Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
bioRxiv ; 2024 May 05.
Article in English | MEDLINE | ID: mdl-38746216

ABSTRACT

Neutrophils (PMNs) reside as a marginated pool within the vasculature, ready for deployment during infection. However, how endothelial cells (ECs) control PMN extravasation and activation to strengthen tissue homeostasis remains ill-defined. Here, we found that the vascular ETS-related gene (ERG) is a generalized mechanism regulating PMN activity in preclinical tissue injury models and human patients. We show that ERG loss in ECs rewired PMN-transcriptome, enriched for genes associated with the CXCR2-CXCR4 signaling. Rewired PMNs compromise mice survival after pneumonia and induced lung vascular inflammatory injury following adoptive transfer into naïve mice, indicating their longevity and inflammatory activity memory. Mechanistically, EC-ERG restricted PMN extravasation and activation by upregulating the deubiquitinase A20 and downregulating the NFκB-IL8 cascade. Rescuing A20 in EC-Erg -/- endothelium or suppressing PMN-CXCR2 signaling rescued EC control of PMN activation. Findings deepen our understanding of EC control of PMN-mediated inflammation, offering potential avenues for targeting various inflammatory diseases. Highlights: ERG regulates trans-endothelial neutrophil (PMN) extravasation, retention, and activationLoss of endothelial (EC) ERG rewires PMN-transcriptomeAdopted transfer of rewired PMNs causes inflammation in a naïve mouse ERG transcribes A20 and suppresses CXCR2 function to inactivate PMNs. In brief/blurb: The authors investigated how vascular endothelial cells (EC) control polymorphonuclear neutrophil (PMN) extravasation, retention, and activation to strengthen tissue homeostasis. They showed that EC-ERG controls PMN transcriptome into an anti-adhesive and anti-inflammatory lineage by synthesizing A20 and suppressing PMNs-CXCR2 signaling, defining EC-ERG as a target for preventing neutrophilic inflammatory injury.

2.
bioRxiv ; 2023 Sep 23.
Article in English | MEDLINE | ID: mdl-37790514

ABSTRACT

IFNγ, a type II interferon secreted by immune cells, augments tissue responses to injury following pathogenic infections leading to lethal acute lung injury (ALI). Alveolar macrophages (AM) abundantly express Toll-like receptor-4 and represent the primary cell type of the innate immune system in the lungs. A fundamental question remains whether AM generation of IFNg leads to uncontrolled innate response and perpetuated lung injury. LPS induced a sustained increase in IFNg levels and unresolvable inflammatory lung injury in the mice lacking RGS2 but not in RGS2 null chimeric mice receiving WT bone marrow or receiving the RGS2 gene in AM. Thus, indicating RGS2 serves as a gatekeeper of IFNg levels in AM and thereby lung's innate immune response. RGS2 functioned by forming a complex with TLR4 shielding Gaq from inducing IFNg generation and AM inflammatory signaling. Thus, inhibition of Gaq blocked IFNg generation and subverted AM transcriptome from being inflammatory to reparative type in RGS2 null mice, resolving lung injury. Highlights: RGS2 levels are inversely correlated with IFNγ in ARDS patient's AM.RGS2 in alveolar macrophages regulate the inflammatory lung injury.During pathogenic insult RGS2 functioned by forming a complex with TLR4 shielding Gαq from inducing IFNγ generation and AM inflammatory signaling. eToc Blurb: Authors demonstrate an essential role of RGS2 in macrophages in airspace to promoting anti-inflammatory function of alveolar macrophages in lung injury. The authors provided new insight into the dynamic control of innate immune response by Gαq and RGS2 axis to prevent ALI.

