Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
Am J Physiol Gastrointest Liver Physiol ; 314(6): G655-G667, 2018 06 01.
Article in English | MEDLINE | ID: mdl-29446653

ABSTRACT

Liver ischemia-reperfusion (I/R) injury occurs through induction of oxidative stress and release of damage-associated molecular patterns (DAMPs), including cytosolic DNA released from dysfunctional mitochondria or from the nucleus. Cyclic guanosine monophosphate-adenosine monophosphate (cGAMP) synthase (cGAS) is a cytosolic DNA sensor known to trigger stimulator of interferon genes (STING) and downstream type 1 interferon (IFN-I) pathways, which are pivotal innate immune system responses to pathogen. However, little is known about the role of cGAS/STING in liver I/R injury. We subjected C57BL/6 (WT), cGAS knockout (cGAS-/-), and STING-deficient (STINGgt/gt) mice to warm liver I/R injury and that found cGAS-/- mice had significantly increased liver injury compared with WT or STINGgt/gt mice, suggesting a protective effect of cGAS independent of STING. Liver I/R upregulated cGAS in vivo and also in vitro in hepatocytes subjected to anoxia/reoxygenation (A/R). We confirmed a previously published finding that hepatocytes do not express STING under normoxic conditions or after A/R. Hepatocytes and liver from cGAS-/- mice had increased cell death and reduced induction of autophagy under hypoxic conditions as well as increased apoptosis. Protection could be restored in cGAS-/- hepatocytes by overexpression of cGAS or by pretreatment of mice with autophagy inducer rapamycin. Our findings indicate a novel protective role for cGAS in the regulation of autophagy during liver I/R injury that occurs independently of STING. NEW & NOTEWORTHY Our studies are the first to document the important role of cGAS in the acute setting of sterile injury induced by I/R. Specifically, we provide evidence that cGAS protects liver from I/R injury in a STING-independent manner.


Subject(s)
Autophagy/physiology , Interferon Type I , Liver , Nucleotides, Cyclic/metabolism , Nucleotidyltransferases/metabolism , Reperfusion Injury , Animals , Apoptosis/physiology , DNA Nucleotidyltransferases/physiology , Interferon Inducers/metabolism , Interferon Type I/genetics , Interferon Type I/metabolism , Liver/blood supply , Liver/metabolism , Liver/pathology , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice , Mice, Inbred C57BL , Protective Agents/metabolism , Reperfusion Injury/metabolism , Reperfusion Injury/prevention & control , Signal Transduction
2.
Am J Physiol Gastrointest Liver Physiol ; 308(10): G852-62, 2015 May 15.
Article in English | MEDLINE | ID: mdl-25747352

ABSTRACT

Myocytes are nonhemopoietic in origin and functionally essential in generating gastrointestinal motility. In endotoxemia, a rapid-onset nonhemopoietic mechanism potently triggers early ileus in a Toll-like receptor 4 (TLR4)/myeloid differentiation primary response gene 88 (MyD88)-dependent manner. Moreover, synergistically with hemopoietic cells, nonhemopoietic cells escalate late ileus via an IL-6 receptor-dependent inflammation-driven pathway. We therefore specifically investigated the role of myocytes in TLR4-triggered inflammation and ileus. TLR4(+/+), TLR4(-/-), bmTLR4(+/+)/TLR4(-/-) chimera, SM22-Cre(-/-)TLR4(flox/flox), and selective myocyte TLR4-deficient (SM22-Cre(+/-)TLR4(flox/flox)) mice were injected intraperitoneally with purified lipopolysaccharide. SM22-driven Cre recombinase activity was selectively detected in cardiac, gastrointestinal, skeletal, and vascular myocytes, of small-sized vessels in a two-color fluorescent Cre reporter mouse. In contrast to nonhemopoietic TLR4 deficiency, deletion of myocyte TLR4 signaling prevented neither endotoxin-induced suppression of spontaneous jejunal contractility in vitro nor early ileus in vivo at 6 h. Circulating plasma colony-stimulating factor 3 was greatly elevated during endotoxemia, independent of myocyte TLR4 signaling or time. TLR4 activation of myocytes contributed significantly to an early enteric IL-6 mRNA induction and systemic IL-6 release, as well as to a late increase in circulating chemokine (C-X-C motif) ligand 1 (CXCL1) and IL-17. Consequently, inhibition of myocyte TLR4 signaling allowed functional recovery of motility by preventing inflammation-driven late ileus at 24 h. Direct TLR4 activation of myocytes is not responsible for nonhemopoietic-mediated early ileus. However, myocytes are proinflammatory cells that potently drive enteric and systemic inflammation, subsequently fueling late mediator-triggered ileus. Specifically, the myocyte TLR4-dependent inflammatory signature of elevated plasma IL-6, CXCL1, and IL-17 is strongly associated with late rodent ileus.


