Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 34
Filter
Add more filters










Publication year range
1.
Curr Protoc ; 2(9): e549, 2022 Sep.
Article in English | MEDLINE | ID: mdl-36102926

ABSTRACT

Immunohistochemistry is an essential technique for the localization and measurement of proteins in cells and tissues. This article describes methods for labeling proteins in adherent and suspension cell cultures and in tissue sections. Choices of antibodies and detection methods are discussed, and detailed troubleshooting guidelines are provided. © 2022 Wiley Periodicals LLC. Basic Protocol 1: Immunofluorescent labeling of cells grown as adherent monolayers Alternate Protocol 1: Immunofluorescent labeling of cells in suspension Basic Protocol 2: Immunofluorescent labeling of tissue sections Alternate Protocol 2: Immunofluorescent labeling using streptavidin-biotin conjugates Alternate Protocol 3: Immunofluorescent double-labeling of tissue sections Alternate Protocol 4: Immunofluorescent double-labeling of tissue sections with two primary antibodies from the same host species.


Subject(s)
Antibodies , Biotin , Antigens , Immunohistochemistry , Proteins , Streptavidin
2.
J Cereb Blood Flow Metab ; 42(12): 2255-2269, 2022 12.
Article in English | MEDLINE | ID: mdl-35854408

ABSTRACT

Epinephrine is the principal resuscitation therapy for pediatric cardiac arrest (CA). Clinical data suggest that although epinephrine increases the rate of resuscitation, it fails to improve neurological outcome, possibly secondary to reductions in microvascular flow. We characterized the effect of epinephrine vs. placebo administered at resuscitation from pediatric asphyxial CA on microvascular and macrovascular cortical perfusion assessed using in vivo multiphoton microscopy and laser speckle flowmetry, respectively, and on brain tissue oxygenation (PbO2), behavioral outcomes, and neuropathology in 16-18-day-old rats. Epinephrine-treated rats had a more rapid return of spontaneous circulation and brisk immediate cortical reperfusion during 1-3 min post-CA vs. placebo. However, at the microvascular level, epinephrine-treated rats had penetrating arteriole constriction and increases in both capillary stalling (no-reflow) and cortical capillary transit time 30-60 min post-CA vs. placebo. Placebo-treated rats had increased capillary diameters post-CA. The cortex was hypoxic post-CA in both groups. Epinephrine treatment worsened reference memory performance vs. shams. Hippocampal neuron counts did not differ between groups. Resuscitation with epinephrine enhanced immediate reperfusion but produced microvascular alterations during the first hour post-resuscitation, characterized by vasoconstriction, capillary stasis, prolonged cortical transit time, and absence of compensatory cortical vasodilation. Targeted therapies mitigating the deleterious microvascular effects of epinephrine are needed.


Subject(s)
Cardiopulmonary Resuscitation , Heart Arrest , Animals , Rats , Microscopy , Cerebrovascular Circulation/physiology , Heart Arrest/drug therapy , Heart Arrest/complications , Epinephrine/pharmacology , Epinephrine/therapeutic use , Resuscitation
3.
Curr Protoc ; 2(1): e342, 2022 Jan.
Article in English | MEDLINE | ID: mdl-35038380

ABSTRACT

The protocols presented here describe steps for cryosectioning tissue samples to be used in light microscopy methodologies including histochemistry, enzyme immunohistochemistry, and immunofluorescence. © 2022 Wiley Periodicals LLC. Basic Protocol 1: Cryosectioning.


Subject(s)
Cryoultramicrotomy , Fluorescent Antibody Technique , Immunohistochemistry , Tissue Fixation
4.
Curr Protoc ; 2(1): e343, 2022 Jan.
Article in English | MEDLINE | ID: mdl-35030299

ABSTRACT

Gravity flow whole-body perfusion maintains effective and reproducible preservation of tissue architecture critical to investigate pathobiology of multiple organs from the same specimen. The purpose of the protocols described within this article is to help the researcher optimize tissue harvest for multisystem pathobiology comparison. The protocols presented here describe tissue harvest for processing and cryopreservation to generate optimal samples for microscopy and parallel biochemical and molecular biology analysis. First, this paper outlines a protocol for tissue perfusion and organ harvest that allows the researcher multiple analysis options from the same research subject simultaneously. Second, this paper outlines a model to optimize ex-vivo tissue fixation for precious human sample preparation. Finally, this paper outlines a methodology for freezing tissue samples to optimize their capacity for biochemical and immunohistochemical analysis. Benefits and alternative approaches to retain cellular morphology in tissue harvest and processing are discussed. Also described within each section are common technical issues to assist problem-solving and troubleshooting. © 2022 Wiley Periodicals LLC. Basic Protocol 1: Whole body in vivo tissue perfusion by gravity flow: preparation and surgical procedures Alternate Protocol: Human ex vivo tissue fixation Basic Protocol 2: Freezing of tissue samples.