3.
Arterioscler Thromb Vasc Biol ; 43(8): 1441-1454, 2023 08.
Article in English | MEDLINE | ID: mdl-37317855

ABSTRACT

BACKGROUND: Endothelial CLICs (chloride intracellular channel proteins) CLIC1 and CLIC4 are required for the GPCRs (G-protein-coupled receptors) S1PR1 (sphingosine-1-phosphate receptor 1) and S1PR3 to activate the small GTPases Rac1 (Ras-related C3 botulinum toxin substrate 1) and RhoA (Ras homolog family member A). To determine whether CLIC1 and CLIC4 function in additional endothelial GPCR pathways, we evaluated CLIC function in thrombin signaling via the thrombin-regulated PAR1 (protease-activated receptor 1) and downstream effector RhoA. METHODS: We assessed the ability of CLIC1 and CLIC4 to relocalize to cell membranes in response to thrombin in human umbilical vein endothelial cells (HUVEC). We examined CLIC1 and CLIC4 function in HUVEC by knocking down expression of each CLIC protein and compared thrombin-mediated RhoA or Rac1 activation, ERM (ezrin/radixin/moesin) phosphorylation, and endothelial barrier modulation in control and CLIC knockdown HUVEC. We generated a conditional murine allele of Clic4 and examined PAR1-mediated lung microvascular permeability and retinal angiogenesis in mice with endothelial-specific loss of Clic4. RESULTS: Thrombin promoted relocalization of CLIC4, but not CLIC1, to HUVEC membranes. Knockdown of CLIC4 in HUVEC reduced thrombin-mediated RhoA activation, ERM phosphorylation, and endothelial barrier disruption. Knockdown of CLIC1 did not reduce thrombin-mediated RhoA activity but prolonged the RhoA and endothelial barrier response to thrombin. Endothelial-specific deletion of Clic4 in mice reduced lung edema and microvascular permeability induced by PAR1 activating peptide. CONCLUSIONS: CLIC4 is a critical effector of endothelial PAR1 signaling and is required to regulate RhoA-mediated endothelial barrier disruption in cultured endothelial cells and murine lung endothelium. CLIC1 was not critical for thrombin-mediated barrier disruption but contributed to the barrier recovery phase after thrombin treatment.


Subject(s)
Receptor, PAR-1 , rhoA GTP-Binding Protein , Humans , Mice , Animals , Receptor, PAR-1/genetics , Receptor, PAR-1/metabolism , rhoA GTP-Binding Protein/metabolism , Thrombin/pharmacology , Thrombin/metabolism , Endothelium/metabolism , Human Umbilical Vein Endothelial Cells/metabolism , Cells, Cultured , Chloride Channels/genetics , Chloride Channels/metabolism , Mitochondrial Proteins/metabolism
4.
Front Pharmacol ; 13: 874197, 2022.
Article in English | MEDLINE | ID: mdl-36204227

ABSTRACT

Efficient phagocytosis of pathogens by the innate immune system during infectious injury is vital for restoring tissue integrity. Impaired phagocytosis, such as in the case of infection with Pseudomonas aeruginosa, a broad-spectrum antibiotic-resistant Gram-negative bacterium, can lead to a life threatening lung disorder, acute lung injury (ALI). Evidence indicates that loss of protease-activated receptor 2 (PAR2) impaired Pseudomonas aeruginosa clearance leading to non-resolvable ALI, but the mechanism remains unclear. Here, we focused on the alveolar macrophages (AMs), the predominant population of lung-resident macrophages involved in sensing bacteria, to understand their role in PAR2-mediated phagocytosis of Pseudomonas aeruginosa. We found that upon binding Pseudomonas aeruginosa, PAR2-expressing but not PAR2-null AMs had increased cAMP levels, which activated Rac1 through protein kinase A. Activated Rac1 increased actin-rich protrusions to augment the phagocytosis of Pseudomonas aeruginosa. Administration of liposomes containing constitutively active Rac1 into PAR2-null mice lungs rescued phagocytosis and enhanced the survival of PAR2-null mice from pneumonia. These studies showed that PAR2 drives the cAMP-Rac1 signaling cascade that activates Pseudomonas aeruginosa phagocytosis in AMs, thereby preventing death from bacterial pneumonia.