Subject(s)
Chemokines/immunology , Ileitis/immunology , Ileitis/pathology , Ileus/immunology , Ileus/pathology , Muscle Cells/immunology , Toll-Like Receptor 4/immunology , Animals , Ileitis/chemically induced , Immunologic Factors/immunology , Lipopolysaccharides , Male , Mice , Mice, Inbred C57BL , Muscle Cells/pathology
3.
Hepatology ; 58(1): 374-87, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23460269

ABSTRACT

UNLABELLED: Ischemia-reperfusion (I/R) injury is a process whereby an initial hypoxic insult and subsequent return of blood flow leads to the propagation of innate immune responses and organ injury. The necessity of the pattern recognition receptor, Toll-like receptor (TLR)4, for this innate immune response has been previously shown. However, TLR4 is present on various cell types of the liver, both immune and nonimmune cells. Therefore, we sought to determine the role of TLR4 in individual cell populations, specifically, parenchymal hepatocytes (HCs), myeloid cells, including Kupffer cells, and dendritic cells (DCs) subsequent to hepatic I/R. When HC-specific (Alb-TLR4(-/-) ) and myeloid-cell-specific (Lyz-TLR4(-/-) ) TLR4 knockout (KO) mice were subjected to warm hepatic ischemia, there was significant protection in these mice, compared to wild type (WT). However, the protection afforded in these two strains was significantly less than global TLR4 KO (TLR4(-/-) ) mice. DC-specific TLR4(-/-) (CD11c-TLR4(-/-) ) mice had significantly increased hepatocellular damage, compared to WT mice. Circulating levels of high-mobility group box 1 (HMGB1) were significantly reduced in Alb-TLR4(-/-) mice, compared to WT, Lyz-TLR4(-/-) , CD11c-TLR4(-/-) mice and equivalent to global TLR4(-/-) mice, suggesting that TLR4-mediated HMGB1 release from HCs may be a source of HMGB1 after I/R. HCs exposed to hypoxia responded by rapidly phosphorylating the mitogen-activated protein kinases, c-Jun-N-terminal kinase (JNK) and p38, in a TLR4-dependent manner; inhibition of JNK decreased release of HMGB1 after both hypoxia in vitro and I/R in vivo. CONCLUSION: These results provide insight into the individual cellular response of TLR4. The parenchymal HC is an active participant in sterile inflammatory response after I/R through TLR4-mediated activation of proinflammatory signaling and release of danger signals, such as HMGB1.


Subject(s)
Hepatocytes/immunology , Immunity, Innate/physiology , Liver/immunology , Reperfusion Injury/immunology , Toll-Like Receptor 4/physiology , Animals , Dendritic Cells/immunology , HMGB1 Protein/metabolism , JNK Mitogen-Activated Protein Kinases/metabolism , Kupffer Cells/immunology , Male , Mice , Mice, Knockout , Myeloid Cells/immunology , Toll-Like Receptor 4/deficiency , p38 Mitogen-Activated Protein Kinases/metabolism
4.
J Surg Res ; 166(2): e149-56, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21276982

