Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
1.
Int J Mol Sci ; 23(21)2022 Oct 27.
Article in English | MEDLINE | ID: mdl-36361794

ABSTRACT

An emerging therapeutic approach in the treatment of infectious disease is to augment the host response through repurposing of well-tolerated, non-antibiotic, host-directed therapeutics. Earlier retrospective studies identify a positive association between statin use and a decreased risk of death due to sepsis or bacteremia. However, more recent randomized control trials fail to detect a therapeutic benefit in these complex infection settings. It is postulated that unrecognized biases in certain observational studies may have led to an overestimation of benefit and that statin use is instead a marker for health status, wealth, and demographic characteristics which may separately affect death due to infection. What remains unresolved is that in vitro and in vivo evidence reproducibly indicates that statin pharmacology limits infection and augments immunomodulatory responses, suggesting that therapeutic benefits may be attainable in certain infection settings, such as intracellular infection by S. aureus. Carefully considering the biological mechanisms capable of driving the relationship between statins and infections and constructing a methodology to avoid potential biases in observational studies would enable the examination of protective effects against infection and limit the risk of underestimating statin efficacy. Such an approach would rely on the examination of statin use in defined infection settings based on an underlying mode-of-action and pharmacology, where the inhibition of HMG-CoA-reductase at the rate-limiting step in cholesterol biosynthesis diminishes not only cholesterol levels but also isoprenoid intermediates central to host cell invasion by S. aureus. Therapeutic benefit in such settings, if existent, may be of clinical importance.


Subject(s)
Hydroxymethylglutaryl-CoA Reductase Inhibitors , Staphylococcal Infections , Humans , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology , Hydroxymethylglutaryl-CoA Reductase Inhibitors/therapeutic use , Staphylococcus aureus , Retrospective Studies , Staphylococcal Infections/drug therapy , Cholesterol
2.
Biochem Biophys Res Commun ; 432(4): 695-700, 2013 Mar 22.
Article in English | MEDLINE | ID: mdl-23410754

ABSTRACT

During severe sepsis, microvesicles that are positive for tissue factor (TF) are at increased levels within blood and in pulmonary lavage. These microvesicles potentially disperse TF, the major initiator of the coagulation cascade, throughout multiple organ systems, initiating fibrin deposition and resultant ischemia. The source of these microvesicles has remained incompletely defined. Although TF(+) microvesicles are shed from cells that express nascent TF transcript in response to injury, recent findings revealed that circulating, full-length TF protein is detectable prior to these nascent transcripts. This finding suggested that the protein is released from constitutive sources as an acute response. We examined whether Staphylococcus aureus, the Gram-positive bacteria that is emerging as one of the most common etiologic agents in sepsis, is capable of stimulating the release of TF(+) microvesicles from a pulmonary cell line that constitutively expresses TF protein. We found that host cell invasion stimulated an acute release of TF(+) microvesicles and that these microvesicles mediated the transfer of the protein to TF-negative endothelial cells. We also found that transfer was inhibited by cholesterol-lowering simvastatin. Taken together, our findings reveal that S. aureus pathogenesis extends to the acute release of TF(+) microvesicles and that inhibiting dispersal by this mechanism may provide a therapeutic target.


Subject(s)
Cytoplasmic Vesicles/metabolism , Host-Pathogen Interactions , Lung/microbiology , Sepsis/microbiology , Staphylococcal Infections/microbiology , Staphylococcus aureus/pathogenicity , Thromboplastin/metabolism , Cell Line , Cytoplasmic Vesicles/drug effects , Humans , Lung/metabolism , Protein Transport/drug effects , Sepsis/metabolism , Simvastatin/pharmacology , Staphylococcal Infections/metabolism
3.
Pharmaceutics ; 13(12)2021 Nov 30.
Article in English | MEDLINE | ID: mdl-34959329

ABSTRACT

An emergent approach to bacterial infection is the use of host rather than bacterial-directed strategies. This approach has the potential to improve efficacy in especially challenging infection settings, including chronic, recurrent infection due to intracellular pathogens. For nearly two decades, the pleiotropic effects of statin drugs have been examined for therapeutic usefulness beyond the treatment of hypercholesterolemia. Interest originated after retrospective studies reported decreases in the risk of death due to bacteremia or sepsis for those on a statin regimen. Although subsequent clinical trials have yielded mixed results and earlier findings have been questioned for biased study design, in vitro and in vivo studies have provided clear evidence of protective mechanisms that include immunomodulatory effects and the inhibition of host cell invasion. Ultimately, the benefits of statins in an infection setting appear to require attention to the underlying host response and to the timing of the dosage. From this examination of statin efficacy, additional novel host-directed strategies may produce adjunctive therapeutic approaches for the treatment of infection where traditional antimicrobial therapy continues to yield poor outcomes. This review focuses on the opportunistic pathogen, Staphylococcus aureus, as a proof of principle in examining the promise and limitations of statins in recalcitrant infection.