Subject(s)
Specimen Handling , Tissue and Organ Harvesting , Humans , Perfusion , Tissue Fixation
5.
Blood Coagul Fibrinolysis ; 33(1): 14-24, 2022 Jan 01.
Article in English | MEDLINE | ID: mdl-34889809

ABSTRACT

Acute kidney injury (AKI) is common after trauma, but contributory factors are incompletely understood. Increases in plasma von Willebrand Factor (vWF) with concurrent decreases in ADAMTS13 are associated with renal microvascular thrombosis in other disease states, but similar findings have not been shown in trauma. We hypothesized that molecular changes in circulating vWF and ADAMTS13 promote AKI following traumatic injury. VWF antigen, vWF multimer composition and ADAMTS13 levels were compared in plasma samples from 16 trauma patients with and without trauma-induced AKI, obtained from the Prehospital Air Medical Plasma (PAMPer) biorepository. Renal histopathology and function, vWF and ADAMTS13 levels were assessed in parallel in a murine model of polytrauma and haemorrhage. VWF antigen was higher in trauma patients when compared with healthy controls [314% (253-349) vs. 100% (87-117)] [median (IQR)], while ADAMTS13 activity was lower [36.0% (30.1-44.7) vs. 100.0% (83.1-121.0)]. Patients who developed AKI showed significantly higher levels of high molecular weight multimeric vWF at 72-h when compared with non-AKI counterparts [32.9% (30.4-35.3) vs. 27.8% (24.6-30.8)]. Murine plasma cystatin C and vWF were elevated postpolytrauma model in mice, with associated decreases in ADAMTS13, and immunohistologic analysis demonstrated renal injury with small vessel plugs positive for fibrinogen and vWF. Following traumatic injury, the vWF-ADAMTS13 axis shifted towards a prothrombotic state in both trauma patients and a murine model. We further demonstrated that vWF-containing, microangiopathic deposits were concurrently produced as the prothrombotic changes were sustained during the days following trauma, potentially contributing to AKI development.


Subject(s)
Acute Kidney Injury , von Willebrand Factor , ADAMTS13 Protein , Animals , Humans , Kidney , Mice , Molecular Weight , Plasma
6.
Mol Med ; 27(1): 18, 2021 02 25.
Article in English | MEDLINE | ID: mdl-33632134

ABSTRACT

BACKGROUND: Hepatic ischemia/reperfusion (I/R) injury can be a major complication following liver surgery contributing to post-operative liver dysfunction. Maresin 1 (MaR1), a pro-resolving lipid mediator, has been shown to suppress I/R injury. However, the mechanisms that account for the protective effects of MaR1 in I/R injury remain unknown. METHODS: WT (C57BL/6J) mice were subjected to partial hepatic warm ischemia for 60mins followed by reperfusion. Mice were treated with MaR1 (5-20 ng/mouse), Boc2 (Lipoxin A4 receptor antagonist), LY294002 (Akt inhibitor) or corresponding controls just prior to liver I/R or at the beginning of reperfusion. Blood and liver samples were collected at 6 h post-reperfusion. Serum aminotransferase, histopathologic changes, inflammatory cytokines, and oxidative stress were analyzed to evaluate liver injury. Signaling pathways were also investigated in vitro using primary mouse hepatocyte (HC) cultures to identify underlying mechanisms for MaR1 in liver I/R injury. RESULTS: MaR1 treatment significantly reduced ALT and AST levels, diminished necrotic areas, suppressed inflammatory responses, attenuated oxidative stress and decreased hepatocyte apoptosis in liver after I/R. Akt signaling was significantly increased in the MaR1-treated liver I/R group compared with controls. The protective effect of MaR1 was abrogated by pretreatment with Boc2, which together with MaR1-induced Akt activation. MaR1-mediated liver protection was reversed by inhibition of Akt. CONCLUSIONS: MaR1 protects the liver against hepatic I/R injury via an ALXR/Akt signaling pathway. MaR1 may represent a novel therapeutic agent to mitigate the detrimental effects of I/R-induced liver injury.