ABSTRACT

BACKGROUND: Post-traumatic inflammatory changes have been identified as major causes of altered organ function and failure. Both hemorrhage and soft tissue damage induce these inflammatory changes. Exposure to heterologous bone in animal models has recently been shown to mimic this inflammatory response in a stable and reproducible fashion. This follow-up study tests the hypothesis that inflammatory responses are comparable between a novel trauma model ("pseudofracture", PFx) and a bilateral femur fracture (BFF) model. MATERIALS AND METHODS: In C57BL/6 mice, markers for remote organ dysfunction and inflammatory responses were compared in four groups (control/sham/BFF/PFx) at the time points 2, 4, and 6 h. RESULTS: Hepatocellular damage in BFF and PFx was highly comparable in extent and evolution, as shown by similar levels of NFkappaB activation and plasma ALT. Pulmonary inflammatory responses were also comparably elevated in both trauma models as early as 2 h after trauma as measured by myeloperoxidase activity (MPO). Muscle damage was provoked in both BFF and PFx mice over the time course, although BFF induced significantly higher AST and CK levels. IL-6 levels were also similar with early and sustained increases over time in both trauma models. CONCLUSIONS: Both BFF and PFx create similar reproducible inflammatory and remote organ responses. PFx will be a useful model to study longer term inflammatory effects that cannot be studied using BFF.


Subject(s)
Crush Syndrome/immunology , Femoral Fractures/immunology , Inflammation/immunology , Leg Injuries/immunology , Soft Tissue Injuries/immunology , Acute Lung Injury/immunology , Acute Lung Injury/pathology , Animals , Aspartate Aminotransferases/blood , Creatine Kinase/blood , Crush Syndrome/pathology , Disease Models, Animal , Femoral Fractures/pathology , Hemorrhage/immunology , Hemorrhage/pathology , Immune Tolerance/physiology , Inflammation/pathology , Interleukin-6/blood , Leg Injuries/pathology , Liver Diseases/immunology , Liver Diseases/pathology , Male , Mice , Mice, Inbred C57BL , Muscle, Skeletal/immunology , Muscle, Skeletal/injuries , Muscle, Skeletal/pathology , Soft Tissue Injuries/pathology
5.
Gastroenterology ; 138(1): 185-96, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19786028

ABSTRACT

BACKGROUND & AIMS: Necrotizing enterocolitis (NEC), the leading cause of gastrointestinal death from gastrointestinal disease in preterm infants, is characterized by exaggerated TLR4 signaling and decreased enterocyte proliferation through unknown mechanisms. Given the importance of beta-catenin in regulating proliferation of many cell types, we hypothesize that TLR4 impairs enterocyte proliferation in NEC via impaired beta-catenin signaling. METHODS: Enterocyte proliferation was detected in IEC-6 cells or in ileum or colon from wild-type, TLR4-mutant, or TLR4(-/-) mice after induction of NEC or endotoxemia. beta-Catenin signaling was assessed by cell fractionation or immunoconfocal microscopy to detect its nuclear translocation. Activation and inhibition of beta-catenin were achieved via cDNA or small interfering RNA, respectively. TLR4 in the intestinal mucosa was inhibited with adenoviruses expressing dominant-negative TLR4. RESULTS: TLR4 activation significantly impaired enterocyte proliferation in the ileum but not colon in newborn but not adult mice and in IEC-6 enterocytes. beta-Catenin activation reversed these effects in vitro. To determine the mechanisms involved, TLR4 activation phosphorylated the upstream inhibitory kinase GSK3beta, causing beta-catenin degradation. NEC in both mouse and humans was associated with decreased beta-catenin and increased mucosal GSK3beta expression. Strikingly, the inhibition of enterocyte beta-catenin signaling in NEC could be reversed, and enterocyte proliferation restored, through adenoviral-mediated inhibition of TLR4 signaling in the small intestinal mucosa. CONCLUSION: We now report a novel pathway linking TLR4 with inhibition of beta-catenin signaling via GSK3beta activation, leading to reduced enterocyte proliferation in vitro and in vivo. These data provide additional insights into the pathogenesis of diseases of intestinal inflammation such as NEC.