4.
Biochem Biophys Res Commun ; 391(1): 443-8, 2010 Jan 01.
Article in English | MEDLINE | ID: mdl-19914208

ABSTRACT

Beta1-containing adhesions at the plasma membrane function as dynamic complexes to provide bidirectional communication between the cell and its environment, yet commonly are used by pathogens to gain host cell entry. Recently, the cholesterol-lowering drug simvastatin was found to inhibit host invasion through beta1-containing adhesion complexes. To better understand the regulatory mechanisms controlling adhesion formation and uptake and the use of these complexes by Staphylococcus aureus, the primary etiologic agent in sepsis, bacteremia and endocarditis, we investigated the mechanism of inhibition by simvastatin. In response to simvastatin, adhesion complexes diminished as well as beta1 trafficking to the plasma membrane required to initiate adhesion formation. Simvastatin stimulated CDC42 activation and coupling to p85, a small-guanosine triphosphatase (GTPase) activating protein (GAP), yet sequestered CDC42 coupled to p85 within the cytosol. Loss of p85 GAP activity through use of genetic strategies decreased host cell invasion as well as beta1 trafficking. From these findings, we propose a mechanism whereby p85 GAP activity localized within membrane compartments facilitates beta1 trafficking. By sequestering p85 within the cytosol, simvastatin restricts the availability and uptake of the receptor used by pathogenic strains to gain host cell entry.


Subject(s)
Host-Pathogen Interactions/drug effects , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology , Integrin beta1/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Simvastatin/pharmacology , Staphylococcal Infections/enzymology , Staphylococcus aureus/pathogenicity , Animals , Bacterial Adhesion/drug effects , Cell Membrane/enzymology , Cytosol/enzymology , Humans , Mice , Staphylococcal Infections/microbiology , Swiss 3T3 Cells , cdc42 GTP-Binding Protein/metabolism
5.
Curr Pharm Biotechnol ; 20(9): 733-744, 2019.
Article in English | MEDLINE | ID: mdl-31258074

ABSTRACT

BACKGROUND: Recurrent pharyngotonsillitis due to Streptococcus pyogenes develops regardless of whether infecting strains are resistant or susceptible to first-line antimicrobials. Causation for recurrent infection is associated with the use of first-line antimicrobials that fail to penetrate deep tissue and host cell membranes, enabling intracellular S. pyogenes to survive throughout repeated rounds of antimicrobial therapy. OBJECTIVE: To determine whether simvastatin, a therapeutic approved for use in the treatment of hypercholesterolemia, and ML141, a first-in-class small molecule inhibitor with specificity for human CDC42, limit host cell invasion by S. pyogenes. METHODS: Assays to assess host cell invasion, bactericidal activity, host cell viability, actin depolymerization, and fibronectin binding were performed using the RAW 267.4 macrophage cell line and Human Umbilical Vein Endothelial Cells (HUVEC) infected with S. pyogenes (90-226) and treated with simvastatin, ML141, structural analogs of ML141, or vehicle control. RESULTS: Simvastatin and ML141 decreased intracellular infection by S. pyogenes in a dose-dependent manner. Inhibition by simvastatin persisted following 1 h washout whereas inhibition by ML141 was reversed. During S. pyogenes infection, actin stress fibers depolymerized in vehicle control treated cells, yet remained intact in simvastatin and in ML141 treated cells. Consistent with the previous characterization of ML141, simvastatin decreased host cell binding to fibronectin. Structural analogs of ML141, designated as the RSM series, decreased intracellular infection through non-cytotoxic, nonbactericidal mechanisms. CONCLUSION: Our findings demonstrate the potential of repurposing simvastatin and of developing CDC42-targeted therapeutics for eradicating intracellular S. pyogenes infection to break the cycle of recurrent infection through a host-directed approach.