Subject(s)
Docosahexaenoic Acids/therapeutic use , Liver Diseases/drug therapy , Protective Agents/therapeutic use , Proto-Oncogene Proteins c-akt/metabolism , Receptors, Formyl Peptide/metabolism , Reperfusion Injury/drug therapy , Alanine Transaminase/blood , Animals , Aspartate Aminotransferases/blood , Cell Survival/drug effects , Cytokines/blood , Docosahexaenoic Acids/pharmacology , Glutathione Peroxidase/metabolism , Hepatocytes/drug effects , Liver/drug effects , Liver/metabolism , Liver/pathology , Liver Diseases/blood , Liver Diseases/metabolism , Liver Diseases/pathology , Male , Malondialdehyde/metabolism , Mice, Inbred C57BL , Oxidative Stress/drug effects , Protective Agents/pharmacology , Receptors, Formyl Peptide/genetics , Reperfusion Injury/blood , Reperfusion Injury/metabolism , Reperfusion Injury/pathology , Signal Transduction/drug effects
7.
Front Immunol ; 11: 229, 2020.
Article in English | MEDLINE | ID: mdl-32328059

ABSTRACT

High-mobility group box-1 (HMGB1), a ubiquitous nuclear protein, acts as a late mediator of lethality when released extracellularly during sepsis. The major source of circulating HMGB1 in sepsis is hepatocytes. However, the mechanism of HMGB1 release of hepatocytes during sepsis is not very clear. We have previously shown that bacterial endotoxin [lipopolysaccharide (LPS)] sensing pathways, including Toll-like receptor (TLR)4 and caspase-11, regulate hepatocyte HMGB1 release in response to LPS. Here, we report the novel function of caspase-11 and gasdermin D (GsdmD) in LPS-induced active HMGB1 released from hepatocytes. HMGB1 release during endotoxemia was caspase-11/GsdmD dependent via an active way in vivo and in vitro. Caspase-11/GsdmD was responsible for HMGB1 translocation from nucleus to the cytoplasm via calcium changing-induced phosphorylation of calcium-calmodulin kinase kinase (camkk)ß during endotoxemia. Cleaved GsdmD accumulated on the endoplasmic reticulum, suggesting this may lead to calcium leak and intracellular calcium increase. Furthermore, we investigated that exosome was an important pathway for HMGB1 release from hepatocytes; this process was dependent on TLR4, independent of caspase-11 and GsdmD in vivo and in vitro. These findings provide a novel mechanism that TLR4 signaling results in an increase in caspase-11 expression, as well as increased exosome release, while caspase-11/GsdmD activation/cleavage leads to accumulation of HMGB1 in the cytoplasm through a process associated with the release of calcium from the endoplasmic reticulum and camkkß activation.


Subject(s)
Caspases, Initiator/metabolism , Exosomes/metabolism , HMGB1 Protein/metabolism , Hepatocytes/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Phosphate-Binding Proteins/metabolism , Sepsis/metabolism , Toll-Like Receptor 4/metabolism , Animals , Calcium/metabolism , Calcium-Calmodulin-Dependent Protein Kinase Kinase/metabolism , Caspases, Initiator/genetics , Endoplasmic Reticulum/metabolism , Humans , Intracellular Signaling Peptides and Proteins/genetics , Lipopolysaccharides/immunology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Phosphate-Binding Proteins/genetics , Signal Transduction , Toll-Like Receptor 4/genetics
8.
Hepatology ; 72(4): 1394-1411, 2020 10.
Article in English | MEDLINE | ID: mdl-31997373

ABSTRACT

BACKGROUND AND AIMS: Itaconate, a metabolite of the tricarboxylic acid cycle, plays anti-inflammatory roles in macrophages during endotoxemia. The mechanisms underlying its anti-inflammatory roles have been shown to be mediated by the modulation of oxidative stress, an important mechanism of hepatic ischemia-reperfusion (I/R) injury. However, the role of itaconate in liver I/R injury is unknown. APPROACH AND RESULTS: We found that deletion of immune-responsive gene 1 (IRG1), encoding for the enzyme producing itaconate, exacerbated liver injury and systemic inflammation. Furthermore, bone marrow adoptive transfer experiments indicated that deletion of IRG1 in both hematopoietic and nonhematopoietic compartments contributes to the protection mediated by IRG1 after I/R. Interestingly, the expression of IRG1 was up-regulated in hepatocytes after I/R and hypoxia/reoxygenation-induced oxidative stress. Modulation of the IRG1 expression levels in hepatocytes regulated hepatocyte cell death. Importantly, addition of 4-octyl itaconate significantly improved liver injury and hepatocyte cell death after I/R. Furthermore, our data indicated that nuclear factor erythroid 2-related factor 2 (Nrf2) is required for the protective effect of IRG1 on mouse and human hepatocytes against oxidative stress-induced injury. Our studies document the important role of IRG1 in the acute setting of sterile injury induced by I/R. Specifically, we provide evidence that the IRG1/itaconate pathway activates Nrf2-mediated antioxidative response in hepatocytes to protect liver from I/R injury. CONCLUSIONS: Our data expand on the importance of IRG1/itaconate in nonimmune cells and identify itaconate as a potential therapeutic strategy for this unfavorable postsurgical complication.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Carboxy-Lyases/physiology , Hepatocytes/metabolism , Liver/blood supply , NF-E2-Related Factor 2/physiology , Reperfusion Injury/prevention & control , Succinates/pharmacology , Animals , Humans , Hydro-Lyases/physiology , Male , Mice , Mice, Inbred C57BL , Oxidative Stress , Signal Transduction/physiology , Succinates/therapeutic use
9.
J Cereb Blood Flow Metab ; 39(5): 913-925, 2019 05.
Article in English | MEDLINE | ID: mdl-29192562

