Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 31
Filter
Add more filters










Publication year range
1.
Nutrients ; 15(7)2023 Mar 30.
Article in English | MEDLINE | ID: mdl-37049536

ABSTRACT

Vitamin D plays a crucial role in modulating the innate immune response by interacting with its intracellular receptor, VDR. In this review, we address vitamin D/VDR signaling and how it contributes to the regulation of intestinal and respiratory microbiota. We additionally review some components of the innate immune system, such as the barrier function of the pulmonary and intestinal epithelial membranes and secretion of mucus, with their respective modulation by vitamin D. We also explore the mechanisms by which this vitamin D/VDR signaling mounts an antimicrobial response through the transduction of microbial signals and the production of antimicrobial peptides that constitute one of the body's first lines of defense against pathogens. Additionally, we highlight the role of vitamin D in clinical diseases, namely inflammatory bowel disease and acute respiratory distress syndrome, where excessive inflammatory responses and dysbiosis are hallmarks. Increasing evidence suggests that vitamin D supplementation may have potentially beneficial effects on those diseases.


Subject(s)
Inflammatory Bowel Diseases , Vitamin D , Humans , Vitamin D/physiology , Intestines , Immunity, Innate , Vitamins , Respiratory System , Receptors, Calcitriol
2.
Exp Ther Med ; 24(6): 763, 2022 Dec.
Article in English | MEDLINE | ID: mdl-36561976

ABSTRACT

Mitochondrial oxidative stress and dysfunction are major pathogenic features of cardiac injury induced by ischemia/reperfusion (I/R). MicroRNA-141 (miR-141) has been implicated in the mitochondrial dysfunction in cell-based models of oxidant stress. Thus, the main aim of the present study was to systematically assess the role of miR-141 in cardiomyocyte injury induced by simulated I/R. The challenge of HL-1 cardiomyocytes with hypoxia/reoxygenation (H/R) decreased cell viability, which was also associated with an increase in miR-141 expression. The H/R-induced cell injury was mitigated by a miR-141 inhibitor and exacerbated by a miR-141 mimic. Furthermore, H/R induced mitochondrial superoxide production, dysfunction (decreased oxygen utilization and membrane depolarization), as well as ultrastructural damage. These mitochondrial effects were mitigated by a miR-141 inhibitor and intensified by a miR-141 mimic. Luciferase reporter assay, reverse transcription-quantitative PCR, and western blot analyses identified sirtuin-1 (Sirt1) and mitofusin-2 (MFN2) as targets of miR-141. The silencing of Sirt1 reduced the MFN2 cardiomyocyte levels and reversed the alleviating effects of miR-141 inhibitor on mitochondrial function during H/R. Collectively, these findings suggest that miR-141 functions as a causative agent in cardiomyocyte injury induced by I/R, primarily by interfering with two mitochondrial regulatory proteins, Sirt1 and MFN2.

3.
Nutrients ; 13(7)2021 Jun 28.
Article in English | MEDLINE | ID: mdl-34203190

ABSTRACT

SARS-CoV-2 infects the respiratory tract and leads to the disease entity, COVID-19. Accordingly, the lungs bear the greatest pathologic burden with the major cause of death being respiratory failure. However, organs remote from the initial site of infection (e.g., kidney, heart) are not spared, particularly in severe and fatal cases. Emerging evidence indicates that an excessive inflammatory response coupled with a diminished antiviral defense is pivotal in the initiation and development of COVID-19. A common finding in autopsy specimens is the presence of thrombi in the lungs as well as remote organs, indicative of immunothrombosis. Herein, the role of SARS-CoV-2 in lung inflammation and associated sequelae are reviewed with an emphasis on immunothrombosis. In as much as vitamin D is touted as a supplement to conventional therapies of COVID-19, the impact of this vitamin at various junctures of COVID-19 pathogenesis is also addressed.


Subject(s)
COVID-19 Drug Treatment , COVID-19/immunology , Inflammation/virology , Pneumonia/virology , Vitamin D/therapeutic use , Animals , COVID-19/virology , Extracellular Traps , Humans , Inflammation/drug therapy , Lung/pathology , Mice , Multiple Organ Failure/virology , Pneumonia/drug therapy , Respiratory Distress Syndrome/virology , SARS-CoV-2 , Thrombosis/immunology , Thrombosis/virology , Vitamins/therapeutic use
4.
Front Cell Infect Microbiol ; 11: 679878, 2021.
Article in English | MEDLINE | ID: mdl-34178722