Subject(s)
Anti-Bacterial Agents/pharmacology , Pyrazoles/pharmacology , Simvastatin/pharmacology , Streptococcal Infections/drug therapy , Streptococcus pyogenes/drug effects , Sulfonamides/pharmacology , cdc42 GTP-Binding Protein/antagonists & inhibitors , Animals , Anti-Bacterial Agents/chemistry , Cells, Cultured , Fibronectins/metabolism , Human Umbilical Vein Endothelial Cells/drug effects , Human Umbilical Vein Endothelial Cells/microbiology , Humans , Macrophages/metabolism , Macrophages/microbiology , Mice , Molecular Structure , Pyrazoles/chemistry , RAW 264.7 Cells , Simvastatin/chemistry , Sulfonamides/chemistry
6.
J Pharmacol Exp Ther ; 326(1): 135-43, 2008 Jul.
Article in English | MEDLINE | ID: mdl-18388257

ABSTRACT

Patients on a statin regimen have a decreased risk of death due to bacterial sepsis. We have found that protection by simvastatin includes the inhibition of host cell invasion by Staphylococcus aureus, the most common etiologic agent of sepsis. Inhibition was due in part to depletion of isoprenoid intermediates within the cholesterol biosynthesis pathway and led to the cytosolic accumulation of the small GTPases CDC42, Rac, and RhoB. Actin stress fiber disassembly required for host invasion was attenuated by simvastatin and by the inhibition of phosphoinositide 3-kinase (PI3K) activity. PI3K relies on coupling to prenylated proteins, such as this subset of small GTPases, for access to membrane-bound phosphoinositide to mediate stress fiber disassembly. Therefore, we examined whether simvastatin restricts PI3K cellular localization. In response to simvastatin, the PI3K isoform p85, coupled to these small-GTPases, was sequestered within the cytosol. From these findings, we propose a mechanism whereby simvastatin restricts p85 localization, inhibiting the actin dynamics required for bacterial endocytosis. This approach may provide the basis for protection at the level of the host in invasive infections by S. aureus.


Subject(s)
Simvastatin/pharmacology , Staphylococcus aureus/drug effects , Terpenes/metabolism , Cell Line , Cells, Cultured , Humans , Integration Host Factors/antagonists & inhibitors , Phosphatidylinositol 3-Kinases/metabolism , Phosphoinositide-3 Kinase Inhibitors , Staphylococcus aureus/cytology
7.
Curr Pharm Biotechnol ; 17(10): 886-93, 2016.
Article in English | MEDLINE | ID: mdl-26927218

ABSTRACT

Statins are potent modulators of immune responses, resulting in their ability to enhance host survival from primary bacterial infections. Alterations in primary immune responses that may be beneficial for survival following infection may also result in alterations in the generation of the immunologic memory response and subsequently affect immune responses mounted during secondary bacterial infection. In this study, we report that levels of total serum IgG2c, following primary infection, were decreased in simvastatin pretreated mice, and investigate the effect of simvastatin treatment, prior to primary infection, on immune responses activated during secondary S. aureus infection. A secondary infection model was implemented whereby simvastatin pretreated and control mice were reinfected with S. aureus 14 days after primary infection, with no additional simvastatin treatment, and assessed for survival and alterations in immune function. While survivability to secondary S. aureus infection was not different between simvastatin pretreated and control mice, memory B and T lymphocyte functions were altered. Memory B cells, isolated 14 days after secondary infection, from simvastatin pretreated mice and stimulated ex vivo produced increased levels of IgG1 compared to memory B cells isolated from control mice, while levels of IgM and IgG2c remained similar. Furthermore, memory B and T lymphocytes from simvastatin pretreated mice exhibited a decreased proliferative response when stimulated ex vivo compared to memory cells isolated from control mice. These findings demonstrate the ability of a short term, low dose simvastatin treatment to modulate memory immune function.


Subject(s)
Immunologic Memory/drug effects , Simvastatin/pharmacology , Staphylococcal Infections/immunology , Staphylococcus aureus , Animals , B-Lymphocytes/immunology , Dose-Response Relationship, Drug , Mice , Receptors, IgG/immunology , Simvastatin/administration & dosage , T-Lymphocytes/immunology
8.
Chest ; 121(3 Suppl): 70S-75S, 2002 Mar.
Article in English | MEDLINE | ID: mdl-11893692

