transforming Growth Factor beta/pharmacology , Animals , Apoptosis/drug effects , Cell Line , Cells, Cultured , Enzyme Activation/drug effects , Epithelial Cells/drug effects , Models, Biological , Myofibroblasts/drug effects , Myofibroblasts/metabolism , Myofibroblasts/pathology , Phenotype , Phosphorylation/drug effects , Rats , Receptors, Transforming Growth Factor beta/metabolism , Smad3 Protein/metabolism , Sus scrofa , Time Factors
7.
FASEB J ; 30(6): 2135-50, 2016 06.
Article in English | MEDLINE | ID: mdl-26884454

ABSTRACT

Matricellular proteins mediate pleiotropic effects during tissue injury and repair. CCN1 is a matricellular protein that has been implicated in angiogenesis, inflammation, and wound repair. In this study, we identified CCN1 as a gene that is differentially up-regulated in alveolar mesenchymal cells of human subjects with rapidly progressive idiopathic pulmonary fibrosis (IPF). Elevated levels of CCN1 mRNA were confirmed in lung tissues of IPF subjects undergoing lung transplantation, and CCN1 protein was predominantly localized to fibroblastic foci. CCN1 expression in ex vivo IPF lung fibroblasts correlated with gene expression of the extracellular matrix proteins, collagen (Col)1a1, Col1a2, and fibronectin as well as the myofibroblast marker, α-smooth muscle actin. RNA interference (RNAi)-mediated knockdown of CCN1 down-regulated the constitutive expression of these profibrotic genes in IPF fibroblasts. TGF-ß1, a known mediator of tissue fibrogenesis, induces gene and protein expression of CCN1 via a mothers against decapentaplegic homolog 3 (SMAD3)-dependent mechanism. Importantly, endogenous CCN1 potentiates TGF-ß1-induced SMAD3 activation and induction of profibrotic genes, supporting a positive feedback loop leading to myofibroblast activation. In vivo RNAi-mediated silencing of CCN1 attenuates fibrogenic responses to bleomycin-induced lung injury. These studies support previously unrecognized, cooperative interaction between the CCN1 matricellular protein and canonical TGF-ß1/SMAD3 signaling that promotes lung fibrosis.-Kurundkar, A. R., Kurundkar, D., Rangarajan, S., Locy, M. L., Zhou, Y., Liu, R.-M., Zmijewski, J., Thannickal, V. J. The matricellular protein CCN1 enhances TGF-ß1/SMAD3-dependent profibrotic signaling in fibroblasts and contributes to fibrogenic responses to lung injury.


Subject(s)
Cysteine-Rich Protein 61/metabolism , Fibroblasts/metabolism , Gene Expression Regulation/physiology , Lung Injury/metabolism , Smad3 Protein/metabolism , Transforming Growth Factor beta1/metabolism , Animals , Cells, Cultured , Cysteine-Rich Protein 61/genetics , Gene Knockdown Techniques , Humans , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Pulmonary Fibrosis/metabolism , RNA Interference , Receptor, Transforming Growth Factor-beta Type I , Receptors, Transforming Growth Factor beta/genetics , Receptors, Transforming Growth Factor beta/metabolism , Signal Transduction/physiology , Smad3 Protein/genetics , Transforming Growth Factor beta1/genetics , Up-Regulation
8.
Pediatr Res ; 77(4): 500-5, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25588190