Subject(s)
Enterocolitis, Necrotizing/metabolism , Enterocytes/cytology , Enterocytes/metabolism , Toll-Like Receptor 4/metabolism , beta Catenin/metabolism , Adenoviridae/genetics , Animals , Cell Division/drug effects , Cell Division/physiology , Cells, Cultured , Colon/pathology , Enterocolitis, Necrotizing/pathology , Glycogen Synthase Kinase 3/metabolism , Glycogen Synthase Kinase 3 beta , Humans , Ileum/pathology , Infant, Newborn , Intestinal Mucosa/metabolism , Intestinal Mucosa/pathology , Lipopolysaccharides/pharmacology , Mice , Mice, Inbred C3H , Mice, Mutant Strains , Signal Transduction/physiology , Toll-Like Receptor 4/genetics
6.
J Biol Chem ; 285(7): 4995-5002, 2010 Feb 12.
Article in English | MEDLINE | ID: mdl-20007974

ABSTRACT

Toll-like receptor-4 (TLR4) is the receptor for bacterial lipopolysaccharide, yet it may also respond to a variety of endogenous molecules. Necrotizing enterocolitis (NEC) is the leading cause of death from gastrointestinal disease in newborn infants and is characterized by intestinal mucosal destruction and impaired enterocyte migration due to increased TLR4 signaling on enterocytes. The endogenous ligands for TLR4 that lead to impaired enterocyte migration remain unknown. High mobility group box-1 (HMGB1) is a DNA-binding protein that is released from injured cells during inflammation. We thus hypothesize that extracellular HMGB1 inhibits enterocyte migration via activation of TLR4 and sought to define the pathways involved. We now demonstrate that murine and human NEC are associated with increased intestinal HMGB1 expression, that serum HMGB1 is increased in murine NEC, and that HMGB1 inhibits enterocyte migration in vitro and in vivo in a TLR4-dependent manner. This finding was unique to enterocytes as HMGB1 enhanced migration of inflammatory cells in vitro and in vivo. In seeking to understand the mechanisms involved, TLR4-dependent HMGB1 signaling increased RhoA activation in enterocytes, increased phosphorylation of focal adhesion kinase, and increased phosphorylation of cofilin, resulting in increased stress fibers and focal adhesions. Using single cell force traction microscopy, the net effect of HMGB1 signaling was a TLR4-dependent increase in cell force adhesion, accounting for the impaired enterocyte migration. These findings demonstrate a novel pathway by which TLR4 activation by HMGB1 delays mucosal repair and suggest a novel potential therapeutic target in the amelioration of intestinal inflammatory diseases like NEC.


Subject(s)
Cell Movement/drug effects , Enterocytes/cytology , HMGB1 Protein/metabolism , HMGB1 Protein/pharmacology , Intestinal Mucosa/metabolism , Toll-Like Receptor 4/metabolism , Actins/metabolism , Animals , Cell Line , Cell Movement/genetics , Chemotaxis/drug effects , Cytoskeleton/drug effects , Cytoskeleton/metabolism , Enterocolitis, Necrotizing/metabolism , Enterocytes/drug effects , Flow Cytometry , Humans , In Vitro Techniques , Infant, Newborn , Intestinal Mucosa/cytology , Macrophages/drug effects , Macrophages/physiology , Mice , Toll-Like Receptor 4/genetics , rhoA GTP-Binding Protein/metabolism
7.
Hepatology ; 49(5): 1695-708, 2009 May.
Article in English | MEDLINE | ID: mdl-19296467

ABSTRACT

UNLABELLED: The liver is the main organ that clears lipopolysaccharide (LPS) and hepatocytes are a major cell-type involved in LPS uptake. LPS tolerance, or desensitization, is important in negative regulation of responses to LPS, but little is known about its mechanisms in hepatocytes. Primary isolated C57BL/6 hepatocytes, and liver in vivo, internalized fluorescent LPS, and this was dependent on Toll-like receptor 4 (TLR4) at the cell surface but not on TLR4-TIR signaling through MyD88. LPS clearance from plasma was also TLR4-dependent. Pretreatment of C57BL/6 hepatocytes with LPS prevented uptake of LPS 24 hours later and this LPS-mediated suppression was dependent on TLR4 signaling through MyD88. Many regulators of TLR4 signaling have been identified and implicated in LPS desensitization, including suppressor of cytokine signaling 1 (SOCS1). SOCS1 mRNA and protein expression increased after LPS stimulation in hepatocytes and in whole liver. LPS uptake in hepatocytes and liver was significantly reduced following infection with adenoviral vectors overexpressing SOCS1. Similarly, inhibition of SOCS1 using small interfering (si)RNA-mediated knockdown prevented LPS desensitization in hepatocytes. SOCS1 is known to interact with Toll/IL-1 receptor associated protein (TIRAP) and cause TIRAP ubiquitination and degradation, which regulates TLR signaling. We have also shown previously that TIRAP regulates LPS uptake in hepatocytes. SOCS1 coimmunoprecipitated with TIRAP in wild type hepatocyte cell lysates up to 8 hours after LPS stimulation, but not at later times. In the same samples, ubiquitinated TIRAP was detected after 4 hours and up to 8 hours after LPS stimulation, but not at later times. CONCLUSION: These data indicate hepatocytes are desensitized by LPS in a TLR4 signaling-dependent manner. LPS-induced SOCS1 upregulation increases degradation of TIRAP and prevents subsequent LPS uptake. The exploitation of these mechanisms of LPS desensitization in the liver may be important in future sepsis therapies.