ABSTRACT

Initiated by numerous factors, acute lung injury is marked by epithelial and endothelial cell perturbation and inflammatory cell influx that leads to surfactant disruption, pulmonary edema, and atelectasis. This syndrome has been associated with a myriad of mediators including cytokines, oxidants, and growth factors. To better understand gene-environmental interactions controlling this complex process, the sensitivity of inbred mouse strains was investigated following acute lung injury that was induced by fine nickel sulfate aerosol. Measuring survival time, protein and neutrophil concentrations in BAL fluid, lung wet-to-dry weight ratio, and histology, we found that these responses varied between inbred mouse strains and that susceptibility is heritable. To assess the progression of acute lung injury, the temporal expression of genes and expressed sequence tags was assessed by complementary DNA microarray analysis. Enhanced expression was noted in genes that were associated with oxidative stress, antiprotease function, and extracellular matrix repair. In contrast, expression levels of surfactant proteins (SPs) and Clara cell secretory protein (ie, transcripts that are constitutively expressed in the lung) decreased markedly. Genome-wide analysis was performed with offspring derived from a sensitive and resistant strain (C57BL/6xA F(1) backcrossed with susceptible A strain). Significant linkage was identified for a locus on chromosome 6 (proposed as Aliq4), a region that we had identified previously following ozone-induced acute lung injury. Two suggestive linkages were identified on chromosomes 1 and 12. Using haplotype analysis to estimate the combined effect of these regions (along with putative modifying loci on chromosomes 9 and 16), we found that five loci interact to account for the differences in survival time of the parental strains. Candidate genes contained in Aliq4 include SP-B, aquaporin 1, and transforming growth factor-alpha. Thus, the functional genomic approaches of large gene set expression (complementary DNA microarray) and genome-wide analyses continue to provide novel insights into the genetic susceptibility of lung injury.


Subject(s)
Chromosome Mapping , Genetic Predisposition to Disease , Respiratory Distress Syndrome/genetics , Aerosols , Animals , Aquaporin 1 , Aquaporins/genetics , Environmental Exposure , Gene Expression , Genetic Linkage , Lung/pathology , Mice , Mice, Inbred Strains , Mice, Transgenic , Nickel , Oligonucleotide Array Sequence Analysis , Ozone , Polytetrafluoroethylene , Proteolipids/genetics , Pulmonary Surfactants/genetics , Quantitative Trait, Heritable , Respiratory Distress Syndrome/etiology , Respiratory Distress Syndrome/pathology , Risk Factors , Transforming Growth Factor alpha/genetics , Transforming Growth Factor alpha/physiology
9.
Chemosphere ; 51(10): 1139-48, 2003 Jun.
Article in English | MEDLINE | ID: mdl-12718980

ABSTRACT

Human exposure to insoluble and soluble nickel compounds is extensive. Besides wide usage in many industries, nickel compounds are contained in cigarette smoke and, in low levels, in ambient particulate matter. Soluble nickel particulate, especially nickel sulfate (NiSO(4)), has been associated with acute lung injury. To begin identifying genes controlling susceptibility to NiSO(4), mean survival times (MSTs) of eight inbred mouse strains were determined after aerosol exposure. Whereas A/J (A) mice were sensitive, C57BL/6J (B6) mice survived nearly twice as long (resistant). Their offspring were similarly resistant, demonstrating heritability as a dominant trait. Quantitative trait locus (QTL) analysis of backcross mice generated from these strains identified a region on chromosome 6 significantly linked to survival time. Regions on chromosomes 1 and 12 were suggestive of linkage and regions on chromosomes 8, 9, and 16 contributed to the response. Haplotype analysis demonstrated that QTLs on chromosomes 6, 9, 12, and 16 could explain the MST difference between the parental strains. To complement QTL analysis results, cDNA microarray analysis was assessed following NiSO(4) exposure of A and B6 mice. Significant expression changes were identified in one or both strains for >100 known genes. Closer evaluation of these changes revealed a temporal pattern of increased cell proliferation, extracellular matrix repair, hypoxia, and oxidative stress, followed by diminished surfactant proteins. Certain expressed sequence tags clustered with known genes, suggesting possible co-regulation and novel roles in pulmonary injury. Together, results from QTL and microarray analyses of nickel-induced acute lung injury survival allowed us to generate a short list of candidate genes.


Subject(s)
DNA Damage , Gene Expression Profiling , Genetic Predisposition to Disease , Irritants/adverse effects , Nickel/adverse effects , Adaptation, Physiological , Animals , Female , Haplotypes , Male , Mice , Mice, Inbred Strains , Oligonucleotide Array Sequence Analysis , Oxidative Stress , Solubility , Survival
10.
Curr Pharm Biotechnol ; 15(8): 727-37, 2014.
Article in English | MEDLINE | ID: mdl-25213310