ABSTRACT

BACKGROUND: Vascular endothelial growth factor (VEGF), a well-characterized regulator of angiogenesis, has been mechanistically implicated in retinal neovascularization and in the pathogenesis of retinopathy of prematurity. However, the ontogeny of VEGF expression in the human fetal retina is not well known. Because retinal vasculature grows with gestational maturation, we hypothesized that VEGF expression also increases in the midgestation human fetal eye as a function of gestational age. METHODS: To identify changes in VEGF gene expression during normal human development, we measured VEGF mRNA by quantitative PCR and measured VEGF protein by enzyme-linked immunosorbent assay and western blots in 10-24 wk gestation fetal vitreous, retina, and serum. RESULTS: VEGF mRNA expression in the retina increased with gestational age. VEGF isoform A, particularly its VEGF121 splice variant, contributed to this positive correlation. Consistent with these findings, we detected increasing VEGF121 protein concentrations in vitreous humor from fetuses of 10-24 wk gestation, while VEGF concentrations decreased in fetal serum. CONCLUSION: VEGF121 mRNA and protein concentrations increase with increasing gestational age in the developing human retina. We speculate that VEGF plays an important role in normal retinal vascular development, and that preterm delivery affects production of this vascular growth factor.


Subject(s)
Gene Expression Regulation, Developmental , RNA, Messenger/metabolism , Retina/embryology , Retinal Neovascularization , Vascular Endothelial Growth Factor A/metabolism , Vitreous Body/embryology , Actins/metabolism , Blotting, Western , Enzyme-Linked Immunosorbent Assay , Gestational Age , Glyceraldehyde-3-Phosphate Dehydrogenases/metabolism , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Infant, Newborn , Infant, Premature , RNA, Ribosomal, 18S/metabolism , Retinopathy of Prematurity/metabolism
9.
Lab Invest ; 94(2): 150-60, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24365747

ABSTRACT

Neonates and young infants exposed to extracorporeal circulation during extracorporeal membrane oxygenation (ECMO) and cardiopulmonary bypass are at risk of developing a systemic inflammatory response syndrome with multi-organ dysfunction. We used a piglet model of ECMO to investigate the hypothesis that epithelial apoptosis is an early event that precedes villous damage during ECMO-related bowel injury. Healthy 3-week-old piglets were subjected to ECMO for up to 8 h. Epithelial apoptosis was measured in histopathological analysis, nuclear imaging, and terminal deoxynucleotidyl transferase dUTP nick end labeling. Plasma intestinal fatty acid-binding protein (I-FABP) levels were measured by enzyme immunoassay. Intestinal mast cells were isolated by fluorescence-assisted cell sorting. Cleaved caspase-8, caspase-9, phospho-p38 MAPK, and fas ligand expression were investigated by immunohistochemistry, western blots, and reverse transcriptase-quantitative PCR. Piglet ECMO was associated with increased gut epithelial apoptosis. Extensive apoptotic changes were noted on villus tips and in scattered crypt cells after 2 h of ECMO. After 8 h, the villi were denuded and apoptotic changes were evident in a majority of crypt cells. Increased circulating I-FABP levels, a marker of gut epithelial injury, showed that epithelial injury occurred during ECMO. We detected increased cleaved caspase-8, but not cleaved caspase-9, in epithelial cells indicating that the extrinsic apoptotic pathway was active. ECMO was associated with increased fas ligand expression in intestinal mast cells, which was induced through activation of the p38 mitogen-activated protein kinase. We conclude that epithelial apoptosis is an early event that initiates gut mucosal injury in a piglet model of ECMO.


Subject(s)
Apoptosis/physiology , Extracorporeal Membrane Oxygenation/adverse effects , Intestinal Mucosa/injuries , Intestinal Mucosa/physiopathology , Animals , Animals, Newborn , Blotting, Western , Caspase 8/metabolism , Caspase 9/metabolism , Cell Nucleus/ultrastructure , DNA Primers/genetics , Enzyme-Linked Immunosorbent Assay , Fas Ligand Protein/metabolism , Fatty Acid-Binding Proteins/blood , Flow Cytometry , Immunohistochemistry , In Situ Nick-End Labeling , Intestinal Mucosa/cytology , Reverse Transcriptase Polymerase Chain Reaction , Statistics, Nonparametric , Swine , p38 Mitogen-Activated Protein Kinases/metabolism
10.
Am J Physiol Gastrointest Liver Physiol ; 303(1): G93-102, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22538401