5.
Cells ; 11(18)2022 09 08.
Article in English | MEDLINE | ID: mdl-36139387

ABSTRACT

Cyclic GMP-AMP synthase (cGAS) is a predominant and ubiquitously expressed cytosolic onfirmedDNA sensor that activates innate immune responses by producing a second messenger, cyclic GMP-AMP (cGAMP), and the stimulator of interferon genes (STING). cGAS contains a highly disordered N-terminus, which can sense genomic/chromatin DNA, while the C terminal of cGAS binds dsDNA liberated from various sources, including mitochondria, pathogens, and dead cells. Furthermore, cGAS cellular localization dictates its response to foreign versus self-DNA. Recent evidence has also highlighted the importance of dsDNA-induced post-translational modifications of cGAS in modulating inflammatory responses. This review summarizes and analyzes cGAS activity regulation based on structure, sub-cellular localization, post-translational mechanisms, and Ca2+ signaling. We also discussed the role of cGAS activation in different diseases and clinical outcomes.


Subject(s)
Membrane Proteins , Nucleotidyltransferases , Chromatin , DNA/metabolism , Interferons/genetics , Membrane Proteins/metabolism , Nucleotidyltransferases/metabolism
6.
Am J Physiol Lung Cell Mol Physiol ; 321(4): L686-L702, 2021 10 01.
Article in English | MEDLINE | ID: mdl-34318714

ABSTRACT

Increased lung vascular permeability and neutrophilic inflammation are hallmarks of acute lung injury. Alveolar macrophages (AMϕ), the predominant sentinel cell type in the airspace, die in massive numbers while fending off pathogens. Recent studies indicate that the AMϕ pool is replenished by airspace-recruited monocytes, but the mechanisms instructing the conversion of recruited monocytes into reparative AMϕ remain elusive. Cyclic AMP (cAMP) is a vascular barrier protective and immunosuppressive second messenger in the lung. Here, we subjected mice expressing GFP under the control of the Lysozyme-M promoter (LysM-GFP mice) to the LPS model of rapidly resolving lung injury to address the impact of mechanisms determining cAMP levels in AMϕ and regulation of mobilization of the reparative AMϕ-pool. RNA-seq analysis of flow-sorted Mϕ identified phosphodiesterase 4b (PDE4b) as the top LPS-responsive cAMP-regulating gene. We observed that PDE4b expression markedly increased at the time of peak injury (4 h) and then decreased to below the basal level during the resolution phase (24 h). Activation of transcription factor NFATc2 was required for the transcription of PDE4b in Mϕ. Inhibition of PDE4 activity at the time of peak injury, using intratracheal rolipram, increased cAMP levels, augmented the reparative AMϕ pool, and resolved lung injury. This response was not seen following conditional depletion of monocytes, thus establishing airspace-recruited PDE4b-sensitive monocytes as the source of reparative AMϕ. Interestingly, adoptive transfer of rolipram-educated AMϕ into injured mice resolved lung edema. We propose suppression of PDE4b as an effective approach to promote reparative AMϕ generation from monocytes for lung repair.


Subject(s)
Acute Lung Injury/pathology , Cyclic AMP/metabolism , Cyclic Nucleotide Phosphodiesterases, Type 4/metabolism , Macrophages, Alveolar/cytology , Monocytes/cytology , NFATC Transcription Factors/metabolism , Adoptive Transfer/methods , Animals , Capillary Permeability/physiology , Cell Differentiation/physiology , Cyclic Nucleotide Phosphodiesterases, Type 4/genetics , Female , Inflammation , Lipopolysaccharides/pharmacology , Macrophages, Alveolar/transplantation , Male , Mice , Mice, Inbred C57BL , Neutrophils/immunology , Phosphodiesterase 4 Inhibitors/pharmacology , Rolipram/pharmacology , Transcriptional Activation/genetics
7.
Inflamm Res ; 70(7): 743-747, 2021 Jul.
Article in English | MEDLINE | ID: mdl-34185111