ABSTRACT

Staphylococcus aureus is a leading causative agent in sepsis, endocarditis, and pneumonia. An emerging concept is that prognosis worsens when the infecting S. aureus strain has the capacity to not only colonize tissue as an extracellular pathogen, but to invade host cells and establish intracellular bacterial populations. In previous work, we identified host CDC42 as a central regulator of endothelial cell invasion by S. aureus. In the current work, we report that ML 141, a first-in-class CDC42 inhibitor, decreases invasion and resultant pathogenesis in a dose-dependent and reversible manner. Inhibition was found to be due in part to decreased remodeling of actin that potentially drives endocytic uptake of bacteria/fibronectin/integrin complexes. ML 141 decreased binding to fibronectin at these complexes, thereby limiting a key pathogenic mechanism used by S. aureus to invade. Structural analogs of ML 141 were synthesized (designated as the RSM series) and a subset identified that inhibit invasion through non-cytotoxic and non-bactericidal mechanisms. Our results support the development of adjunctive therapeutics targeting host CDC42 for mitigating invasive infection at the level of the host.


Subject(s)
Small Molecule Libraries/pharmacology , Staphylococcus aureus/drug effects , Bacterial Adhesion , Cells, Cultured , Endothelial Cells/drug effects , Endothelial Cells/microbiology , Humans , Myotonin-Protein Kinase/antagonists & inhibitors
11.
Curr Pharm Biotechnol ; 14(2): 233-41, 2013.
Article in English | MEDLINE | ID: mdl-23228241

ABSTRACT

Staphylococcus aureus is the most prevalent etiologic agent of sepsis. Statins, primarily prescribed for their cholesterol-lowering capabilities, may be beneficial for treating sepsis due to their anti-inflammatory properties. This study examined the effect of low dose, short term simvastatin pretreatment in conjunction with antibiotic treatment on host survival and demonstrated that pretreatment with simvastatin increased survival of C57BL/6 mice in response to S. aureus infection. In vitro studies revealed that short term simvastatin pretreatment did not reduce S. aureus-stimulated expression of surface proteins necessary for macrophage presentation of antigen to T cells, such as MHC Class II and costimulatory molecules CD80 and CD86, but did reduce both basal and S. aureus-stimulated levels of C5aR. Additionally, this work demonstrated the ability of simvastatin to dampen macrophage responses initiated not only by bacteria directly but by membrane vesicles shed in response to infection, revealing a new mechanism of immune modulation by statins. These data demonstrate the ability of short term simvastatin pretreatment to modulate immune responses and identify new insights into the underlying mechanisms of the anti-inflammatory properties of simvastatin that may decrease the pathophysiological effects leading to sepsis.


Subject(s)
Anti-Inflammatory Agents/therapeutic use , Hydroxymethylglutaryl-CoA Reductase Inhibitors/therapeutic use , Sepsis/drug therapy , Simvastatin/therapeutic use , Staphylococcal Infections/drug therapy , Animals , Cell Line , Female , Human Umbilical Vein Endothelial Cells , Humans , Macrophage Activation/drug effects , Macrophages/drug effects , Macrophages/immunology , Male , Mice , Mice, Inbred C57BL , Sepsis/immunology , Sepsis/microbiology , Staphylococcal Infections/immunology , Staphylococcal Infections/microbiology , Staphylococcus aureus
12.
J Food Prot ; 76(4): 674-9, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23575132

ABSTRACT

Quality control procedures during food processing may involve direct inoculation of food samples onto appropriate selective media for subsequent enumeration. However, sublethally injured bacteria often fail to grow, enabling them to evade detection and intervention measures and ultimately threaten the health of consumers. This study compares traditional selective and nonselective agar-based overlays versus two commercial systems (Petrifilm and Easygel) for recovery of injured E. coli B-41560 and O157:H7 strains. Bacteria were propagated in tryptic soy broth (TSB), ground beef slurry, and infant milk formula to a density of 10(6) to 10(8) CFU/ml and then were stressed for 6 min either in lactic acid (pH 4.5) or heat shocked for 3 min at 60°C. Samples were pour plated in basal layers of either tryptic soy agar (TSA), sorbitol MacConkey agar (SMAC), or violet red bile agar (VRB) and were resuscitated for 4 h prior to addition of agar overlays. Other stressed bacteria were plated directly onto Petrifilm and Easygel. Results indicate that selective and nonselective agar overlays recovered significantly higher numbers (greater than 1 log) of acid- and heat-injured E. coli O157:H7 from TSB, ground beef, and infant milk formula compared with direct plating onto selective media, Petrifilm, or Easygel, while no significant differences among these media combinations were observed for stressed E. coli B-41560. Nonstressed bacteria from TSB and ground beef were also recovered at densities significantly higher in nonselective TSA-TSA and in VRB-VRB and SMAC-SMAC compared with Petrifilm and Easygel. These data underscore the need to implement food safety measures that address sublethally injured pathogens such as E. coli O157:H7 in order to avoid underestimation of true densities for target pathogens.