ABSTRACT

Necrotizing enterocolitis (NEC) is an inflammatory bowel necrosis of premature infants. In tissue samples of NEC, we identified numerous macrophages and a few neutrophils but not many lymphocytes. We hypothesized that these pathoanatomic characteristics of NEC represent a common tissue injury response of the gastrointestinal tract to a variety of insults at a specific stage of gut development. To evaluate developmental changes in mucosal inflammatory response, we used trinitrobenzene sulfonic acid (TNBS)-induced inflammation as a nonspecific insult and compared mucosal injury in newborn vs. adult mice. Enterocolitis was induced in 10-day-old pups and adult mice (n = 25 animals per group) by administering TNBS by gavage and enema. Leukocyte populations were enumerated in human NEC and in murine TNBS-enterocolitis using quantitative immunofluorescence. Chemokine expression was measured using quantitative polymerase chain reaction, immunoblots, and immunohistochemistry. Macrophage recruitment was investigated ex vivo using intestinal tissue-conditioned media and bone marrow-derived macrophages in a microchemotaxis assay. Similar to human NEC, TNBS enterocolitis in pups was marked by a macrophage-rich leukocyte infiltrate in affected tissue. In contrast, TNBS-enterocolitis in adult mice was associated with pleomorphic leukocyte infiltrates. Macrophage precursors were recruited to murine neonatal gastrointestinal tract by the chemokine CXCL5, a known chemoattractant for myeloid cells. We also demonstrated increased expression of CXCL5 in surgically resected tissue samples of human NEC, indicating that a similar pathway was active in NEC. We concluded that gut mucosal injury in the murine neonate is marked by a macrophage-rich leukocyte infiltrate, which contrasts with the pleomorphic leukocyte infiltrates in adult mice. In murine neonatal enterocolitis, macrophages were recruited to the inflamed gut mucosa by the chemokine CXCL5, indicating that CXCL5 and its cognate receptor CXCR2 merit further investigation as potential therapeutic targets in NEC.


Subject(s)
Intestinal Mucosa/pathology , Macrophages/physiology , Aging/physiology , Animals , Animals, Newborn , Blotting, Western , Chemokine CXCL5/physiology , Chemotaxis, Leukocyte/drug effects , Chemotaxis, Leukocyte/physiology , Denaturing Gradient Gel Electrophoresis , Enzyme-Linked Immunosorbent Assay , Humans , Immunohistochemistry , In Vitro Techniques , Infant, Newborn , Infant, Premature , Inflammation/pathology , Intestinal Diseases/chemically induced , Intestinal Diseases/pathology , Intestinal Mucosa/cytology , Mice , Neutrophil Infiltration/physiology , Polymerase Chain Reaction , Risk Factors , Trinitrobenzenesulfonic Acid
11.
Am J Pathol ; 180(2): 626-35, 2012 02.
Article in English | MEDLINE | ID: mdl-22155109

ABSTRACT

An immature intestinal epithelial barrier may predispose infants and children to many intestinal inflammatory diseases, such as infectious enteritis, inflammatory bowel disease, and necrotizing enterocolitis. Understanding the factors that regulate gut barrier maturation may yield insight into strategies to prevent these intestinal diseases. The claudin family of tight junction proteins plays an important role in regulating epithelial paracellular permeability. Previous reports demonstrate that rodent intestinal barrier function matures during the first 3 weeks of life. We show that murine paracellular permeability markedly decreases during postnatal maturation, with the most significant change occurring between 2 and 3 weeks. Here we report for the first time that commensal bacterial colonization induces intestinal barrier function maturation by promoting claudin 3 expression. Neonatal mice raised on antibiotics or lacking the toll-like receptor adaptor protein MyD88 exhibit impaired barrier function and decreased claudin 3 expression. Furthermore, enteral administration of either live or heat-killed preparations of the probiotic Lactobacillus rhamnosus GG accelerates intestinal barrier maturation and induces claudin 3 expression. However, live Lactobacillus rhamnosus GG increases mortality. Taken together, these results support a vital role for intestinal flora in the maturation of intestinal barrier function. Probiotics may prevent intestinal inflammatory diseases by regulating intestinal tight junction protein expression and barrier function. The use of heat-killed probiotics may provide therapeutic benefit while minimizing adverse effects.