ABSTRACT

Decreased cerebral blood flow (CBF) after cardiac arrest (CA) contributes to secondary ischemic injury in infants and children. We previously reported cortical hypoperfusion with tissue hypoxia early in a pediatric rat model of asphyxial CA. In order to identify specific alterations as potential therapeutic targets to improve cortical hypoperfusion post-CA, we characterize the CBF alterations at the cortical microvascular level in vivo using multiphoton microscopy. We hypothesize that microvascular constriction and disturbances of capillary red blood cell (RBC) flow contribute to cortical hypoperfusion post-CA. After resuscitation from 9 min asphyxial CA, transient dilation of capillaries and venules at 5 min was followed by pial arteriolar constriction at 30 and 60 min (19.6 ± 1.3, 19.3 ± 1.2 µm at 30, 60 min vs. 22.0 ± 1.2 µm at baseline, p < 0.05). At the capillary level, microcirculatory disturbances were highly heterogeneous, with RBC stasis observed in 25.4% of capillaries at 30 min post-CA. Overall, the capillary plasma mean transit time was increased post-CA by 139.7 ± 51.5%, p < 0.05. In conclusion, pial arteriolar constriction, the no-reflow phenomenon and increased plasma transit time were observed post-CA. Our results detail the microvascular disturbances in a pediatric asphyxial CA model and provide a powerful platform for assessing specific vascular-targeted therapies.


Subject(s)
Brain/blood supply , Cerebrovascular Circulation , Heart Arrest/complications , Microcirculation , No-Reflow Phenomenon/etiology , Animals , Brain/physiopathology , Heart Arrest/physiopathology , Male , No-Reflow Phenomenon/physiopathology , Rats, Sprague-Dawley , Vasodilation
10.
Blood Adv ; 2(6): 638-648, 2018 03 27.
Article in English | MEDLINE | ID: mdl-29563120

ABSTRACT

Thrombocytopenia impairs host defense and hemostasis in sepsis. However, the mechanisms of how platelets regulate host defense are not fully understood. High-mobility group box 1 (HMGB1), a danger-associated molecular pattern protein, is released during infection and contributes to the pathogenesis of sepsis. Platelets express HMGB1, which is released on activation and has been shown to play a critical role in thrombosis, monocyte recruitment, and neutrophil extracellular trap (NET) production. However, the contribution of platelet HMGB1 to host defense is unknown. To determine the role of platelet HMGB1 in polymicrobial sepsis, platelet-specific HMGB1 knockout (HMGB1 platelet factor 4 [PF4]) mice were generated and were subjected to cecal ligation and puncture (CLP), a clinically relevant intra-abdominal sepsis model. Compared with HMGB1 Flox mice and wild-type (WT) mice, HMGB1 PF4 mice showed significantly higher bacterial loads in the peritoneum and blood, an exaggerated systemic inflammation response, and significantly greater mortality after CLP. Deletion of HMGB1 in platelets was associated with lower platelet-derived chemokines (PF4 and RANTES) in the peritoneal cavity, and a decrease of platelet-neutrophil interaction in the lung after CLP. In vitro, neutrophils cocultured with activated HMGB1 knockout platelets showed fewer platelet-neutrophil aggregates, reduced reactive oxygen species (ROS) burst as compared with control. Taken together, these data reveal an unrecognized role of platelet HMGB1 in the regulation of neutrophil recruitment and activation via modulation of platelet activation during sepsis.


Subject(s)
Abdominal Cavity/microbiology , Bacterial Load , Blood Platelets/metabolism , HMGB1 Protein/genetics , Sepsis/genetics , Sepsis/microbiology , Adoptive Transfer , Animals , Cell Communication , Disease Models, Animal , Extracellular Traps/immunology , Extracellular Traps/metabolism , Male , Mice , Mice, Transgenic , Neutrophil Activation/immunology , Neutrophils/immunology , Neutrophils/metabolism , Neutrophils/microbiology , Platelet Activation , Reactive Oxygen Species/metabolism , Sepsis/metabolism , Sepsis/mortality
11.
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
12.
Curr Protoc Cytom ; 83: 12.2.1-12.2.14, 2018 01 18.
Article in English | MEDLINE | ID: mdl-29345327

ABSTRACT

Modern digital microscopy combines the equipment of classical light microscopy with a computerized imaging system. The technique comprises image formation by optics, image registration by a camera, and saving of image data in a computer file. This chapter describes limitations that are particular to each of these processes, including optical resolution, efficiency of image registration, characteristics of image file formats, and data management. Further suggestions are given which serve, in turn, to help construct a set of guidelines aimed at optimization of digital microscopic imaging. © 2018 by John Wiley & Sons, Inc.