Subject(s)
Desensitization, Immunologic , Hepatocytes/immunology , Lipopolysaccharides/immunology , Liver/immunology , Suppressor of Cytokine Signaling Proteins/metabolism , Adaptor Proteins, Vesicular Transport/metabolism , Animals , Hepatocytes/metabolism , Lipopolysaccharides/metabolism , Liver/metabolism , Lymphocyte Antigen 96/metabolism , Membrane Glycoproteins/metabolism , Mice , Mice, Inbred C3H , Mice, Inbred C57BL , Mice, Knockout , Myeloid Differentiation Factor 88/metabolism , Receptors, Interleukin-1/metabolism , Suppressor of Cytokine Signaling 1 Protein , Toll-Like Receptor 4/metabolism , Up-Regulation
8.
J Immunol ; 181(7): 5015-23, 2008 Oct 01.
Article in English | MEDLINE | ID: mdl-18802105

ABSTRACT

The chromatin-binding factor high-mobility group box 1 (HMGB1) functions as a proinflammatory cytokine and late mediator of mortality in murine endotoxemia. Although serine phosphorylation of HMGB1 is necessary for nucleocytoplasmic shuttling before its cellular release, the protein kinases involved have not been identified. To investigate if calcium/calmodulin-dependent protein kinase (CaMK) IV serine phosphorylates and mediates the release of HMGB1 from macrophages (Mphi) stimulated with LPS, RAW 264.7 cells or murine primary peritoneal Mphi were incubated with either STO609 (a CaMKIV kinase inhibitor), KN93 (a CaMKIV inhibitor), or we utilized cells from which CaMKIV was depleted by RNA interference (RNAi) before stimulation with LPS. We also compared the LPS response of primary Mphi isolated from CaMKIV(+/+) and CaMKIV(-/-) mice. In both cell types LPS induced activation and nuclear translocation of CaMKIV, which preceded HMGB1 nucleocytoplasmic shuttling. However, Mphi treated with KN93, STO609, or CaMKIV RNAi before LPS showed reduced nucleocytoplasmic shuttling of HMGB1 and release of HMGB1 into the supernatant. Additionally, LPS induced serine phosphorylation of HMGB1, which correlated with an interaction between CaMKIV and HMGB1 and with CaMKIV phosphorylation of HMGB1 in vitro. In cells, both HMGB1 phosphorylation and interaction with CaMKIV were inhibited by STO609 or CaMKIV RNAi. Similarly, whereas CaMKIV(+/+) Mphi showed serine phosphorylation of HMGB1 in response to LPS, this phosphorylation was attenuated in CaMKIV(-/-) Mphi. Collectively, our results demonstrate that CaMKIV promotes the nucleocytoplasmic shuttling of HMGB1 and suggest that the process may be mediated through CaMKIV-dependent serine phosphorylation of HMGB1.