Subject(s)
Claudins/metabolism , Intestinal Mucosa/metabolism , Lacticaseibacillus rhamnosus/physiology , Probiotics/pharmacology , Animals , Animals, Newborn , Anti-Bacterial Agents/pharmacology , Claudin-3 , Female , Intestines/microbiology , Male , Mice , Mice, Inbred C57BL , Myeloid Differentiation Factor 88/deficiency , Permeability , RNA, Messenger/metabolism , Signal Transduction/physiology , Toll-Like Receptors/physiology
12.
Mol Carcinog ; 50(7): 516-27, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21308804

ABSTRACT

Organ transplant recipients (OTRs) develop multiple aggressive and metastatic non-melanoma skin cancers (NMSCs). Yet, the underlying mechanism remains elusive. Employing a variety of immune-compromised murine models, immunoblotting, immunohistochemical and immunofluorescence techniques, we show that human squamous xenograft tumors in nude mice grow faster and become significantly larger in size following treatment with the immunosuppressive drug, cyclosporine A (CsA). Re-injected tumor cells isolated from CsA-treated xenografts continued to form larger tumors in nude mice than those from vehicle-controls and retained the CsA-signatures of calcineurin signaling inhibition. Similar results were obtained when these tumors were grown in SCID-beige mice or in immuno-competent mice inoculated with syngeinic tumor cells. Consistently, tumors in the CsA group manifested enhanced cellular proliferation and decreased apoptosis. Tumors in CsA-treated animals also showed an augmented epithelial-mesenchymal transition (EMT) characterized by an increased expression of fibronectin, α-SMA, vimentin, N-cadherin, MMP-9/-2, snail and twist with a concomitant decrease in E-cadherin. CsA-treated xenograft tumors manifested increased TGFß1 expression and TGFß-dependent signaling characterized by increased nuclear p-Smad 2/3. Our data demonstrate that CsA alters the phenotype of skin SCCs to an invasive and aggressive tumor-type by enhancing expression of proteins regulating EMT acting through the TGFß1 signaling pathway providing at least one unique mechanism by which multiple aggressive and metastatic NMSCs develop in OTRs.


Subject(s)
Carcinoma, Squamous Cell/pathology , Cyclosporine/pharmacology , Epithelial-Mesenchymal Transition/drug effects , Signal Transduction , Skin Neoplasms/pathology , Transforming Growth Factor beta/metabolism , Animals , Apoptosis/drug effects , Blotting, Western , Carcinoma, Squamous Cell/metabolism , Cell Division/drug effects , Cell Line, Tumor , Female , Fluorescent Antibody Technique , Humans , In Situ Nick-End Labeling , Mice , Mice, Nude , Reverse Transcriptase Polymerase Chain Reaction , Skin Neoplasms/metabolism , Transplantation, Heterologous
13.
Am J Physiol Gastrointest Liver Physiol ; 299(3): G614-22, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20539009