ABSTRACT

OBJECTIVE: Isoproterenol (ISO) is widely used agent to study the effects of interventions which could prevent or attenuate the development of myocardial infarction. The sequence of pathological event's revealed that increased myocardial tissue oxygen demand and energy dysregulation exist early during Iso-induced cardiac toxicity. Later, tissue hypoxia results in increased oxidative stress, inflammation and fibrosis along with cardiac dysfunction in this model. The canonical Wnt/ß-catenin pathway has been reported to directly implicate in inducing cardiomyocyte hypertrophy and remodelling. However, less is known about the role of non-canonical Wnt signalling in cardiac diseases. METHOD: Certain evidences have suggested that the activation of Wnt could up-regulate key energy sensor and cell growth regulator mTOR (Mechanistic target of rapamycin) by inhibition of GSK-3ß mediator. RESULT: The GSK-3ß could negatively influence the mTOR activity and produce energy dysregulation during stress or hypoxic conditions. This suggests that the inhibition of GSK-3ß by Wnt signalling could up-regulate mTOR levels and thereby restore early myocardial tissue energy balance and prevent cardiac toxicity in rodents. CONCLUSION: We hereby discuss a novel therapeutic role of the ß-catenin independent, Wnt-GSK3-mTOR axis in attenuation of Iso-induced cardiotoxicity in rodents.


Subject(s)
Adrenergic beta-Agonists , Cardiotoxicity/metabolism , Energy Metabolism , Glycogen Synthase Kinase 3 beta/metabolism , Isoproterenol , TOR Serine-Threonine Kinases/metabolism , Wnt Signaling Pathway , Animals , Disease Models, Animal , beta Catenin
8.
Front Immunol ; 11: 2091, 2020.
Article in English | MEDLINE | ID: mdl-33072072

ABSTRACT

Macrophages play a central role in dictating the tissue response to infection and orchestrating subsequent repair of the damage. In this context, macrophages residing in the lungs continuously sense and discriminate among a wide range of insults to initiate the immune responses important to host-defense. Inflammatory tissue injury also leads to activation of proteases, and thereby the coagulation pathway, to optimize injury and repair post-infection. However, long-lasting inflammatory triggers from macrophages can impair the lung's ability to recover from severe injury, leading to increased lung vascular permeability and neutrophilic injury, hallmarks of Acute Lung Injury (ALI). In this review, we discuss the roles of toll-like receptor 4 (TLR4) and protease activating receptor 2 (PAR2) expressed on the macrophage cell-surface in regulating lung vascular inflammatory signaling.


Subject(s)
Acute Lung Injury/immunology , Blood Vessels/immunology , Lung/immunology , Macrophages/immunology , Receptor, PAR-2/immunology , Signal Transduction/immunology , Toll-Like Receptor 4/immunology , Acute Lung Injury/pathology , Animals , Blood Vessels/injuries , Blood Vessels/pathology , Capillary Permeability/immunology , Humans , Lung/blood supply , Macrophages/pathology
9.
FASEB J ; 34(9): 12805-12819, 2020 09.
Article in English | MEDLINE | ID: mdl-32772419

ABSTRACT

Increased endothelial permeability leads to excessive exudation of plasma proteins and leukocytes in the interstitium, which characterizes several vascular diseases including acute lung injury. The myosin light chain kinase long (MYLK-L) isoform is canonically known to regulate the endothelial permeability by phosphorylating myosin light chain (MLC-P). Compared to the short MYLK isoform, MYLK-L contains an additional stretch of ~919 amino acid at the N-terminus of unknown function. We show that thapsigargin and thrombin-induced SOCE was markedly reduced in Mylk-L-/- endothelial cells (EC) or MYLK-L-depleted human EC. These agonists also failed to increase endothelial permeability in MYLK-L-depleted EC and Mylk-L-/- lungs, thus demonstrating the novel role of MYLK-L-induced SOCE in increasing vascular permeability. MYLK-L augmented SOCE by increasing endoplasmic reticulum (ER)-plasma membrane (PM) junctions and STIM1 translocation to these junctions. Transduction of N-MYLK domain (amino acids 1-919 devoid of catalytic activity) into Mylk-L-/- EC rescued SOCE to the level seen in control EC in a STIM1-dependent manner. N-MYLK-induced SOCE augmented endothelial permeability without MLC-P via an actin-binding motif, DVRGLL. Liposomal-mediated delivery of N-MYLK mutant but not ∆DVRGLL-N-MYLK mutant in Mylk-L-/- mice rescued vascular permeability increase in response to endotoxin, indicating that targeting of DVRGLL motif within MYLK-L may limit SOCE-induced vascular hyperpermeability.


