Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 122
Filter
Add more filters

Publication year range
1.
Int J Mol Sci ; 24(3)2023 Jan 21.
Article in English | MEDLINE | ID: mdl-36768463

ABSTRACT

Diabetes mellitus is a chronic disease characterized by metabolic dysregulation which is frequently associated with diabetic foot ulcers that result from a severely compromised innate immune system. The high levels of blood glucose characteristic of diabetes cause an increase in circulating inflammatory mediators, which accelerate cellular senescence and dampen antimicrobial activity within dermal tissue. In diabetic wounds, bacteria and fungi proliferate in a protective biofilm forming a structure that a compromised host defense system cannot easily penetrate, often resulting in chronic infections that require antimicrobial intervention to promote the healing process. The designed host defense peptide (dHDP) RP557 is a synthesized peptide whose sequence has been derived from naturally occurring antimicrobial peptides (AMPs) that provide the first line of defense against invading pathogens. AMPs possess an amphipathic α-helix or ß-sheet structure and a net positive charge that enables them to incorporate into pathogen membranes and perturb the barrier function of Gram-positive and Gram-negative bacteria along with fungi. The capacity of skin to resist infections is largely dependent upon the activity of endogenous AMPs that provided the basis for the design and testing of RP557 for the resolution of wound infections. In the current study, the topical application of RP557 stopped bacterial growth in the biofilm of methicillin-resistant Staphylococcus aureus (MRSA) USA300 infected wounds on the flanks of clinically relevant diabetic TALLYHO mice. Topical application of RP557 reduced bacterial load and accelerated wound closure, while wound size in control diabetic mice continued to expand. These studies demonstrate that RP557 reduces or eliminates an infection in its biofilm and restores wound-healing capacity.


Subject(s)
Anti-Bacterial Agents , Antimicrobial Cationic Peptides , Diabetes Mellitus, Experimental , Methicillin-Resistant Staphylococcus aureus , Wound Infection , Animals , Mice , Anti-Bacterial Agents/administration & dosage , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/therapeutic use , Antimicrobial Cationic Peptides/administration & dosage , Antimicrobial Cationic Peptides/pharmacology , Antimicrobial Cationic Peptides/therapeutic use , Diabetes Mellitus, Experimental/complications , Diabetes Mellitus, Experimental/drug therapy , Gram-Negative Bacteria , Gram-Positive Bacteria , Methicillin-Resistant Staphylococcus aureus/drug effects , Wound Infection/drug therapy , Administration, Topical
2.
J Immunol ; 205(10): 2806-2820, 2020 11 15.
Article in English | MEDLINE | ID: mdl-33055281

ABSTRACT

Intermediate monocytes (iMo; CD14+CD16+) increase in number in the circulation of patients with unstable coronary artery disease (CAD), and their recruitment to inflamed arteries is implicated in events leading to mortality following MI. Monocyte recruitment to inflamed coronary arteries is initiated by high affinity ß2-integrin (CD11c/CD18) that activates ß1-integrin (VLA-4) to bind endothelial VCAM-1. How integrin binding under shear stress mechanosignals a functional shift in iMo toward an inflammatory phenotype associated with CAD progression is unknown. Whole blood samples from patients treated for symptomatic CAD including non-ST elevation MI, along with healthy age-matched subjects, were collected to assess chemokine and integrin receptor levels on monocytes. Recruitment on inflamed human aortic endothelium or rVCAM-1 under fluid shear stress was assessed using a microfluidic-based artery on a chip (A-Chip). Membrane upregulation of high affinity CD11c correlated with concomitant activation of VLA-4 within focal adhesive contacts was required for arrest and diapedesis across inflamed arterial endothelium to a greater extent in non-ST elevation MI compared with stable CAD patients. The subsequent conversion of CD11c from a high to low affinity state under fluid shear activated phospho-Syk- and ADAM17-mediated proteolytic cleavage of CD16. This marked the conversion of iMo to an inflammatory phenotype associated with nuclear translocation of NF-κB and production of IL-1ß+ We conclude that CD11c functions as a mechanoregulator that activates an inflammatory state preferentially in a majority of iMo from cardiac patients but not healthy patients.