ABSTRACT

Necrotizing enterocolitis (NEC) is a devastating intestinal disease of premature infants. Epidermal growth factor (EGF) is one of the most promising candidates in NEC prophylaxis. Autophagy regulates cell homeostasis, but uncontrolled activation of autophagy may lead to cellular injury. The aim was to evaluate the effects of EGF on intestinal autophagy in epithelial cells and in the rat NEC model and measure autophagy in NEC patients. Intestinal epithelial cells (IEC-6) and the rat NEC model were used to study the effect of EGF on intestinal autophagy. Protein levels of Beclin 1 and LC3II were measured in the intestinal epithelium in both in vivo and in vitro models. Ultrastructural changes in intestinal epithelium were studied by electron microscopy. Expression of Beclin 1, LC3II, and p62 protein was evaluated in biopsies from NEC patients. Autophagy was induced in IEC-6 cells and inhibited by adding EGF into the culture. In the rat NEC model, EGF treatment of NEC reduced expression of Beclin 1 and LC3II in ileal epithelium. Morphologically, typical signs of autophagy were observed in the epithelium of the NEC group, but not in the EGF group. A strong signal for Beclin 1 and LC3II was detected in the intestine from patients with NEC. Autophagy is activated in the intestinal epithelium of NEC patients and in the ileum of NEC rats. Supplementation of EGF blocks intestinal autophagy in both in vivo and in vitro conditions. Results from this study indicate that EGF-mediated protection against NEC injury is associated with regulation of intestinal autophagy.


Subject(s)
Autophagy , Enterocolitis, Necrotizing/drug therapy , Epidermal Growth Factor/pharmacology , Epidermal Growth Factor/therapeutic use , Intestinal Mucosa/pathology , Administration, Oral , Animals , Animals, Newborn , Apoptosis Regulatory Proteins/metabolism , Beclin-1 , Cell Line , Disease Models, Animal , Enterocolitis, Necrotizing/pathology , Epidermal Growth Factor/administration & dosage , Gene Expression Regulation/drug effects , Gene Expression Regulation/physiology , Humans , Incidence , Intestinal Mucosa/drug effects , Microtubule-Associated Proteins/metabolism , Rats , Rats, Sprague-Dawley
14.
Pediatr Res ; 68(2): 128-33, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20442689

ABSTRACT

Extracorporeal membrane oxygenation (ECMO) is an important life-support system used in neonates and young children with intractable cardiorespiratory failure. In this study, we used our porcine neonatal model of venoarterial ECMO to investigate whether ECMO causes gut barrier dysfunction. We subjected 3-wk-old previously healthy piglets to venoarterial ECMO for up to 8 h and evaluated gut mucosal permeability, bacterial translocation, plasma levels of bacterial products, and ultrastructural changes in gut epithelium. We also measured plasma lipopolysaccharide (LPS) levels in a small cohort of human neonates receiving ECMO. In our porcine model, ECMO caused a rapid increase in gut mucosal permeability within the first 2 h of treatment, leading to a 6- to 10-fold rise in circulating bacterial products. These changes in barrier function were associated with cytoskeletal condensation in epithelial cells, which was explained by phosphorylation of a myosin II regulatory light chain. In support of these findings, we also detected elevated plasma LPS levels in human neonates receiving ECMO, indicating a similar loss of gut barrier function in these infants. On the basis of these data, we conclude that ECMO is an independent cause of gut barrier dysfunction and bacterial translocation may be an important contributor to ECMO-related inflammation.


Subject(s)
Animals, Newborn , Cell Membrane Permeability , Extracorporeal Membrane Oxygenation/adverse effects , Intestinal Mucosa/pathology , Animals , Bacteria/metabolism , Child , Cytoskeleton/metabolism , Cytoskeleton/ultrastructure , Gene Expression , Humans , Infant, Newborn , Intestinal Absorption , Intestinal Mucosa/metabolism , Intestinal Mucosa/microbiology , Intestinal Mucosa/ultrastructure , Lipopolysaccharides/blood , Swine , Tight Junctions/physiology , Tight Junctions/ultrastructure
15.
Am J Physiol Cell Physiol ; 298(6): C1538-48, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20219948