Subject(s)
Image Processing, Computer-Assisted/instrumentation , Image Processing, Computer-Assisted/methods , Microscopy/instrumentation , Microscopy/methods
13.
J Leukoc Biol ; 103(2): 175-183, 2018 02.
Article in English | MEDLINE | ID: mdl-28801344

ABSTRACT

Hemorrhagic shock (HS) renders patients susceptible to development of systemic inflammatory response syndrome (SIRS) and multiple organ dysfunction syndrome (MODS) through mechanisms that are, as yet, unclear. Cell necroptosis, a form of regulated inflammatory cell death, is one of the mechanisms that controls cell release of inflammatory mediators from innate immune cells, such as polymorphonuclear neutrophils (PMNs), and critically regulates the progress of inflammation. In this study, we investigated the mechanisms of alveolar macrophage (AMϕ) effects on PMN necroptosis following HS. With the use of in vivo and ex vivo HS models, we reveal a novel function of shock-activated AMϕ in promoting PMN necroptosis. We demonstrate that exosomes released from HS-activated AMϕ induce mainly NADPH oxidase-derived reactive oxygen species (ROS) production inside PMNs and subsequent promotion of necroptosis. These findings explore a previously unidentified pathway of AMϕ-PMN cross-talk, which causes enhanced PMN necroptosis and subsequent exaggerated post-HS lung inflammation. The targeting of this PMN death pathway may serve as a new therapeutic strategy for treatment of post-HS SIRS.


Subject(s)
Macrophages, Alveolar/pathology , Neutrophils/pathology , Shock, Hemorrhagic/pathology , Animals , Coculture Techniques , Disease Models, Animal , Exosomes/pathology , Humans , Inflammation/pathology , Male , Mice , Mice, Inbred C57BL , NADPH Oxidases/metabolism , Necrosis , Primary Cell Culture , Reactive Oxygen Species/metabolism
14.
Front Immunol ; 8: 1672, 2017.
Article in English | MEDLINE | ID: mdl-29234326

ABSTRACT

Trauma combined with hemorrhagic shock (HS/T) leads to systemic inflammation, which results in organ injury. Toll-like Receptor 4 (TLR4)-signaling activation contributes to the initiation of inflammatory pathways following HS/T but its cell-specific roles in this setting are not known. We assessed the importance of TLR4 on leukocytes of myeloid lineage and dendritic cells (DCs) to the early systemic inflammatory response following HS/T. Mice were subjected to HS/T and 20 inflammatory mediators were measured in plasma followed by Dynamic Bayesian Network (DBN) Analysis. Organ damage was assessed by histology and plasma ALT levels. The role of TLR4 was determined using TLR4-/-, MyD88-/-, and Trif-/- C57BL/6 (B6) mice, and by in vivo administration of a TLR4-specific neutralizing monoclonal antibody (mAb). The contribution of TLR4 expressed by myeloid leukocytes and DC was determined by generating cell-specific TLR4-/- B6 mice, including Lyz-Cre × TLR4loxP/loxP, and CD11c-Cre × TLR4loxP/loxP B6 mice. Adoptive transfer of bone marrow-derived TLR4+/+ or TLR4-/- DC into TLR4-/- mice confirmed the contribution of TLR4 on DC to the systemic inflammatory response after HS/T. Using both global knockout mice and the TLR4-blocking mAb 1A6 we established a central role for TLR4 in driving systemic inflammation. Using cell-selective TLR4-/- B6 mice, we found that TLR4 expression on both myeloid cells and CD11chigh DC is required for increases in systemic cytokine levels and organ damage after HS/T. We confirmed the capacity of TLR4 on CD11chigh DC to promote inflammation and liver damage using adoptive transfer of TLR4+/+ conventional (CD11chigh) DC into TLR4-/- mice. DBN inference identified CXC chemokines as proximal drivers of dynamic changes in the circulating levels of cytokines/chemokines after HS/T. TLR4 on DC was found to contribute selectively to the elevations in these proximal drivers. TLR4 on both myeloid cells and conventional DC is required for the initial systemic inflammation and organ damage in a mouse model of HS/T. This includes a role for TLR4 on DC in promoting increases in the early inflammatory networks identified in HS/T. These data establish DC along with macrophages as essential to the recognition of tissue damage and stress following tissue trauma with HS.