Subject(s)
CD11c Antigen/metabolism , Coronary Artery Disease/immunology , Endothelium, Vascular/immunology , Monocytes/immunology , Non-ST Elevated Myocardial Infarction/immunology , Adult , Aged , Allosteric Regulation/immunology , Aorta/cytology , Case-Control Studies , Cell Culture Techniques , Cell Line , Cell Membrane/metabolism , Coronary Artery Disease/blood , Coronary Artery Disease/surgery , Coronary Vessels/cytology , Coronary Vessels/immunology , Endothelial Cells/cytology , Endothelial Cells/immunology , Endothelial Cells/metabolism , Endothelium, Vascular/metabolism , Female , Humans , Integrin alpha4beta1/metabolism , Lab-On-A-Chip Devices , Male , Microfluidic Analytical Techniques/instrumentation , Middle Aged , Non-ST Elevated Myocardial Infarction/blood , Non-ST Elevated Myocardial Infarction/surgery , Percutaneous Coronary Intervention , Recombinant Proteins/immunology , Recombinant Proteins/metabolism , Transendothelial and Transepithelial Migration/immunology , Vascular Cell Adhesion Molecule-1/immunology , Vascular Cell Adhesion Molecule-1/metabolism
3.
Proc Natl Acad Sci U S A ; 116(22): 10917-10926, 2019 05 28.
Article in English | MEDLINE | ID: mdl-31088972

ABSTRACT

T cell cytokines contribute to immunity against Staphylococcus aureus, but the predominant T cell subsets involved are unclear. In an S. aureus skin infection mouse model, we found that the IL-17 response was mediated by γδ T cells, which trafficked from lymph nodes to the infected skin to induce neutrophil recruitment, proinflammatory cytokines IL-1α, IL-1ß, and TNF, and host defense peptides. RNA-seq for TRG and TRD sequences in lymph nodes and skin revealed a single clonotypic expansion of the encoded complementarity-determining region 3 amino acid sequence, which could be generated by canonical nucleotide sequences of TRGV5 or TRGV6 and TRDV4 However, only TRGV6 and TRDV4 but not TRGV5 sequences expanded. Finally, Vγ6+ T cells were a predominant γδ T cell subset that produced IL-17A as well as IL-22, TNF, and IFNγ, indicating a broad and substantial role for clonal Vγ6+Vδ4+ T cells in immunity against S. aureus skin infections.


Subject(s)
Interleukin-17/physiology , Staphylococcal Infections/immunology , Staphylococcus aureus/pathogenicity , T-Lymphocyte Subsets/immunology , T-Lymphocytes/immunology , Animals , Disease Models, Animal , Humans , Lymph Nodes/immunology , Mice , Staphylococcal Infections/microbiology
4.
Arterioscler Thromb Vasc Biol ; 40(1): 72-85, 2020 01.
Article in English | MEDLINE | ID: mdl-31619061