Subject(s)
Calcium-Calmodulin-Dependent Protein Kinase Type 4/physiology , Cell Nucleus/metabolism , Cytoplasm/metabolism , HMGB1 Protein/metabolism , Lipopolysaccharides/pharmacology , Macrophages, Peritoneal/enzymology , Active Transport, Cell Nucleus/immunology , Animals , Cell Line , Cells, Cultured , Cytoplasm/immunology , Macrophages, Peritoneal/immunology , Macrophages, Peritoneal/metabolism , Mice , Nuclear Proteins/metabolism , Phosphorylation , Serine/metabolism
9.
Am J Physiol Cell Physiol ; 292(2): C824-31, 2007 Feb.
Article in English | MEDLINE | ID: mdl-16914531

ABSTRACT

Nitric oxide (NO) acts as a vasoregulatory molecule that inhibits vascular smooth muscle cell (SMC) proliferation. Studies have illustrated that NO inhibits SMC proliferation via the extracellular signal-regulated kinase (ERK) pathway, leading to increased protein levels of the cyclin-dependent kinase inhibitor p21(Waf1/Cip1). The ERK pathway can be pro- or antiproliferative, and it has been demonstrated that the activation status of the small GTPase RhoA determines the proliferative fate of ERK signaling, whereby inactivation of RhoA influences ERK signaling to increase p21(Waf1/Cip1) and inhibit proliferation. The purpose of these investigations was to examine the effect of NO on RhoA activation/S-nitrosation and to test the hypothesis that inhibition of SMC proliferation by NO is dependent on inactivation of RhoA. NO decreases activation of RhoA, as demonstrated by RhoA GTP-binding assays, affinity precipitation, and phalloidin staining of the actin cytoskeleton. Additionally, these effects are independent of cGMP. NO decreases SMC proliferation, and gene transfer of constitutively active RhoA (RhoA(63L)) diminished the antiproliferative effects of NO, as determined by thymidine incorporation. Western blots of p21(Waf1/Cip1) correlated with changes in proliferation. S-nitrosation of recombinant RhoA protein and immunoprecipitated RhoA was demonstrated by Western blotting for nitrosocysteine and by measurement of NO release. Furthermore, NO decreases GTP loading of recombinant RhoA protein. These findings indicate that inactivation of RhoA plays a role in NO-mediated SMC antiproliferation and that S-nitrosation is associated with decreased GTP binding of RhoA. Nitrosation of RhoA and other proteins likely contributes to cGMP-independent effects of NO.


Subject(s)
Cell Proliferation , Myocytes, Smooth Muscle/physiology , Nitric Oxide/metabolism , rhoA GTP-Binding Protein/metabolism , Animals , Aorta, Thoracic/cytology , Cells, Cultured , Cyclic GMP/metabolism , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Extracellular Signal-Regulated MAP Kinases/metabolism , Nitric Oxide/pharmacology , Nitrosation , Phosphorylation , Rats , Rats, Sprague-Dawley , Signal Transduction
10.
J Biol Chem ; 279(43): 44327-34, 2004 Oct 22.
Article in English | MEDLINE | ID: mdl-15280387

ABSTRACT

A properly functioning immune system is dependent on programmed cell death/apoptosis at virtually every stage of lymphocyte development and activity. Carbon monoxide (CO), an enzymatic product of heme oxyenase-1, has been shown to possess anti-apoptotic effects in a number of different model systems. The purpose of the present study was to expand on this knowledge to determine the role of CO in the well established model of Fas/CD95-induced apoptosis in Jurkat cells, and to determine the mechanism by which CO can modulate T-cell apoptosis. Exposure of Jurkat cells to CO resulted in augmentation in Fas/CD95-induced apoptosis, which correlated with CO-induced up-regulation of the pro-apoptotic protein FADD as well as activation of caspase-8, -9, and -3 while simultaneously down-regulating the anti-apoptotic protein BCL-2. These effects of CO were lost with overexpression of the small interfering RNA of FADD. CO, as demonstrated previously in endothelial cells, was also anti-apoptotic in Jurkat cells against tumor necrosis factor and etoposide. We further demonstrate that this pro-apoptotic effect of CO was independent of reactive oxygen species production and involved inhibition in Fas/CD95-induced activation of the pro-survival ERK MAPK. We conclude that in contrast to other studies showing the anti-apoptotic effects of CO, Fas/CD95-induced cell death in Jurkat cells is augmented by exposure to CO and that this occurs in part via inhibition in the activation of ERK MAPK. These data begin to elucidate specific differences with regard to the effects of CO and cell death pathways and provide important and valuable insight into potential mechanisms of action.