Subject(s)
Agar/chemistry , Colony Count, Microbial/methods , Escherichia coli O157/growth & development , Escherichia coli/growth & development , Food Contamination/analysis , Food Handling/methods , Consumer Product Safety , Humans , Hydrogen-Ion Concentration , Infant , Infant Food/microbiology , Kinetics , Meat Products/microbiology , Temperature
14.
Curr Pharm Biotechnol ; 12(9): 1455-62, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21401521

ABSTRACT

Epidemiologic studies suggest that the incidence and severity of sepsis are ameliorated in patients on statins (3- hydroxy-3-methylglutaryl coenzyme A reductase inhibitors) for cholesterol lowering indications. We sought to understand the mechanism underlying such protection and hypothesized that simvastatin would be protective in mice against acute infection with Staphylococcus aureus, the primary etiologic agent in sepsis. Mice were treated with simvastatin or buffer for two weeks and were subsequently challenged with S. aureus intratracheally or intravenously. Relative to buffer-treated mice, bacterial killing was enhanced 4-fold (p=0.02), systemic dissemination was reduced, and lethality was decreased (hazard ratio 8.8, 95% CI 2.5 to 31.3, p=0.001) in mice that were pretreated with simvastatin for two weeks. Systemic inflammatory response was abrogated and the local elaboration of inflammatory mediators was diminished. Serum concentrations of pro-fibrinolytic protein C were elevated (p=0.034), while the concentration of pro-coagulant tissue factor in bronchoalveolar lavage fluids was attenuated (reduced 25%), p=0.001, in simvastatin-treated mice. Taken together, these data indicate that extended treatment with simvastatin is protective during infection with S. aureus through enhanced bacterial clearance, anti-inflammatory, and anti-coagulant activities. These studies provide insights into the mechanism by which statins confer protection in acute infection, support the notion that statins may be effective adjuncts in the treatment of sepsis, and provide a rationale for randomized control trials in patients that are at a high risk for infection characterized by coagulopathy.


Subject(s)
Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology , Pneumonia, Staphylococcal/prevention & control , Simvastatin/pharmacology , Animals , Bacterial Load , Bronchoalveolar Lavage Fluid/chemistry , Bronchoalveolar Lavage Fluid/cytology , C-Reactive Protein/metabolism , Cytokines/genetics , Disease Models, Animal , Gene Expression/drug effects , Mice , Peroxidase/metabolism , Pneumonia, Staphylococcal/metabolism , Pneumonia, Staphylococcal/microbiology , Pneumonia, Staphylococcal/pathology , Protein C/metabolism , Thromboplastin/metabolism
15.
Article in English | MEDLINE | ID: mdl-21401522
16.
Curr Pharm Biotechnol ; 12(9): 1440, 2011 Sep.
Article in English | MEDLINE | ID: mdl-22309021

Subject(s)
Hemostasis , Animals , Drug Design
17.
Am J Respir Cell Mol Biol ; 34(1): 15-27, 2006 Jan.
Article in English | MEDLINE | ID: mdl-16166746

ABSTRACT

Previous studies have shown that mice deficient in the tyrosine kinase domain (TK-/-) of the receptor Mst1r have an increased susceptibility to nickel (Ni)-induced acute lung injury (ALI). Mst1r TK-/- mice have decreased survival times, alterations in cytokine and nitric oxide regulation, and an earlier onset of pulmonary pathology compared with control mice, suggesting that Mst1r signaling, in part, may regulate the response to ALI. To examine the role of Mst1r in ALI in more detail, we compared the gene expression profiles of murine lung mRNA from control and Mst1r TK-/- mice at baseline and after 24 h of particulate Ni sulfate exposure. Microarray analyses showed a total of 343 transcripts that were significantly changed, either by Ni treatment, or between genotypes. Genes responsible for inflammation, edema, and lymphocyte function were altered in the Mst1r TK-/- mice. Interestingly, the genes for several granzymes were increased in Mst1r TK-/- mice before Ni exposure, compared with controls. In addition, the Mst1r TK-/- lungs showed clusters of cells near the vascular endothelium and airways. Immunohistochemistry indicates these clusters are composed of macrophages, T cells, and neutrophils, and that the clusters display granzyme protein production. These results suggest that Mst1r signaling may be involved in the regulation of macrophage and T-lymphocyte activation in vivo during injury. This assessment of gene expression indicates the importance of genetic factors in contributing to lung injury, and points to strategies for intervention in the progression of inflammatory diseases.