ABSTRACT

The respiratory tract is the major site of infection by SARS-CoV-2, the virus causing COVID-19. The pulmonary infection can lead to acute respiratory distress syndrome (ARDS) and ultimately, death. An excessive innate immune response plays a major role in the development of ARDS in COVID-19 patients. In this scenario, activation of lung epithelia and resident macrophages by the virus results in local cytokine production and recruitment of neutrophils. Activated neutrophils extrude a web of DNA-based cytoplasmic material containing antimicrobials referred to as neutrophil extracellular traps (NETs). While NETs are a defensive strategy against invading microbes, they can also serve as a nidus for accumulation of activated platelets and coagulation factors, forming thrombi. This immunothrombosis can result in occlusion of blood vessels leading to ischemic damage. Herein we address evidence in favor of a lung-centric immunothrombosis and suggest a lung-centric therapeutic approach to the ARDS of COVID-19.


Subject(s)
COVID-19 , Extracellular Traps , Respiratory Distress Syndrome , Humans , Lung , SARS-CoV-2
5.
Front Immunol ; 11: 558712, 2020.
Article in English | MEDLINE | ID: mdl-33101273

ABSTRACT

Sepsis/endotoxemia activates the NLRP3 inflammasome of macrophages leading to the maturation and release of IL-1ß, an important mediator of the inflammatory response. Reactive oxygen species have been implicated in NLRP3 inflammasome activation. Further, our preliminary studies indicated that LPS challenge of cardiac fibroblasts could phosphorylate protein kinase R (PKR) on threonine 451 and increase message for pro-IL-1 ß. Thus, the major aim of the present study was to address the role of PKR and the oxidant, peroxynitrite, in the two-tiered function of the NLRP3 inflammasome (priming and activation). Materials and Methods: Isolated murine fibroblasts were primed with LPS (1 µg/ml) for 6 h and subsequently activated by an ATP (3 mM) challenge for 30 min to induce optimum functioning of the inflammasome. Increased levels of NLRP3 and pro-IL-1ß protein (Western) were used as readouts for inflammasome priming, while activation of caspase 1 (p20) (Western) and secretion of IL-1ß (ELISA) were indicative of inflammasome activation. Results: Inhibition of PKR (PKR inhibitor or siRNA) prior to priming with LPS prevented the LPS-induced increase in NLRP3 and pro-IL-1ß expression. Further, inhibition of PKR after priming, but before activation, did not affect NLRP3 or pro-IL-1ß protein levels, but markedly reduced the activation of caspase 1 and secretion of mature IL-1ß. In a similar fashion, a peroxynitrite decomposition catalyst (Fe-TPPS) prevented both the priming and activation of the NLRP3 inflammasome. Finally, pretreatment of the fibroblasts with Fe-TPPS prevented the LPS-induced PKR phosphorylation (T451). Conclusion: Our results indicate that peroxynitrite-/PKR pathway modulates priming and activation of NLRP3 inflammasome in LPS/ATP challenged cardiac fibroblasts.


Subject(s)
Fibroblasts/drug effects , Fibroblasts/metabolism , Inflammasomes/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Peroxynitrous Acid/pharmacology , eIF-2 Kinase/metabolism , Animals , Mice , Models, Biological , Phosphorylation , Signal Transduction/drug effects
7.
J Steroid Biochem Mol Biol ; 200: 105663, 2020 06.
Article in English | MEDLINE | ID: mdl-32194242

ABSTRACT

Vitamin D plays a pivotal role in intestinal homeostasis. Vitamin D can impact the function of virtually every cell in the gut by binding to its intracellular receptor (VDR) and subsequently transcribing relevant genes. In the lumen, the mucus layer and the underlying epithelium serve to keep resident microbiota at bay. Vitamin D ensures an appropriate level of antimicrobial peptides in the mucus and maintains epithelial integrity by reinforcing intercellular junctions. Should bacteria penetrate the epithelial layer and enter the interstitium, immune sentinel cells (e.g. macrophages, dendritic cells, and innate lymphoid cells) elicit inflammation and trigger the adaptive immune response by activating Th1/Th17 cells. Vitamin D/VDR signaling in these cells ensures clearance of the bacteria. Subsequently, vitamin D also quiets the adaptive immune system by suppressing the Th1/Th17 cells and favoring Treg cells. The importance of vitamin D/VDR signaling in intestinal homeostasis is evidenced by the development of a chronic inflammatory state (e.g. IBD) when this signaling system is disrupted.