ABSTRACT

OBJECTIVE: A Mediterranean diet supplemented with olive oil and nuts prevents cardiovascular disease in clinical studies, but the underlying mechanisms are incompletely understood. We investigated whether the preventive effect of the diet could be due to inhibition of atherosclerosis and foamy monocyte formation in Ldlr-/- mice fed with a diet in which milkfat in a Western diet (WD) was replaced with extra-virgin olive oil and nuts (EVOND). Approach and Results: Ldlr-/- mice were fed EVOND or a Western diet for 3 (or 6) months. Compared with the Western diet, EVOND decreased triglyceride and cholesterol levels but increased unsaturated fatty acid concentrations in plasma. EVOND also lowered intracellular lipid accumulation in circulating monocytes, indicating less formation of foamy monocytes, compared with the Western diet. In addition, compared with the Western diet, EVOND reduced monocyte expression of inflammatory cytokines, CD36, and CD11c, with decreased monocyte uptake of oxLDL (oxidized LDL [low-density lipoprotein]) ex vivo and reduced CD11c+ foamy monocyte firm arrest on vascular cell adhesion molecule-1 and E-selectin-coated slides in an ex vivo shear flow assay. Along with these changes, EVOND compared with the Western diet reduced the number of CD11c+ macrophages in atherosclerotic lesions and lowered atherosclerotic lesion area of the whole aorta and aortic sinus. CONCLUSIONS: A diet enriched in extra-virgin olive oil and nuts, compared with a Western diet high in saturated fat, lowered plasma cholesterol and triglyceride levels, inhibited foamy monocyte formation, inflammation, and adhesion, and reduced atherosclerosis in Ldlr-/- mice.


Subject(s)
Atherosclerosis/diet therapy , Diet, Western , Dietary Fats, Unsaturated/pharmacology , Fatty Acids/adverse effects , Lipid Metabolism/physiology , Monocytes/metabolism , Animals , Atherosclerosis/metabolism , Atherosclerosis/pathology , Disease Models, Animal , Lipoproteins, LDL/metabolism , Male , Mice , Monocytes/pathology
5.
J Mol Cell Cardiol ; 140: 30-41, 2020 03.
Article in English | MEDLINE | ID: mdl-32087218

ABSTRACT

AIMS: Mammalian target of rapamycin (mTOR) inhibitors used in drug-eluting stents (DES) to control restenosis have been found to delay endothelialization and increase incidence of late-stent thrombosis through mechanisms not completely understood. We revealed that mTOR inhibition (mTORi) upregulated the expression of cell growth suppressor IRF-1 in primary human arterial endothelial cells (HAEC). This study aimed to examine how mTOR-regulated IRF-1 expression contributes to the suppressive effect of mTORi on arterial endothelial proliferation. METHODS AND RESULTS: Western blotting, quantitative PCR, and a dual-luciferase reporter assay indicated that mTOR inhibitors rapamycin and torin 1 upregulated IRF-1 expression and increased its transcriptional activity. IRF-1 in turn contributed to the suppressive effect of mTORi by mediating HAEC apoptosis and cell cycle arrest in part through upregulation of caspase 1 and downregulation of cyclin D3, as revealed by CCK-8 assay, Annexin V binding assay, measurement of activated caspase 3, BrdU incorporation assay, and matrigel tube formation assay. In a mouse model of femoral artery wire injury, administration of rapamycin inhibited EC recovery, an effect alleviated by EC deficiency of IRF-1. Chromatin immunoprecipitation assay with HAEC and rescue expression of wild type or dominant-negative IRF-1 in EC isolated from Irf1-/- mice confirmed transcriptional regulation of IRF-1 on the expression of CASP1 and CCND3. Furthermore, mTORi activated multiple PKC members, among which PKCζ was responsible for the growth-inhibitory effect on HAEC. Activated PKCζ increased IRF1 transcription through JAK/STAT-1 and NF-κB signaling. Finally, overexpression of wild type or mutant raptor incapable of binding mTOR indicated that mTOR-free raptor contributed to PKCζ activation in mTOR-inhibited HAEC. CONCLUSIONS: The study reveals an IRF-1-mediated mechanism that contributes to the suppressive effects of mTORi on HAEC proliferation. Further study may facilitate the development of effective strategies to reduce the side effects of DES used in coronary interventions.