15.
J Biol Chem ; 291(29): 15093-107, 2016 07 15.
Article in English | MEDLINE | ID: mdl-27226571

ABSTRACT

The sensing of double-stranded RNA (dsRNA) in the liver is important for antiviral defenses but can also contribute to sterile inflammation during liver injury. Hepatocytes are often the target of viral infection and are easily injured by inflammatory insults. Here we sought to establish the pathways involved in the production of type I interferons (IFN-I) in response to extracellular poly(I:C), a dsRNA mimetic, in hepatocytes. This was of interest because hepatocytes are long-lived and, unlike most immune cells that readily die after activation with dsRNA, are not viewed as cells with robust antimicrobial capacity. We found that poly(I:C) leads to rapid up-regulation of inducible nitric oxide synthase (iNOS), double-stranded RNA-dependent protein kinase (PKR), and Src. The production of IFN-ß was dependent on iNOS, PKR, and Src and partially dependent on TLR3/Trif. iNOS and Src up-regulation was partially dependent on TLR3/Trif but entirely dependent on PKR. The phosphorylation of TLR3 on tyrosine 759 was shown to increase in parallel to IFN-ß production in an iNOS- and Src-dependent manner, and Src was found to directly interact with TLR3 in the endosomal compartment of poly(I:C)-treated cells. Furthermore, we identified a robust NO/cGMP/PKG-dependent feedforward pathway for the amplification of iNOS expression. These data identify iNOS/NO as an integral component of IFN-ß production in response to dsRNA in hepatocytes in a pathway that involves the coordinated activities of TLR3/Trif and PKR.


Subject(s)
Hepatocytes/immunology , Hepatocytes/metabolism , Interferon-beta/biosynthesis , Nitric Oxide Synthase Type II/metabolism , RNA, Double-Stranded/immunology , RNA, Double-Stranded/pharmacology , Toll-Like Receptor 3/metabolism , eIF-2 Kinase/metabolism , src-Family Kinases/metabolism , Adaptor Proteins, Vesicular Transport/deficiency , Adaptor Proteins, Vesicular Transport/genetics , Adaptor Proteins, Vesicular Transport/metabolism , Animals , Cells, Cultured , Endosomes/drug effects , Endosomes/metabolism , Hepatocytes/drug effects , Lipopolysaccharides/pharmacology , Mice , Mice, Inbred C57BL , Mice, Knockout , Models, Biological , Nitric Oxide Synthase Type II/deficiency , Nitric Oxide Synthase Type II/genetics , Phosphorylation/drug effects , Poly I-C/pharmacology , RNA Interference , Signal Transduction/drug effects , Toll-Like Receptor 3/deficiency , Toll-Like Receptor 3/genetics , Tyrosine/chemistry , Up-Regulation/drug effects , eIF-2 Kinase/deficiency , eIF-2 Kinase/genetics , src-Family Kinases/antagonists & inhibitors , src-Family Kinases/genetics
16.
Sci Signal ; 8(361): ra11, 2015 Jan 27.
Article in English | MEDLINE | ID: mdl-25628461

ABSTRACT

Proteolytic cleavage of the tumor necrosis factor (TNF) receptor (TNFR) from the cell surface contributes to anti-inflammatory responses and may be beneficial in reducing the excessive inflammation associated with multiple organ failure and mortality during sepsis. Using a clinically relevant mouse model of polymicrobial abdominal sepsis, we found that the production of inducible nitric oxide synthase (iNOS) in hepatocytes led to the cyclic guanosine monophosphate (cGMP)-dependent activation of the protease TACE (TNF-converting enzyme) and the shedding of TNFR. Furthermore, treating mice with a cGMP analog after the induction of sepsis increased TNFR shedding and decreased systemic inflammation. Similarly, increasing the abundance of cGMP with a clinically approved phosphodiesterase 5 inhibitor (sildenafil) also decreased markers of systemic inflammation, protected against organ injury, and increased circulating amounts of TNFR1 in mice with sepsis. We further confirmed that a similar iNOS-cGMP-TACE pathway was required for TNFR1 shedding by human hepatocytes in response to the bacterial product lipopolysaccharide. Our data suggest that increasing the bioavailability of cGMP might be beneficial in ameliorating the inflammation associated with sepsis.