Subject(s)
Intestinal Mucosa , Vitamin D/metabolism , Adaptive Immunity , Animals , Gastrointestinal Microbiome , Homeostasis , Humans , Immunity, Innate , Immunomodulation , Intestinal Mucosa/immunology , Intestinal Mucosa/metabolism , Intestinal Mucosa/microbiology
8.
Int J Biochem Cell Biol ; 105: 94-103, 2018 12.
Article in English | MEDLINE | ID: mdl-30381241

ABSTRACT

Diabetes mellitus (DM) has a negative impact on clinical outcomes for patients with myocardial infarction. The aim of the present study was to assess whether decreased myocardial levels of Sirtuin1 (Sirt1) contribute to the increased susceptibility of the diabetic myocardium to ischemia/reperfusion (I/R) injury. In vivo, myocardial levels of Sirt1 expression and activity were decreased in mice with STZ-induced DM. Increasing Sirt1 activity prevented the DM-induced exacerbation of myocardial mitochondrial fission, apoptosis and dysfunction elicited by I/R. In vitro, anoxia/reoxygenation (A/R) challenge of cardiomyocytes (CM) that were preconditioned with high glucose (HG-CM) resulted in an exacerbation of the A/R-induced mitochondrial fission, oxidant production and CM apoptosis; effects reversed by increasing Sirt1 protein/activity. Inhibition of Drp1 prevented the exacerbated CM mitochondrial fission and oxidant production after A/R challenge of HG-CM. Decreased Sirt1 in HG-CM was associated with decreased Akt phosphorylation. Inhibition of Akt had no effect on CM Sirt1 levels, but further increased Drp1 activation. Increasing Sirt1 levels prevented the decrease in Akt phosphorylation and Drp1 activation in A/R challenged HG-CM. In conclusion: our data indicate that the increased vulnerability of the diabetic myocardium to I/R-induced apoptosis/dysfunction is attributable, in part, to decreased myocardial Sirt1 activity which leads to a decrease in Akt activation, an increase in Drp1 activity, culminating in excessive mitochondrial fission and ROS production.


Subject(s)
Diabetes Mellitus, Experimental/complications , Dynamins/metabolism , Mitochondrial Dynamics/physiology , Myocardial Reperfusion Injury/complications , Proto-Oncogene Proteins c-akt/metabolism , Sirtuin 1/metabolism , Animals , Apoptosis , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Experimental/pathology , Down-Regulation , Male , Mice , Mice, Inbred C57BL , Mitochondria, Heart/metabolism , Mitochondria, Heart/pathology , Myocardial Infarction/complications , Myocardial Infarction/metabolism , Myocardial Infarction/pathology , Myocardial Reperfusion Injury/metabolism , Myocardial Reperfusion Injury/pathology , Myocardium/metabolism , Myocardium/pathology , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Reactive Oxygen Species/metabolism , Sirtuin 1/genetics
9.
Pathophysiology ; 24(4): 213-228, 2017 Dec.
Article in English | MEDLINE | ID: mdl-29102280

ABSTRACT

Microvascular dysfunction is well established as an early and rate-determining factor in the injury response of tissues to ischemia and reperfusion (I/R). Severe endothelial cell dysfunction, which can develop without obvious morphological cell injury, is a major underlying cause of the microvascular abnormalities that accompany I/R. While I/R-induced microvascular dysfunction is manifested in different ways, two responses that have received much attention in both the experimental and clinical setting are impaired capillary perfusion (no-reflow) and endothelial barrier failure with a transition to hemorrhage. These responses are emerging as potentially important determinants of the severity of the tissue injury response, and there is growing clinical evidence that they are predictive of clinical outcome following reperfusion therapy. This review provides a summary of animal studies that have focused on the mechanisms that may underlie the genesis of no-reflow and hemorrhage following reperfusion of ischemic tissues, and addresses the clinical evidence that implicates these vascular events in the responses of the ischemic brain (stroke) and heart (myocardial infarction) to reperfusion therapy. Inasmuch as reactive oxygen species (ROS) and matrix metalloproteinases (MMP) are frequently invoked as triggers of the microvascular dysfunction elicited by I/R, the potential roles and sources of these mediators are also discussed. The available evidence in the literature justifies the increased interest in the development of no-reflow and hemorrhage in heart and brain following reperfusion therapy, and suggests that these vascular events may be predictive of poor clinical outcome and warrant the development of targeted treatment strategies.