Subject(s)
Calcium-Binding Proteins/metabolism , Capillary Permeability , Cell Membrane/enzymology , Endoplasmic Reticulum/enzymology , Myosin-Light-Chain Kinase/metabolism , Acute Lung Injury/metabolism , Animals , Human Umbilical Vein Endothelial Cells , Humans , Isoenzymes/metabolism , Lung/metabolism , Lung/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Neoplasm Proteins/metabolism , Stromal Interaction Molecule 1/metabolism
10.
Cell Rep ; 27(3): 793-805.e4, 2019 04 16.
Article in English | MEDLINE | ID: mdl-30995477

ABSTRACT

Alveolar macrophages (AMs), upon sensing pathogens, trigger host defense by activating toll-like receptor 4 (TLR4), but the counterbalancing mechanisms that deactivate AM inflammatory signaling and prevent lethal edema, the hallmark of acute lung injury (ALI), remain unknown. Here, we demonstrate the essential role of AM protease-activating receptor 2 (PAR2) in rapidly suppressing inflammation to prevent long-lasting injury. We show that thrombin, released during TLR4-induced lung injury, directly activates PAR2 to generate cAMP, which abolishes Ca2+ entry through the TRPV4 channel. Deletion of PAR2 and thus the accompanying cAMP generation augments Ca2+ entry via TRPV4, causing sustained activation of the transcription factor NFAT to produce long-lasting TLR4-mediated inflammatory lung injury. Rescuing thrombin-sensitive PAR2 expression or blocking TRPV4 activity in PAR2-null AMs restores their capacity to resolve inflammation and reverse lung injury. Thus, activation of the thrombin-induced PAR2-cAMP cascade in AMs suppresses TLR4 inflammatory signaling to reinstate tissue integrity.


Subject(s)
Calcium Signaling , Cyclic AMP/metabolism , Inflammation/prevention & control , Macrophages, Alveolar/metabolism , Receptor, PAR-2/metabolism , TRPV Cation Channels/metabolism , Toll-Like Receptor 4/metabolism , Acute Lung Injury/metabolism , Acute Lung Injury/pathology , Animals , Calcium/metabolism , Cytokines/metabolism , Disease Models, Animal , Inflammation/metabolism , Lipopolysaccharides/toxicity , Macrophages, Alveolar/cytology , Macrophages, Alveolar/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , NFATC Transcription Factors/metabolism , RNA Interference , RNA, Small Interfering/metabolism , Receptor, PAR-2/deficiency , Receptor, PAR-2/genetics , TRPV Cation Channels/antagonists & inhibitors , TRPV Cation Channels/genetics , Thrombin/metabolism
11.
Eur J Pharmacol ; 765: 406-14, 2015 Oct 15.
Article in English | MEDLINE | ID: mdl-26341014

ABSTRACT

Stress and stress related disorders are a major cause of morbidity and mortality and understanding stress mechanisms is of great importance for devising appropriate therapeutic measures in such situations. The brain and its complex neurotransmitter systems regulate physiological and behavioral responses to a variety of stressors. Several other factors like age, gender and emotionality of the organism, as well as type, intensity and duration of the stressor may decide the nature and extent of stress effects. Nitric oxide (NO) is widely distributed in the brain and its role in Central nervous system (CNS) pathophysiology has been suggested. Recent studies have shown that free radicals and in particular NO may play a crucial role in the regulation of stress effects. All the various factors, mentioned above, that might influence stress responsiveness have been discussed with reference to regulatory role of NO during stress and it appears that NO may act as a therapeutic target for development of novel strategies against stress related disorders.


Subject(s)
Brain/metabolism , Nitric Oxide/metabolism , Oxidative Stress/physiology , Stress, Physiological/physiology , Animals , Free Radicals/metabolism , Humans , Reactive Oxygen Species/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...