Subject(s)
Gene Expression Profiling , Irritants/toxicity , Nickel/toxicity , Receptor Protein-Tyrosine Kinases/physiology , Respiratory Distress Syndrome , Animals , Cluster Analysis , Lung/cytology , Lung/pathology , Lung/physiology , Macrophages/metabolism , Mice , Mice, Knockout , Molecular Sequence Data , Oligonucleotide Array Sequence Analysis , RNA, Messenger/metabolism , Receptor Protein-Tyrosine Kinases/genetics , Respiratory Distress Syndrome/chemically induced , Respiratory Distress Syndrome/immunology , Respiratory Distress Syndrome/physiopathology , Serine Endopeptidases/genetics , Serine Endopeptidases/metabolism , Signal Transduction/physiology , T-Lymphocytes/metabolism
18.
Am J Respir Cell Mol Biol ; 34(1): 73-82, 2006 Jan.
Article in English | MEDLINE | ID: mdl-16166738

ABSTRACT

Often fatal, acute lung injury has a complicated etiology. Previous studies from our laboratory in mice have demonstrated that survival during acute lung injury is a complex trait governed by multiple loci. We also found that the increase in metallothionein (MT) is one of the greatest noted in transcriptome-wide analyses of gene expression. To assess the role of MT in nickel-induced acute lung injury, the survival of Mt-transgenic, Mt1/2(+/+), and Mt1/2(-/-) mice was compared. Pulmonary inflammation and global gene expression were compared in Mt1/2(+/+) and Mt1/2(-/-) mice. Gene-targeted Mt1/2(-/-) mice were more susceptible than Mt1/2(+/+) mice to nickel-induced inflammation, surfactant-associated protein B transcript loss, and lethality. Similarly, Mt-transgenic mice exhibited increased survival. MAPPFinder analyses also noted significant decreases in genes involved in protein processing (e.g., ubiquitination, folding), which were greater in Mt1/2(-/-) mice as compared with Mt1/2(+/+) mice early in the progression of acute lung injury, possibly due to a zinc-mediated transcript destabilization. In contrast, transcript levels of genes associated with the inflammatory response, extracellular matrix regulation, and coagulation/fibrinolysis were increased more in Mt1/2(-/-) mice as compared with Mt1/2(+/+) mice late in the development of acute lung injury. Thus, MT ultimately improves survival in the progression of acute lung injury in mice. Transcriptome-wide analysis suggests that this survival may be mediated through changes in the destabilization of transcripts associated with protein processing, the subsequent augmentation of transcripts controlling inflammation, extracellular matrix regulation, coagulation/fibrinolysis, and disruption of surfactant homeostasis.


Subject(s)
Metallothionein/metabolism , Respiratory Distress Syndrome/genetics , Respiratory Distress Syndrome/physiopathology , Animals , Disease Progression , Gene Expression Profiling , Gene Expression Regulation , Humans , Metallothionein/genetics , Mice , Mice, Inbred Strains , Mice, Knockout , Nickel/toxicity , Oligonucleotide Array Sequence Analysis , Respiratory Distress Syndrome/chemically induced , Respiratory Distress Syndrome/metabolism , Survival Rate
19.
Am J Physiol Heart Circ Physiol ; 286(2): H796-805, 2004 Feb.
Article in English | MEDLINE | ID: mdl-14563664

ABSTRACT

The phosphoinositide 3-kinase (PI3K) inhibitor LY-294002 decreased steady-state contraction in neonatal rat ventricular myocytes (NRVM). To determine whether the effect on steady-state contraction could be due to decreased intracellular Ca(2+) content, Ca(2+) content was assessed with fluorescent plate reader analysis by using the caffeine-releasable Ca(2+) stores as an index of sarcoplasmic reticulum (SR) Ca(2+) content. Caffeine-releasable Ca(2+) content was diminished in a dose-dependent manner with LY-294002, suggesting that the decrease in steady-state contraction was due to diminished intracellular Ca(2+) content. Activation of the L-type Ca(2+) channel by BAY K 8644 was attenuated by LY-294002, suggesting the effect of LY-294002 is to reduce Ca(2+) influx at this channel. To investigate whether additional proteins involved in excitation-contraction (EC) coupling are likewise regulated by PI3K activity, the effects of compounds acting at sarco(endo)plasmic reticulum Ca(2+)-ATPase (SERCA2a), the ryanodine receptor, and the Na/Ca exchanger (NCX) were compared with LY-294002. Inhibition of SERCA2a by thapsigargin increased basal Ca(2+) levels in contrast to LY-294002, indicating that SERCA2a activity is sustained in the presence of LY-294002. Ryanodine decreased SR Ca(2+) content. The additive effect with coadministration of LY-294002 could be attributed to a decrease in Ca(2+) influx at the L-type Ca(2+) channel. The NCX inhibitor Ni(2+) was used to investigate whether the decrease in intracellular Ca(2+) content with LY-294002 could be due to inhibition of the NCX reverse-mode activity. The minimal effect of LY-294002 with Ni(2+) suggests that the primary effect of LY-294002 on EC coupling occurs through inhibition of PI3K-mediated L-type Ca(2+) channel activity.