10.
Redox Biol ; 6: 524-551, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26484802

ABSTRACT

Reperfusion injury, the paradoxical tissue response that is manifested by blood flow-deprived and oxygen-starved organs following the restoration of blood flow and tissue oxygenation, has been a focus of basic and clinical research for over 4-decades. While a variety of molecular mechanisms have been proposed to explain this phenomenon, excess production of reactive oxygen species (ROS) continues to receive much attention as a critical factor in the genesis of reperfusion injury. As a consequence, considerable effort has been devoted to identifying the dominant cellular and enzymatic sources of excess ROS production following ischemia-reperfusion (I/R). Of the potential ROS sources described to date, xanthine oxidase, NADPH oxidase (Nox), mitochondria, and uncoupled nitric oxide synthase have gained a status as the most likely contributors to reperfusion-induced oxidative stress and represent priority targets for therapeutic intervention against reperfusion-induced organ dysfunction and tissue damage. Although all four enzymatic sources are present in most tissues and are likely to play some role in reperfusion injury, priority and emphasis has been given to specific ROS sources that are enriched in certain tissues, such as xanthine oxidase in the gastrointestinal tract and mitochondria in the metabolically active heart and brain. The possibility that multiple ROS sources contribute to reperfusion injury in most tissues is supported by evidence demonstrating that redox-signaling enables ROS produced by one enzymatic source (e.g., Nox) to activate and enhance ROS production by a second source (e.g., mitochondria). This review provides a synopsis of the evidence implicating ROS in reperfusion injury, the clinical implications of this phenomenon, and summarizes current understanding of the four most frequently invoked enzymatic sources of ROS production in post-ischemic tissue.


Subject(s)
Reactive Oxygen Species/metabolism , Reperfusion Injury/metabolism , Animals , Cell Hypoxia , Humans , Oxidation-Reduction , Oxidative Stress
11.
Compr Physiol ; 5(3): 1541-83, 2015 Jul 01.
Article in English | MEDLINE | ID: mdl-26140727

ABSTRACT

The gastrointestinal (GI) circulation receives a large fraction of cardiac output and this increases following ingestion of a meal. While blood flow regulation is not the intense phenomenon noted in other vascular beds, the combined responses of blood flow, and capillary oxygen exchange help ensure a level of tissue oxygenation that is commensurate with organ metabolism and function. This is evidenced in the vascular responses of the stomach to increased acid production and in intestine during periods of enhanced nutrient absorption. Complimenting the metabolic vasoregulation is a strong myogenic response that contributes to basal vascular tone and to the responses elicited by changes in intravascular pressure. The GI circulation also contributes to a mucosal defense mechanism that protects against excessive damage to the epithelial lining following ingestion of toxins and/or noxious agents. Profound reductions in GI blood flow are evidenced in certain physiological (strenuous exercise) and pathological (hemorrhage) conditions, while some disease states (e.g., chronic portal hypertension) are associated with a hyperdynamic circulation. The sacrificial nature of GI blood flow is essential for ensuring adequate perfusion of vital organs during periods of whole body stress. The restoration of blood flow (reperfusion) to GI organs following ischemia elicits an exaggerated tissue injury response that reflects the potential of this organ system to generate reactive oxygen species and to mount an inflammatory response. Human and animal studies of inflammatory bowel disease have also revealed a contribution of the vasculature to the initiation and perpetuation of the tissue inflammation and associated injury response.


Subject(s)
Gastrointestinal Tract/blood supply , Hyperemia/etiology , Oxygen Consumption , Vasoconstriction , Animals , Autonomic Nervous System/metabolism , Autonomic Nervous System/physiology , Gastrointestinal Tract/physiology , Gastrointestinal Tract/physiopathology , Humans , Microvessels/metabolism , Microvessels/physiology
12.
Biochim Biophys Acta ; 1852(10 Pt A): 2075-85, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26209013