Subject(s)
Endothelial Cells/metabolism , Endothelium, Vascular/metabolism , Interferon Regulatory Factor-1/metabolism , TOR Serine-Threonine Kinases/metabolism , Animals , Apoptosis/drug effects , Apoptosis/genetics , Cell Cycle Checkpoints/drug effects , Cell Cycle Checkpoints/genetics , Cell Proliferation/drug effects , Cell Proliferation/genetics , Cells, Cultured , Femoral Artery/injuries , Humans , Interferon Regulatory Factor-1/genetics , Mice , Mice, Knockout , Naphthyridines/pharmacology , Signal Transduction/drug effects , Signal Transduction/genetics , Sirolimus/pharmacology , TOR Serine-Threonine Kinases/antagonists & inhibitors , TOR Serine-Threonine Kinases/genetics , Transfection
6.
FASEB J ; 33(11): 12888-12899, 2019 11.
Article in English | MEDLINE | ID: mdl-31499005

ABSTRACT

Endothelial up-regulation of VCAM-1 at susceptible sites in arteries modulates the recruitment efficiency of inflammatory monocytes that initiates atherosclerotic lesion formation. We reported that hydrodynamic shear stress (SS) mechanoregulates inflammation in human aortic endothelial cells through endoplasmic reticulum (ER) stress via activation of the transcription factor x-box binding protein 1 (XBP1). Here, a microfluidic flow channel that produces a linear gradient of SS along a continuous monolayer of endothelium was used to delve the mechanisms underlying transcriptional regulation of TNF-α-stimulated VCAM-1 expression. High-resolution immunofluorescence imaging enabled continuous detection of platelet endothelial cell adhesion molecule 1 (PECAM-1)-dependent, outside-in signaling as a function of SS magnitude. Differential expression of VCAM-1 and intercellular adhesion molecule 1 (ICAM-1) was regulated by the spatiotemporal activation of MAPKs, ER stress markers, and transcription factors, which was dependent on the mechanosensing of SS through PECAM-1 and PI3K. Inhibition of p38 specifically abrogated the rise to peak VCAM-1 at low SS (2 dyn/cm2), whereas inhibition of ERK1/2 attenuated peak ICAM-1 at high SS (12 dyn/cm2). A shear stress-regulated temporal rise in p38 phosphorylation activated the nuclear translocation of XBP1, which together with the transcription factor IFN regulatory factor 1, promoted maximum VCAM-1 expression. These data reveal a mechanism by which SS sensitizes the endothelium to a cytokine-induced ER stress response to spatially regulate inflammation promoting atherosclerosis.-Bailey, K. A., Moreno, E., Haj, F. G., Simon, S. I., Passerini, A. G. Mechanoregulation of p38 activity enhances endoplasmic reticulum stress-mediated inflammation by arterial endothelium.


Subject(s)
Arteries/pathology , Endoplasmic Reticulum Stress , Endothelium, Vascular/pathology , p38 Mitogen-Activated Protein Kinases/metabolism , Arteries/enzymology , Cell Nucleus/metabolism , Cells, Cultured , Endothelium, Vascular/enzymology , Female , Humans , Interferon Regulatory Factor-1/metabolism , Male , Phosphorylation , Transcription, Genetic , Vascular Cell Adhesion Molecule-1/genetics
7.
Am J Physiol Heart Circ Physiol ; 316(2): H408-H420, 2019 02 01.
Article in English | MEDLINE | ID: mdl-30462552