Subject(s)
Calcium/metabolism , Myocardial Contraction/physiology , Myocytes, Cardiac/physiology , Phosphatidylinositol 3-Kinases/physiology , 3-Pyridinecarboxylic acid, 1,4-dihydro-2,6-dimethyl-5-nitro-4-(2-(trifluoromethyl)phenyl)-, Methyl ester/pharmacology , Animals , Animals, Newborn , Calcium Channels, L-Type/drug effects , Calcium Channels, L-Type/physiology , Calcium-Transporting ATPases/metabolism , Cells, Cultured , Chromones/pharmacology , Endoplasmic Reticulum/drug effects , Endoplasmic Reticulum/physiology , Enzyme Inhibitors/pharmacology , Heart Ventricles/drug effects , Kinetics , Morpholines/pharmacology , Myocytes, Cardiac/enzymology , Phosphoinositide-3 Kinase Inhibitors , Piperazines/pharmacology , Platelet-Derived Growth Factor/pharmacology , Rats , Rats, Sprague-Dawley , Ryanodine Receptor Calcium Release Channel/physiology , Sarcoplasmic Reticulum/drug effects , Sarcoplasmic Reticulum/physiology , Sarcoplasmic Reticulum Calcium-Transporting ATPases , Sodium-Calcium Exchanger/metabolism , Ventricular Function
20.
Am J Respir Cell Mol Biol ; 28(2): 188-98, 2003 Feb.
Article in English | MEDLINE | ID: mdl-12540486

ABSTRACT

The role of nitric oxide (NO) in acute lung injury remains controversial. Although inhaled NO increases oxygenation in clinical trials, inhibiting NO-synthase (NOS) can be protective. To examine the latter, nickel-exposed mice were treated with saline or NOS inhibitor, N(G)-nitro-L-arginine methyl ester (L-NAME). Initial microarray analysis of nickel-induced gene expression of saline-treated mice revealed increased inflammatory mediator, matrix injury-repair, and hypoxia-induced factor-mediated sequences and decreased lung-specific (e.g., surfactant-associated protein B and C) sequences. Compared with saline control, L-NAME-treated mice had enhanced survival with attenuated serum nitrate/nitrite, endothelial NOS activity, and lavage neutrophils and protein. Although initial cytokine (i.e., interferon-gamma, interleukins-1beta and -6, macrophage inflammatory protein-2, monocyte chemotactic protein-1, and tumor necrosis factor-alpha) gene expression was similar between groups, subsequent larger cytokine increases only occurred in saline-treated mice. Similarly, surfactant protein gene expression decreased initially in both groups yet was restored subsequently with L-NAME treatment. Interestingly, the role of inducible NOS (iNOS) in these responses seems minimal. iNOS gene expression was unaltered, iNOS activity and nitrotyrosine residues were undetectable, and an iNOS antagonist, aminoguanidine, failed to increase survival. Rather, systemic L-NAME treatment appears to attenuate pulmonary endothelial NOS activity, subsequent cytokine expression, inflammation, and protein permeability, and thereby restores surfactant gene expression and increases survival.


Subject(s)
Lung/drug effects , Lung/metabolism , Nickel/toxicity , Nitric Oxide/antagonists & inhibitors , Pulmonary Surfactants/metabolism , Animals , Bronchoalveolar Lavage Fluid/chemistry , Bronchoalveolar Lavage Fluid/cytology , Cytokines/genetics , Enzyme Inhibitors/pharmacology , Gene Expression/drug effects , Lung Injury , Mice , Mice, Inbred A , NG-Nitroarginine Methyl Ester/pharmacology , Neutrophils/drug effects , Nitric Oxide Synthase/antagonists & inhibitors , Nitric Oxide Synthase Type II , Nitric Oxide Synthase Type III , Pulmonary Surfactant-Associated Protein A/genetics , Pulmonary Surfactant-Associated Protein B/genetics , Pulmonary Surfactant-Associated Protein C/genetics
SELECTION OF CITATIONS
SEARCH DETAIL