ABSTRACT

Diabetic cardiomyopathy (DiCM) is characterized by myocardial fibrosis and dysfunction. In rodent models of diabetes myocardial HMGB1 increases while IL-33 decreases. The major cardiac cell type expressing HMGB1 is the myocyte while the primary IL-33 expressing cell is the fibroblast. The aim of this study was to delineate the extracellular communication pathway(s) between cardiomyocytes and fibroblasts that contributes to murine DiCM. The streptozotocin (STZ)-induced murine model of diabetes and a cardiomyocyte/fibroblast co-culture challenged with high glucose were used. In STZ mice, myocardial HMGB1 expression was increased while IL-33 expression decreased (immunofluorescence and Western blot). In addition, STZ mice had an increased myocardial collagen deposition and myocardial dysfunction (pressure-volume loop analysis). An HMGB1 inhibitor (A-box) or exogenous IL-33 prevented the myocardial collagen deposition and dysfunction. In the cardiomyocyte/fibroblast co-culture model, HG increased cardiomyocyte HMGB1 secretion, decreased fibroblast IL-33 expression, and increased fibroblast collagen I production. Further, using A-box and HMGB1 shRNA transfected myocytes, we found that cardiomyocyte-derived HMGB1 dramatically potentiated the HG-induced down-regulation of IL-33 and the increase in collagen I expression in the fibroblasts. The potentiating effects of the cardiomyocytes was diminished when toll-like receptor 4 deficient (TLR4(-/-)) fibroblasts were co-cultured with wild-type myocytes. Finally, TLR4(-/-) mice with diabetes had increased myocardial expression of HMGB1, but failed to down-regulate IL-33. The diabetes-induced myocardial collagen deposition and cardiac dysfunction were significantly attenuated in TLR4(-/-) mice. In conclusion, our findings support a role for "cardiomyocyte HMGB1-fibroblast TLR4/IL-33 axis" in the development of myocardial fibrosis and dysfunction in a murine model of diabetes.

13.
PLoS One ; 9(9): e107639, 2014.
Article in English | MEDLINE | ID: mdl-25216263

ABSTRACT

Myocardial contractile dysfunction in sepsis is associated with the increased morbidity and mortality. Although the underlying mechanisms of the cardiac depression have not been fully elucidated, an exaggerated inflammatory response is believed to be responsible. Nucleotide-binding oligomerization domain-like receptor containing pyrin domain 3 (NLRP3) inflammasome is an intracellular platform that is involved in the maturation and release of interleukin (IL)-1ß. The aim of the present study is to evaluate whether sepsis activates NLRP3 inflammasome/caspase-1/IL-1ß pathway in cardiac fibroblasts (CFs) and whether this cytokine can subsequently impact the function of cardiomyocytes (cardiac fibroblast-myocyte cross-talk). We show that treatment of CFs with lipopolysaccharide (LPS) induces upregulation of NLRP3, activation of caspase-1, as well as the maturation (activation) and release of IL-1ß. In addition, the genetic (small interfering ribonucleic acid [siRNA]) and pharmacological (glyburide) inhibition of the NLRP3 inflammasome in CFs can block this signaling pathway. Furthermore, the inhibition of the NLRP3 inflammasome in cardiac fibroblasts ameliorated the ability of LPS-challenged CFs to impact cardiomyocyte function as assessed by intracellular cyclic adenosine monophosphate (cAMP) responses in cardiomyocytes. Salient features of this the NLP3 inflammasome/ caspase-1 pathway were confirmed in in vivo models of endotoxemia/sepsis. We found that inhibition of the NLRP3 inflammasome attenuated myocardial dysfunction in mice with LPS and increased the survival rate in mice with feces-induced peritonitis. Our results indicate that the activation of the NLRP3 inflammasome in cardiac fibroblasts is pivotal in the induction of myocardial dysfunction in sepsis.


Subject(s)
Cardiomyopathies/genetics , Carrier Proteins/biosynthesis , Inflammasomes/genetics , Sepsis/complications , Animals , Cardiomyopathies/complications , Cardiomyopathies/pathology , Carrier Proteins/genetics , Caspase 1/metabolism , Fibroblasts/drug effects , Fibroblasts/metabolism , Gene Expression Regulation/drug effects , Humans , Interleukin-1beta/metabolism , Lipopolysaccharides/toxicity , Mice , Myocardial Contraction/drug effects , NLR Family, Pyrin Domain-Containing 3 Protein , Sepsis/chemically induced , Sepsis/pathology , Signal Transduction/drug effects
15.
Free Radic Biol Med ; 52(3): 556-592, 2012 Feb 01.
Article in English | MEDLINE | ID: mdl-22154653