Subject(s)
Coinfection/metabolism , Cyclic GMP/metabolism , Hepatocytes/metabolism , Receptors, Tumor Necrosis Factor/metabolism , Sepsis/metabolism , Signal Transduction/physiology , ADAM Proteins/metabolism , ADAM17 Protein , Analysis of Variance , Animals , Blotting, Western , Caspase 1/genetics , Enzyme Activation/physiology , Gentian Violet , Humans , Lipopolysaccharides , Mice , Mice, Inbred C57BL , Mice, Knockout , Microscopy, Fluorescence , Myeloid Differentiation Factor 88/genetics , Nitric Oxide Synthase Type II/metabolism , Proteolysis , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction
17.
J Immunol ; 193(5): 2405-15, 2014 Sep 01.
Article in English | MEDLINE | ID: mdl-25070845

ABSTRACT

Autophagy, an evolutionarily conserved homeostasis process regulating biomass quantity and quality, plays a critical role in the host response to sepsis. Recent studies show its calcium dependence, but the calcium-sensitive regulatory cascades have not been defined. In this study, we describe a novel mechanism in which calcium/calmodulin-dependent protein kinase IV (CaMKIV), through inhibitory serine phosphorylation of GSK-3ß and inhibition of FBXW7 recruitment, prevents ubiquitin proteosomal degradation of mammalian target of rapamycin (mTOR) and thereby augments autophagy in both the macrophage and the kidney. Under the conditions of sepsis studied, mTOR expression and activity were requisite for autophagy, a paradigm countering the current perspective that prototypically, mTOR inhibition induces autophagy. CaMKIV-mTOR-dependent autophagy was fundamentally important for IL-6 production in vitro and in vivo. Similar mechanisms were operant in the kidney during endotoxemia and served a cytoprotective role in mitigating acute kidney injury. Thus, CaMKIV-mTOR-dependent autophagy is conserved in both immune and nonimmune/parenchymal cells and is fundamental for the respective functional and adaptive responses to septic insult.


Subject(s)
Acute Kidney Injury/immunology , Autophagy/drug effects , Calcium-Calmodulin-Dependent Protein Kinase Type 4/immunology , Gene Expression Regulation/drug effects , Lipopolysaccharides/toxicity , Macrophages/immunology , TOR Serine-Threonine Kinases/immunology , Acute Kidney Injury/chemically induced , Acute Kidney Injury/genetics , Acute Kidney Injury/pathology , Animals , Autophagy/genetics , Autophagy/immunology , Calcium-Calmodulin-Dependent Protein Kinase Type 4/genetics , Cell Line , F-Box Proteins/genetics , F-Box Proteins/immunology , F-Box-WD Repeat-Containing Protein 7 , Gene Expression Regulation/immunology , Glycogen Synthase Kinase 3/genetics , Glycogen Synthase Kinase 3/immunology , Glycogen Synthase Kinase 3 beta , Inflammation/chemically induced , Inflammation/genetics , Inflammation/immunology , Interleukin-6/genetics , Interleukin-6/immunology , Macrophages/pathology , Mice , Mice, Knockout , TOR Serine-Threonine Kinases/genetics , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/immunology
18.
Circ Cardiovasc Genet ; 7(5): 615-24, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25049041

ABSTRACT

BACKGROUND: Growing evidence indicates that the presence of toll-like receptor 4 (TLR4) on platelets is a key regulator of platelet number and function. Platelets exposed to TLR4 agonists may serve to activate other cells such as neutrophils and endothelial cells in sepsis and other inflammatory conditions. The functional significance of platelet TLR4 in hemorrhagic shock (HS), however, remains unexplored. METHODS AND RESULTS: Using thromboelastography and platelet aggregometry, we demonstrate that platelet function is impaired in a mouse model of HS with resuscitation. Further analysis using cellular-specific TLR4 deletion in mice revealed that platelet TLR4 is essential for platelet activation and function in HS with resuscitation and that platelet TLR4 regulates the development of coagulopathy after hemorrhage and resuscitation. Transfusion of TLR4-negative platelets into mice resulted in protection from coagulopathy and restored platelet function. Additionally, platelet-specific TLR4 knockout mice were protected from lung and liver injury and exhibited a marked reduction in systemic inflammation as measured by circulating interleukin-6 after HS with resuscitation. CONCLUSIONS: We demonstrate for the first time that platelet TLR4 is an essential mediator of the inflammatory response as well as platelet activation and function in HS and resuscitation.


Subject(s)
Blood Coagulation Disorders/metabolism , Blood Platelets/metabolism , Gene Expression Regulation , Shock, Hemorrhagic/metabolism , Toll-Like Receptor 4/metabolism , Animals , Blood Coagulation Disorders/genetics , Blood Platelets/physiology , Endotoxemia , Gene Deletion , Hemostasis , Inflammation , Interleukin-6/blood , Macrophages/cytology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Peroxidase/metabolism , Platelet Aggregation , Platelet Function Tests , Resuscitation , Thrombelastography/methods
19.
Free Radic Biol Med ; 63: 361-7, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23722163