ABSTRACT

HDL and its major protein component apolipoprotein A-I (apoA-I) exert anti-inflammatory effects, inhibit monocyte chemotaxis/adhesion, and reduce vascular macrophage content in inflammatory conditions. In this study, we tested the hypothesis that the apoA-I mimetic 4F modulates the function of monocyte-derived macrophages (MDMs) by regulating the expression of key cell surface receptors on MDMs. Primary human monocytes and THP-1 cells were treated with 4F, apoA-I, or vehicle for 7 days and analyzed for expression of cell surface markers, adhesion to human endothelial cells, phagocytic function, cholesterol efflux capacity, and lipid raft organization. 4F and apoA-I treatment decreased the expression of HLA-DR, CD86, CD11b, CD11c, CD14, and Toll-like receptor-4 (TLR-4) compared with control cells, suggesting the induction of monocyte differentiation. Both treatments abolished LPS-induced mRNA for monocyte chemotactic protein-1 (MCP-1), macrophage inflammatory protein-1 (MIP-1), regulated on activation, normal T-expressed and presumably secreted (RANTES), IL-6, and TNF-alpha but significantly upregulated LPS-induced IL-10 expression. Moreover, 4F and apoA-I induced a 90% reduction in the expression of CD49d, a ligand for the VCAM-1 receptor, with a concurrent decrease in monocyte adhesion (55% reduction) to human endothelial cells and transendothelial migration (34 and 27% for 4F and apoA-I treatments) compared with vehicle treatment. In addition, phagocytosis of dextran-FITC beads was inhibited by 4F and apoA-I, a response associated with reduced expression of CD32. Finally, 4F and apoA-I stimulated cholesterol efflux from MDMs, leading to cholesterol depletion and disruption of lipid rafts. These data provide evidence that 4F, similar to apoA-I, induces profound functional changes in MDMs, possibly due to differentiation to an anti-inflammatory phenotype.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Apolipoprotein A-I/pharmacology , Macrophages/drug effects , Antigens, CD/metabolism , Cell Adhesion/drug effects , Cell Adhesion Molecules/metabolism , Cells, Cultured , Cholesterol/metabolism , Cytokines/genetics , Cytokines/metabolism , Endothelial Cells/drug effects , Endothelial Cells/immunology , Endothelial Cells/metabolism , HLA-DR Antigens/metabolism , Humans , Inflammation Mediators/metabolism , Leukocyte Rolling/drug effects , Lipopolysaccharides/pharmacology , Macrophages/immunology , Macrophages/metabolism , Membrane Microdomains/drug effects , Membrane Microdomains/metabolism , Molecular Mimicry , Phagocytosis/drug effects , Phenotype , RNA, Messenger/metabolism
16.
Lab Invest ; 90(1): 128-39, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19901912

ABSTRACT

Extracorporeal membrane oxygenation (ECMO) is a life-saving support system used in neonates and young children with severe cardiorespiratory failure. Although ECMO has reduced mortality in these critically ill patients, almost all patients treated with ECMO develop a systemic inflammatory response syndrome (SIRS) characterized by a 'cytokine storm', leukocyte activation, and multisystem organ dysfunction. We used a neonatal porcine model of ECMO to investigate whether rising plasma concentrations of inflammatory cytokines during ECMO reflect de novo synthesis of these mediators in inflamed tissues, and therefore, can be used to assess the severity of ECMO-related SIRS. Previously healthy piglets (3-week-old) were subjected to venoarterial ECMO for up to 8 h. SIRS was assessed by histopathological analysis, measurement of neutrophil activation (flow cytometry), plasma cytokine concentrations (enzyme immunoassays), and tissue expression of inflammatory genes (PCR/western blots). Mast cell degranulation was investigated by measurement of plasma tryptase activity. Porcine neonatal ECMO was associated with systemic inflammatory changes similar to those seen in human neonates. Tumor necrosis factor-alpha (TNF-alpha) and interleukin-8 (IL-8) concentrations rose rapidly during the first 2 h of ECMO, faster than the tissue expression of these cytokines. ECMO was associated with increased plasma mast cell tryptase activity, indicating that increased plasma concentrations of inflammatory cytokines during ECMO may result from mast cell degranulation and associated release of preformed cytokines stored in mast cells. TNF-alpha and IL-8 concentrations rose faster in plasma than in the peripheral tissues during ECMO, indicating that rising plasma levels of these cytokines immediately after the initiation of ECMO may not reflect increasing tissue synthesis of these cytokines. Mobilization of preformed cellular stores of inflammatory cytokines such as in mucosal mast cells may have an important pathophysiological role in ECMO-related SIRS.