ABSTRACT

Inflammation is a complex and potentially life-threatening condition that involves the participation of a variety of chemical mediators, signaling pathways, and cell types. The microcirculation, which is critical for the initiation and perpetuation of an inflammatory response, exhibits several characteristic functional and structural changes in response to inflammation. These include vasomotor dysfunction (impaired vessel dilation and constriction), the adhesion and transendothelial migration of leukocytes, endothelial barrier dysfunction (increased vascular permeability), blood vessel proliferation (angiogenesis), and enhanced thrombus formation. These diverse responses of the microvasculature largely reflect the endothelial cell dysfunction that accompanies inflammation and the central role of these cells in modulating processes as varied as blood flow regulation, angiogenesis, and thrombogenesis. The importance of endothelial cells in inflammation-induced vascular dysfunction is also predicated on the ability of these cells to produce and respond to reactive oxygen and nitrogen species. Inflammation seems to upset the balance between nitric oxide and superoxide within (and surrounding) endothelial cells, which is necessary for normal vessel function. This review is focused on defining the molecular targets in the vessel wall that interact with reactive oxygen species and nitric oxide to produce the characteristic functional and structural changes that occur in response to inflammation. This analysis of the literature is consistent with the view that reactive oxygen and nitrogen species contribute significantly to the diverse vascular responses in inflammation and supports efforts that are directed at targeting these highly reactive species to maintain normal vascular health in pathological conditions that are associated with acute or chronic inflammation.


Subject(s)
Blood Vessels/metabolism , Inflammation/metabolism , Reactive Nitrogen Species/metabolism , Reactive Oxygen Species/metabolism , Animals , Blood Coagulation , Blood Vessels/pathology , Capillary Permeability , Humans , Inflammation/pathology , Neovascularization, Pathologic/metabolism , Thrombosis/metabolism , Vasomotor System/metabolism , Vasomotor System/pathology
16.
Ann N Y Acad Sci ; 1207 Suppl 1: E29-43, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20961304

ABSTRACT

Two of the principal functions of intestinal lymphatics are to assist in the maintenance of interstitial volume within relatively normal limits during alterations in capillary filtration (e.g., acute portal hypertension) and the removal of absorbed water and chylomicrons. The contribution of lymphatics to the prevention of interstitial overhydration or dehydration during alterations in transcapillary filtration is similar in the small intestine and colon. While the lymphatics of the small intestine contribute substantially to the removal of absorbed water (particularly at low and moderate absorption rates), the contribution of colonic lymphatics to the removal of the fluid absorbate is negligible. This difference is attributed to the relative caliber and location of lymphatics in the mucosal layer of the small and large intestines. In the small intestine, large lacteals lie in close proximity to transporting epithelium, while colonic lymph vessels are rather sparse and confined to the basal portion of the mucosa. In the small intestine, the lymphatics assume a more important role in removing absorbed water during lipid absorption than during glucose absorption.


Subject(s)
Body Water , Intestinal Mucosa/physiology , Lymphatic Vessels/physiology , Animals , Humans
17.
J Immunol ; 184(3): 1492-8, 2010 Feb 01.
Article in English | MEDLINE | ID: mdl-20028656

ABSTRACT

High mobility group box 1 (HMGB1) is an alarmin actively secreted by immune cells and passively released by necrotic nonimmune cells. HMGB1 has been implicated in both cardiac contractile dysfunction and the lethality associated with sepsis/endotoxemia. The aim of the current study was to assess whether viable cardiomyocytes could produce HMGB1 and whether HMGB1 can affect myocardial contractility. LPS was used as a model of sepsis/endotoxemia in mice and isolated cardiac myocytes. LPS increased myocardial expression of HMGB1 in vivo (immunohistochemistry) and production and secretion of HMGB1 by viable cardiac myocytes in vitro (Western). LPS increased the phosphorylation status of PI3Kgamma in cardiac myocytes, an effect not observed in TLR4(-/-) myocytes. Genetic (PI3Kgamma(-/-)) or pharmacologic (AS605240) blockade of PI3Kgamma ameliorated the LPS-induced 1) cardiomyocyte production and secretion of HMGB1 in vitro and 2) HMGB1 expression in the myocardium in vivo. The LPS-induced depression of myocardial contractility was prevented by the HMGB1 antagonist, A-box. Genetic (PI3Kgamma(-/-)) or pharmacologic (AS605240) blockade of PI3Kgamma ameliorated the LPS-induced decrease in myocardial contractility. No evidence of inflammatory infiltrate was noted in any of the in vivo studies. The findings of the current study indicate that 1) LPS can induce HMGB1 secretion by viable cardiac myocytes through a TLR4/PI3Kgamma signaling pathway, and 2) HMGB1 plays a role in the LPS-induced myocardial contractile dysfunction. The results of the current study also have broader implications (i.e., that viable parenchymal cells, such as cardiac myocytes, participate in the alarmin response).