ABSTRACT

TRAF-interacting protein with a forkhead-associated domain (TIFA) is a tumor necrosis factor receptor-associated factor 6 (TRAF6) binding protein that mediates IL-1 signaling. We recently reported that TIFA mRNA is significantly upregulated early in the liver after trauma and hemorrhagic shock. In this study, we sought to characterize the upregulation of TIFA by hypoxia-reoxygenation and investigate its role in hypoxia-induced signaling. TIFA expression was detected by qRT-PCR and Western blotting in both mouse hemorrhagic shock with resuscitation (HS-R) and hepatocytes exposed to hypoxia-reoxygenation. Involvement of TLR4 and MyD88 was assessed using cells from TLR4(-/-) and MyD88(-/-) mice. The interaction of TIFA with TRAF6 and IRAK-1 was investigated using coimmunoprecipitation in vitro. RNAi was performed to knock down the endogenous expression of the TIFA gene in hepatocytes. High-mobility-group box 1 protein (HMGB1) expression was detected by Western blotting and ELISA, and the activation of NF-κB and MAPK was measured with EMSA and Western blotting. The results showed that TIFA expression was upregulated after HS-R in vivo and hypoxia-reoxygenation in vitro. Further analysis revealed that hypoxia-reoxygenation-induced upregulation of TIFA was TLR4- and MyD88-dependent. Moreover, TIFA was found to associate with TRAF6 constitutively, whereas its association with IRAK-1 was seen only after hypoxia-reoxygenation. Suppression of TIFA by siRNA reduced NF-κB activation and HMGB1 upregulation and release after hypoxia-reoxygenation. Taken together, these data suggest that TIFA is involved in the regulation of cell signaling in hypoxia-reoxygenation. The increase in TIFA level appears to be a feed-forward mechanism involved in TLR4/MyD88-dependent signaling, leading to NF-κB activation and HMGB1 release.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , HMGB1 Protein/metabolism , Hypoxia/metabolism , Myeloid Differentiation Factor 88/metabolism , Toll-Like Receptor 4/metabolism , Adaptor Proteins, Signal Transducing/genetics , Animals , Gene Expression Regulation , Hypoxia/pathology , Interleukin-1/genetics , Interleukin-1/metabolism , Mice , Shock, Hemorrhagic/metabolism , Shock, Hemorrhagic/pathology , Signal Transduction/genetics , Up-Regulation
20.
Shock ; 40(2): 115-21, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23656919

ABSTRACT

INTRODUCTION: Ischemia/reperfusion (I/R) of the liver contributes to the pathobiology of liver injury in transplantation, liver surgery, and hemorrhagic shock. Ischemia/reperfusion induces an inflammatory response that is driven, in part, by Toll-like receptor 4 (TLR) signaling. CD14 is known to participate in the function of TLR4. We hypothesized that CD14 would be involved in the pathobiology of warm hepatic I/R. METHODS: Using a 70% liver inflow inclusion model, CD14 knockout and wild-type (WT) mice were subjected to 1-h warm ischemia followed by reperfusion. CD14 mRNA, circulating transaminase, interleukin 6, soluble CD14, and high-mobility group box 1 (HMGB1) levels were measured. CD14 neutralizing antibody or isotype control antibody was given before ischemia or reperfusion for CD14 blockade in WT mice. Recombinant HMGB1 was given before reperfusion in some experiments to test if liver injury worsens. RESULTS: There was an upregulation of CD14 mRNA in reperfused livers together with increased soluble CD14 levels in the circulation. Compared with WT control mice, CD14 knockout mice had much lower alanine aminotransferase and interleukin 6 levels at 6 and 24 h following I/R, and much less liver necrosis by histology. TUNEL (terminal deoxynucleotidyl-transferase dUTP nick end labeling) staining displayed less apoptosis at 24 h in the absence of CD14. CD14 blockage by neutralizing antibody also attenuated liver injury and the inflammatory response in C57BL/6 mice following I/R, but did not provide additional protection to TLR4 mutant C3H/Hej mice. CD14 deficiency did not change circulating HMGB1 levels following I/R (6 h). A dose of recombinant HMGB1, which worsened hepatic injury when given before reperfusion in WT mice, did not increase liver damage in CD14-deficient mice. CONCLUSIONS: CD14 is actively involved in hepatic I/R injury. Its deficiency or blockade ischemia attenuates liver injury and inflammatory response. CD14 mediates liver damage and inflammatory responses in the setting of warm hepatic I/R in mice.


Subject(s)
Lipopolysaccharide Receptors/metabolism , Liver/metabolism , Liver/pathology , Reperfusion Injury/metabolism , Animals , Blotting, Western , Enzyme-Linked Immunosorbent Assay , HMGB1 Protein/blood , Interleukin-6/metabolism , Lipopolysaccharide Receptors/genetics , Mice , Mice, Knockout , Reperfusion Injury/blood , Reperfusion Injury/genetics , Reverse Transcriptase Polymerase Chain Reaction
SELECTION OF CITATIONS
SEARCH DETAIL
...