Subject(s)
Apoptosis , Carbon Monoxide/metabolism , fas Receptor/biosynthesis , Adenoviridae/genetics , Annexin A5/pharmacology , Blotting, Western , Carbon Monoxide/chemistry , Caspase 3 , Caspase 8 , Caspase 9 , Caspases/metabolism , Cell Death , Cell Line, Tumor , Down-Regulation , Enzyme Inhibitors/pharmacology , Flow Cytometry , HSP70 Heat-Shock Proteins/metabolism , Humans , Inhibitor of Apoptosis Proteins , Jurkat Cells , MAP Kinase Signaling System , Propidium/pharmacology , Proteins/metabolism , Proto-Oncogene Proteins c-bcl-2/metabolism , RNA/metabolism , RNA, Small Interfering/metabolism , Time Factors , Transfection , Up-Regulation , fas Receptor/chemistry
11.
Methods Mol Biol ; 279: 225-34, 2004.
Article in English | MEDLINE | ID: mdl-15199249

ABSTRACT

The varied biological effects of nitric oxide (NO) have led to intense research into its diverse physiologic and pathophysiologic roles in multiple disease processes. It has been implicated in the development of altered vasomotor tone, intimal hyperplasia, atherosclerosis, impotence, host defense, and wound healing. Using the modern technologies of recombinant DNA and gene transfer using adenoviral vectors, the effects of NO derived from various NO synthase (NOS) enzymes can be studied in a variety of tissues and the therapeutic applications of NOS is possible. Such uses of NOS gene transfer have been investigated extensively in the vasculature where NO is critical to regulating vascular homeostasis. NOS gene therapy has the theoretical advantage of allowing NO delivery to be localized, thereby limiting potential adverse effects of NO. The benefits of adenoviral vectors in gene transfer include relatively high transduction efficiencies, both replicating and nonreplicating cells may be infected, and the high titers of adenovirus that can be produced. The methods described in this chapter include the cloning of the iNOS cDNA into a recombinant adenoviral vector, large-scale production of that vector AdiNOS preparation, and the use of the vector to transduce tissue in vitro and in vivo.


Subject(s)
Adenoviridae/genetics , Aorta/metabolism , Myocytes, Smooth Muscle/metabolism , Nitric Oxide Synthase/genetics , Animals , Cloning, Molecular , Gene Transfer Techniques , Mice , Nitric Oxide/metabolism , Nitric Oxide Synthase/metabolism , Nitric Oxide Synthase Type II , Rats , Transplantation
12.
Circulation ; 108(7): 876-81, 2003 Aug 19.
Article in English | MEDLINE | ID: mdl-12874183

ABSTRACT

BACKGROUND: The 42/44-kD mitogen-activated protein kinases (extracellular signal-regulated kinases, ERKs) regulate smooth muscle cell (SMC) cell-cycle progression and can either promote or inhibit proliferation depending on the activation status of the small GTPase RhoA. RhoA is involved in the regulation of the actin cytoskeleton and converges on multiple signaling pathways. However, the mechanism by which RhoA modulates ERK signaling is not well defined. The purpose of this investigation was to examine whether RhoA regulates ERK downstream signaling and cellular proliferation through its effects on the cytoskeleton and the nuclear localization of ERK. METHODS AND RESULTS: Treatment of SMCs with Clostridia botulinum C3 exoenzyme, which inhibits RhoA activation, decreased SMC proliferation to 24+/-7% of that of controls and increased p21Waf1/Cip1 transcription and protein levels. These effects of RhoA were reversed by inhibition of ERK phosphorylation. However, inactivation of RhoA did not alter levels of ERK phosphorylation but did increase nuclear localization of phosphorylated ERK. In addition, immunostaining demonstrated that phosphorylated ERK associated with the actin cytoskeleton, which was disrupted by C3 exoenzyme. Leptomycin B, an inhibitor of Crm1 that results in ERK nuclear accumulation, similarly increased p21Waf1/Cip1. CONCLUSIONS: RhoA inhibition increased levels of phosphorylated ERK in the cell nucleus. Inhibition of RhoA or pharmacological inhibition of nuclear export resulted in increased p21Waf1/Cip1 expression and decreased SMC proliferation, effects that were partially dependent on ERK. RhoA regulation of the actin cytoskeleton may determine ERK subcellular localization and its subsequent effects on SMC proliferation.