Subject(s)
Cardiomyopathies/immunology , Cytokines/biosynthesis , HMGB1 Protein/biosynthesis , Myocytes, Cardiac/immunology , Myocytes, Cardiac/metabolism , Phosphatidylinositol 3-Kinases/physiology , Signal Transduction/immunology , Toll-Like Receptor 4/physiology , Animals , Animals, Newborn , Cardiomyopathies/enzymology , Cardiomyopathies/physiopathology , Cell Survival/immunology , Cells, Cultured , Class Ib Phosphatidylinositol 3-Kinase , Cytokines/metabolism , Cytokines/physiology , Disease Models, Animal , Endotoxemia/enzymology , Endotoxemia/immunology , Endotoxemia/physiopathology , HMGB1 Protein/metabolism , HMGB1 Protein/physiology , Isoenzymes/deficiency , Isoenzymes/genetics , Isoenzymes/physiology , Lipopolysaccharides/toxicity , Mice , Mice, Inbred C57BL , Mice, Knockout , Myocardial Contraction/genetics , Myocardial Contraction/immunology , Phosphatidylinositol 3-Kinases/deficiency , Phosphatidylinositol 3-Kinases/genetics , Signal Transduction/genetics , Toll-Like Receptor 4/deficiency , Toll-Like Receptor 4/genetics
18.
Free Radic Biol Med ; 46(10): 1338-45, 2009 May 15.
Article in English | MEDLINE | ID: mdl-19249346

ABSTRACT

It has been reported that polymorphonuclear leukocyte (PMN) infiltration into the myocardial interstitium is involved in sepsis-induced myocardial dysfunction. The aim of this study was to evaluate the role of NADPH oxidase in the sepsis-induced conversion of cardiomyocytes to a proinflammatory phenotype. Using an in vitro approach we evaluated the role of NADPH oxidase in cardiomyocyte CXC chemokine production and its ability to promote PMN transendothelial migration under septic conditions. Treatment of cardiac myocytes with septic plasma (1) activated NADPH oxidase (p47phox phosphorylation) and increased its activity (O(2)(-) production) and (2) converted them to a proinflammatory phenotype; both effects were prevented by blockade of NADPH oxidase. NF-kappaB nuclear translocation was increased in cardiomyocytes conditioned with septic plasma, a response prevented by blockade of NADPH oxidase. The increase in NF-kappaB activation/translocation was associated with phosphorylation of both IKK and the p65 subunit of NF-kappaB. Blockade of NADPH oxidase prevented phosphorylation of IKK, but not p65. Blockade approaches indicated that p38 MAP kinase (previously implicated in NF-kappaB activation) did not play a role in the NADPH oxidase pathway, either upstream or downstream. Collectively, the results of this study and those of previous reports indicate that the conversion of cardiomyocytes to a proinflammatory phenotype in sepsis involves two distinct pathways: NADPH oxidase-mediated phosphorylation of IKK and p38 MAP kinase-mediated phosphorylation of p65.


Subject(s)
Cell Nucleus/metabolism , Myocytes, Cardiac/metabolism , NADPH Oxidases/metabolism , NF-kappa B/metabolism , Peritonitis/physiopathology , Active Transport, Cell Nucleus , Animals , Cell Differentiation , Cell Movement , I-kappa B Kinase/metabolism , Inflammation , Membrane Glycoproteins/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Myocytes, Cardiac/immunology , Myocytes, Cardiac/pathology , NADPH Oxidase 2 , NADPH Oxidases/genetics , Neutrophils/pathology , Peritonitis/pathology , Sepsis/physiopathology , Signal Transduction , Transcription Factor RelA/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism
19.
J Immunol ; 181(12): 8735-44, 2008 Dec 15.
Article in English | MEDLINE | ID: mdl-19050294

ABSTRACT

Alveolar macrophages (AMphi) have been implicated in the polymorphonuclear leukocyte (PMN) recruitment to the lungs during sepsis. Using an in vivo murine model of sepsis (feces in the peritoneum), we show that peritonitis leads to increased activation of AMphi and PMN migration into pulmonary alveoli. To assess cellular mechanisms, an in vitro construct of the pulmonary vascular-interstitial interface (murine AMphi, pulmonary endothelial cells, and PMN) and a chimera approach were used. Using immunologic (Abs) and genetic blockade (CXCR2-deficient AMphi), we show that CXC chemokines in septic plasma are responsible for the activation of AMphi. The activated AMphi can promote PMN transendothelial migration, even against a concentration gradient of septic plasma, by generating platelet-activating factor and H(2)O(2). Platelet-activating factor/H(2)O(2) induce an oxidant stress in the adjacent endothelial cells, an event that appears to be a prerequisite for PMN transendothelial migration, since PMN migration is abrogated across Cu/Zn-superoxide dismutase overexpressing endothelial cells. Using gp91-deficient endothelial cells, we show that NADPH oxidase plays an important role in the AMphi-induced PMN transendothelial migration. Pharmacologic/small interfering RNA blockade of Src kinase inhibits AMphi-induced endothelial NADPH oxidase activation and PMN migration. Collectively, our findings indicate that the PMN transendothelial migration induced by septic AMphi is dependent on the generation of superoxide in endothelial cells via the Src kinase/NADPH oxidase signaling pathway.