Subject(s)
Cytokines/metabolism , Extracorporeal Membrane Oxygenation/adverse effects , Inflammation Mediators/metabolism , Intestinal Mucosa/metabolism , Systemic Inflammatory Response Syndrome/etiology , Systemic Inflammatory Response Syndrome/metabolism , Animals , Animals, Newborn , C-Reactive Protein/metabolism , Cell Degranulation , Cytokines/blood , Cytokines/genetics , Female , Hemodynamics , Inflammation Mediators/blood , Interleukin-8/blood , Leukocyte Count , Male , Mast Cells/metabolism , Neutrophil Activation , Osmolar Concentration , Swine , Systemic Inflammatory Response Syndrome/pathology , Systemic Inflammatory Response Syndrome/physiopathology , Time Factors , Transcriptional Activation , Tumor Necrosis Factor-alpha/biosynthesis , Tumor Necrosis Factor-alpha/blood , Tumor Necrosis Factor-alpha/metabolism
17.
Am J Physiol Gastrointest Liver Physiol ; 297(1): G1-G10, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19443732

ABSTRACT

Macrophages are first seen in the fetal intestine at 11-12 wk and rapidly increase in number during the 12- to 22-wk period of gestation. The development of macrophage populations in the fetal intestine precedes the appearance of lymphocytes and neutrophils and does not require the presence of dietary or microbial antigens. In this study, we investigated the role of chemerin, a recently discovered, relatively selective chemoattractant for macrophages, in the recruitment of macrophage precursors to the fetal intestine. Chemerin mRNA/protein expression was measured in jejunoileal tissue from 10- to 24-wk human fetuses, neonates operated for intestinal obstruction, and adults undergoing bariatric surgery. The expression of chemerin in intestinal epithelial cells (IECs) was confirmed by using cultured primary IECs and IEC-like cell lines in vitro. The regulatory mechanisms involved in chemerin expression were investigated by in silico and immunolocalization techniques. IECs in the fetal, but not mature, intestine express chemerin. Chemerin expression peaked in the fetal intestine at 20-24 wk and then decreased to original low levels by full term. During the 10- to 24-wk period, chemerin accounted for most of the macrophage chemotactic activity of cultured fetal IECs. The maturational changes in chemerin expression correlated with the expression of retinoic acid receptor-beta in the intestine. Chemerin is an important mediator of epithelial-macrophage cross talk in the fetal/premature, but not in the mature, intestine. Understanding the regulation of the gut macrophage pool is an important step in development of novel strategies to boost mucosal immunity in premature infants and other patient populations at risk of microbial translocation.


Subject(s)
Chemokines/metabolism , Chemotaxis , Epithelial Cells/immunology , Ileum/immunology , Jejunum/immunology , Macrophages/immunology , Adult , Amino Acid Sequence , Caco-2 Cells , Chemokines/genetics , Culture Media, Conditioned/metabolism , Fetus/immunology , Gene Expression Regulation, Developmental , Gestational Age , Humans , Ileum/embryology , Immunohistochemistry , Infant, Newborn , Intercellular Signaling Peptides and Proteins , Jejunum/embryology , Molecular Sequence Data , Promoter Regions, Genetic , RNA, Messenger/metabolism , Receptors, Retinoic Acid/metabolism , Reverse Transcriptase Polymerase Chain Reaction
SELECTION OF CITATIONS
SEARCH DETAIL