ABSTRACT

Increased expression of vascular cell adhesion molecule 1 (VCAM-1) on the aortic endothelium is an early marker of atherogenesis, promoted in part by elevated levels of inflammatory cytokines such as TNF-α. Mammalian target of rapamycin (mTOR) is a ubiquitous signaling molecule that has been considered to contribute to diverse cellular processes through mTOR complex 1 (mTORC1) or complex 2 (mTORC2). This study aimed to elucidate the role of mTOR signaling in TNF-α-induced VCAM-1 expression by the arterial endothelium. Primary human aortic endothelial cells (HAECs) were treated with low-dose (0.1 ng/ml) TNF-α, and VCAM-1 expression was measured by real-time quantitative PCR, Western blot analysis, and flow cytometry. Inhibition of mTOR through siRNA-mediated depletion or treatment with chemical inhibitors rapamycin or torin 1 suppressed VCAM1 transcription, which translated to inhibition of VCAM-1 surface expression by HAECs and concomitant decreased adhesion of monocytes. A promoter luciferase assay and chromatin immunoprecipitation indicated that mTOR regulated VCAM1 transcription through a mechanism involving transcription factor GATA6. Activation of PKC-α and an increase in miR-200a-3p expression, caused by mTOR inhibition but not disruption of mTORC1 or mTORC2 singly or together, decreased TNF-α-induced GATA6 expression and its enrichment at the VCAM1 promoter. In conclusion, mTOR inhibition activates PKC-α independently of disruption of mTORC1 and/or mTORC2, which challenges the conventional wisdom regarding mTOR signaling. Moreover, mTOR signals through transcriptional and posttranscriptional mechanisms to elicit maximal cytokine-induced endothelial inflammation that precedes atherosclerosis. NEW & NOTEWORTHY Both mammalian target of rapamycin (mTOR) complex 1 (mTORC1) and mTORC2 contribute to PKC-α activation in the human aortic endothelium. Inhibition of mTOR is not equivalent to disruption of mTORC1 and/or mTORC2 in affecting human aortic endothelial cell signaling. Specifically, inhibition of mTOR causes PKC-α activation and miR-200a-3p upregulation, which independently suppresses TNF-α-induced transcription factor GATA6 expression and subsequently inhibits VCAM-1 expression and monocytic cell adhesion onto the aortic endothelium.


Subject(s)
Atherosclerosis/metabolism , Cell Adhesion , Endothelial Cells/metabolism , GATA6 Transcription Factor/metabolism , TOR Serine-Threonine Kinases/metabolism , Vascular Cell Adhesion Molecule-1/metabolism , Aorta/cytology , Aorta/metabolism , Cells, Cultured , Down-Regulation , Endothelial Cells/drug effects , Endothelial Cells/physiology , Endothelium, Vascular/cytology , Endothelium, Vascular/metabolism , GATA6 Transcription Factor/genetics , Humans , Monocytes/physiology , Protein Kinase C-alpha/metabolism , Signal Transduction , TOR Serine-Threonine Kinases/genetics , Tumor Necrosis Factor-alpha/pharmacology , Vascular Cell Adhesion Molecule-1/genetics
8.
Blood ; 130(19): 2101-2110, 2017 11 09.
Article in English | MEDLINE | ID: mdl-28811304

ABSTRACT

E-selectin extends from the plasma membrane of inflamed endothelium and serves to capture leukocytes from flowing blood via long-lived catch-bonds that support slow leukocyte rolling under shear stress. Its ligands are glycosylated with the tetrasaccharide sialyl Lewisx (sLex), which contributes to bond affinity and specificity. E-selectin-mediated rolling transmits signals into neutrophils that trigger activation of high-affinity ß2-integrins necessary for transition to shear-resistant adhesion and transendothelial migration. Rivipansel is a glycomimetic drug that inhibits E-selectin-mediated vaso-occlusion induced by integrin-dependent sickle-red blood cell-leukocyte adhesion. How Rivipansel antagonizes ligand recognition by E-selectin and blocks outside-in signaling of integrin-mediated neutrophil arrest while maintaining rolling immune-surveillance is unknown. Here, we demonstrate that sLex expressed on human L-selectin is preferentially bound by E-selectin and, on ligation, initiates secretion of MRP8/14 that binds TLR4 to elicit the extension of ß2-integrin to an intermediate affinity state. Neutrophil rolling over E-selectin at precise shear stress transmits tension and catch-bond formation with L-selectin via sLex, resulting in focal clusters that deliver a distinct signal to upshift ß2-integrins to a high-affinity state. Rivipansel effectively blocked formation of selectin catch-bonds, revealing a novel mechanotransduction circuit that rapidly converts extended ß2-integrins to high-affinity shear-resistant bond clusters with intracellular adhesion molecule 1 on inflamed endothelium.