Subject(s)
Chemotaxis, Leukocyte/immunology , Endothelial Cells/immunology , Macrophages, Alveolar/immunology , Membrane Glycoproteins/physiology , NADPH Oxidases/physiology , Neutrophils/immunology , Peritonitis/immunology , Protein-Tyrosine Kinases/physiology , Signal Transduction/immunology , Animals , Bronchoalveolar Lavage Fluid/cytology , Bronchoalveolar Lavage Fluid/immunology , CSK Tyrosine-Protein Kinase , Cells, Cultured , Chemotaxis, Leukocyte/genetics , Endothelial Cells/enzymology , Endothelial Cells/pathology , Macrophages, Alveolar/enzymology , Macrophages, Alveolar/pathology , Membrane Glycoproteins/deficiency , Membrane Glycoproteins/genetics , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , NADPH Oxidase 2 , NADPH Oxidases/deficiency , NADPH Oxidases/genetics , Neutrophils/enzymology , Neutrophils/pathology , Peritonitis/enzymology , Peritonitis/genetics , Protein-Tyrosine Kinases/deficiency , Protein-Tyrosine Kinases/genetics , Pulmonary Alveoli/enzymology , Pulmonary Alveoli/immunology , Pulmonary Alveoli/pathology , Sepsis/enzymology , Sepsis/genetics , Sepsis/immunology , Signal Transduction/genetics , Superoxide Dismutase/metabolism , src-Family Kinases
20.
J Immunol ; 181(3): 2145-54, 2008 Aug 01.
Article in English | MEDLINE | ID: mdl-18641353

ABSTRACT

PECAM-1 is expressed on endothelial cells and leukocytes. Its extracellular domain has been implicated in leukocyte diapedesis. In this study, we used PECAM-1(-/-) mice and relevant cells derived from them to assess the role of PECAM-1 in an experimental model of acute colonic inflammation with a predominant innate immune response, i.e., 2,4,6-trinitrobenzine sulfonic acid (TNBS). Using chimeric approaches, we addressed the points of control exerted by PECAM-1 along the macrophage-endothelial cell-polymorphonuclear neutrophil (PMN) axis. In vivo, TNBS-induced colitis was ameliorated in PECAM-1(-/-) mice, an event attributed to PECAM-1 on hematopoietic cells rather than to PECAM-1 on endothelial cells. The in vivo innate immune response was mimicked in vitro by using a construct of the vascular-interstitial interface, i.e., PMN transendothelial migration was induced by colonic lavage fluid (CLF) from TNBS mice or macrophages (MPhi) challenged with CLF. Using the construct, we confirmed that endothelial cell PECAM-1 does not play a role in PMN transendothelial migration. Although MPhi activation (NF-kappaB nuclear binding) and function (keratinocyte-derived chemokine production) induced by CLF was diminished in PECAM-1(-/-) MPhi, this did not affect their ability to promote PMN transendothelial migration. By contrast, PECAM-1(-/-) PMN did not adhere to or migrate across endothelial cell monolayers in response to CLF. Further, as compared with PECAM-1(+/+) PMN, PECAM-1(-/-) PMN were less effective in orientating their CXCR2 receptors (polarization) in the direction of a chemotactic gradient. Collectively, our findings indicate that PECAM-1 modulation of PMN function (at a step before diapedesis) most likely contributes to the inflammation in a colitis model with a strong innate immune component.


Subject(s)
Colitis/immunology , Colitis/metabolism , Endothelial Cells/immunology , Immunity, Innate/immunology , Macrophages/immunology , Neutrophils/immunology , Platelet Endothelial Cell Adhesion Molecule-1/metabolism , Acute Disease , Animals , Cell Adhesion , Cell Movement/immunology , Cells, Cultured , Colitis/chemically induced , Colitis/pathology , Macrophages/cytology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Molecular Mimicry/immunology , Neutrophils/cytology , Platelet Endothelial Cell Adhesion Molecule-1/genetics , Trinitrobenzenesulfonic Acid/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...