Subject(s)
Cell Nucleus/metabolism , Cyclins/metabolism , Mitogen-Activated Protein Kinases/metabolism , Muscle, Smooth, Vascular/metabolism , Receptors, Cytoplasmic and Nuclear , rhoA GTP-Binding Protein/metabolism , ADP Ribose Transferases/pharmacology , Active Transport, Cell Nucleus/drug effects , Active Transport, Cell Nucleus/physiology , Animals , Botulinum Toxins/pharmacology , Cell Division/drug effects , Cell Division/physiology , Cells, Cultured , Cyclin-Dependent Kinase Inhibitor p21 , Cyclin-Dependent Kinases/antagonists & inhibitors , Cyclins/genetics , Cytoskeleton/metabolism , Enzyme Inhibitors/pharmacology , Fatty Acids, Unsaturated/pharmacology , Genes, Reporter , Karyopherins/antagonists & inhibitors , Muscle, Smooth, Vascular/cytology , Phosphorylation/drug effects , Rats , Rats, Sprague-Dawley , Signal Transduction/physiology , rhoA GTP-Binding Protein/antagonists & inhibitors , Exportin 1 Protein
13.
Infect Immun ; 70(7): 3433-42, 2002 Jul.
Article in English | MEDLINE | ID: mdl-12065483

ABSTRACT

The liver is an important site of host-microbe interaction. Although hepatocytes have been reported to be responsive to lipopolysaccharide (LPS), the global gene expression changes by LPS and mechanism(s) by which LPS stimulates cultured hepatocytes remain uncertain. Cultures of primary mouse hepatocytes were incubated with LPS to assess its effects on the global gene expression, hepatic transcription factors, and mitogen-activated protein (MAP) kinase activation. DNA microarray analysis indicated that LPS modulates the selective expression of more than 80 genes and expressed sequence tags. We have shown previously that hepatocytes express CD14, which is required both for uptake and responsiveness to LPS. In other cells, responsiveness to microbial products requires expression of Toll-like receptors (TLR) and their associated accessory molecules. Hepatocytes expressed TLR1 through TLR9 as well as MyD88 and MD-2 transcripts, as shown by reverse transcriptase PCR analysis, indicating that hepatocytes express all known microbe recognition molecules. The MAP kinase extracellular signal-regulated kinase 1/2 was phosphorylated in response to LPS in mouse hepatocytes, and the levels of phosphorylation were lower in hepatocytes from TLR4-null mice. NF-kappa B activation was reduced in TLR4-mutant or -null hepatocytes compared to control hepatocytes, and this defect was partially restored by adenoviral transduction of mouse TLR4. Thus, hepatocytes respond to nanogram concentrations of LPS through a TLR4 response pathway.


Subject(s)
Drosophila Proteins , Gene Expression , Lipopolysaccharides/pharmacology , Membrane Glycoproteins/physiology , NF-kappa B/metabolism , Receptors, Cell Surface/physiology , Signal Transduction , Adaptor Proteins, Signal Transducing , Adenoviridae , Animals , Antigens, Differentiation/genetics , Antigens, Surface/genetics , Cells, Cultured , Genetic Vectors , Hepatocytes/cytology , Hepatocytes/drug effects , Humans , Lymphocyte Antigen 96 , Membrane Glycoproteins/genetics , Mice , Mice, Inbred C3H , Mice, Inbred C57BL , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3 , Mitogen-Activated Protein Kinases/metabolism , Myeloid Differentiation Factor 88 , Oligonucleotide Array Sequence Analysis , Receptors, Cell Surface/genetics , Receptors, Immunologic/genetics , Toll-Like Receptor 1 , Toll-Like Receptor 4 , Toll-Like Receptor 9 , Toll-Like Receptors , Transcription Factor AP-1/metabolism , Tumor Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL
...