Subject(s)
Anemia, Sickle Cell/metabolism , CD18 Antigens/metabolism , Cell Migration Inhibition , E-Selectin/metabolism , Endothelium, Vascular/metabolism , L-Selectin/metabolism , Leukocyte Rolling , Mechanotransduction, Cellular , Neutrophils/metabolism , Transendothelial and Transepithelial Migration , ATP-Binding Cassette Transporters/metabolism , Adult , Anemia, Sickle Cell/drug therapy , Anemia, Sickle Cell/pathology , Calgranulin B/metabolism , Cell Adhesion/drug effects , Endothelium, Vascular/pathology , Female , Humans , Male , Neutrophils/pathology , Oligosaccharides/metabolism , Shear Strength , Sialyl Lewis X Antigen , Toll-Like Receptor 4/metabolism
9.
J Immunol ; 199(5): 1772-1782, 2017 09 01.
Article in English | MEDLINE | ID: mdl-28733486

ABSTRACT

The immune response to Staphylococcus aureus infection in skin involves the recruitment of polymorphonuclear neutrophils (PMNs) from the bone marrow via the circulation and local granulopoiesis from hematopoietic stem and progenitor cells (HSPCs) that also traffic to infected skin wounds. We focus on regulation of PMN number and function and the role of pore-forming α-toxin (AT), a virulence factor that causes host cell lysis and elicits inflammasome-mediated IL-1ß secretion in wounds. Infection with wild-type S. aureus enriched in AT reduced PMN recruitment and resulted in sustained bacterial burden and delayed wound healing. In contrast, PMN recruitment to wounds infected with an isogenic AT-deficient S. aureus strain was unimpeded, exhibiting efficient bacterial clearance and hastened wound resolution. HSPCs recruited to infected wounds were unaffected by AT production and were activated to expand PMN numbers in proportion to S. aureus abundance in a manner regulated by TLR2 and IL-1R signaling. Immunodeficient MyD88-knockout mice infected with S. aureus experienced lethal sepsis that was reversed by PMN expansion mediated by injection of wild-type HSPCs directly into wounds. We conclude that AT-induced IL-1ß promotes local granulopoiesis and effective resolution of S. aureus-infected wounds, revealing a potential antibiotic-free strategy for tuning the innate immune response to treat methicillin-resistant S. aureus infection in immunodeficient patients.


Subject(s)
Bacterial Toxins/immunology , Granulocytes/immunology , Hematopoietic Stem Cells/physiology , Hemolysin Proteins/immunology , Staphylococcal Infections/immunology , Staphylococcus aureus/physiology , Virulence Factors/immunology , Wound Infection/immunology , Animals , Bacterial Load , Bacterial Toxins/genetics , Cell Differentiation , Cell Proliferation , Granulocytes/microbiology , Hemolysin Proteins/genetics , Immunomodulation , Mice , Mice, Inbred C57BL , Mice, Knockout , Mutation/genetics , Myeloid Differentiation Factor 88/genetics , Receptors, Interleukin-1/metabolism , Signal Transduction , Toll-Like Receptor 2/metabolism , Virulence Factors/genetics
10.
Blood ; 127(3): 314-24, 2016 Jan 21.
Article in English | MEDLINE | ID: mdl-26468229

ABSTRACT

Chemokines are required for leukocyte recruitment and appropriate host defense and act through G protein-coupled receptors (GPCRs), which induce downstream signaling leading to integrin activation. Although the α and ß subunits of the GPCRs are the first intracellular molecules that transduce signals after ligand binding and are therefore indispensable for downstream signaling, relatively little is known about their contribution to lymphocyte function-associated antigen 1 (LFA-1) activation and leukocyte recruitment. We used knockout mice and short hairpin RNA to knock down guanine nucleotide binding protein (GNB) isoforms (GNB1, GNB2, GNB4, and GNB5) in HL60 cells and primary murine hematopoietic cells. Neutrophil function was assessed by using intravital microscopy, flow chamber assays, and chemotaxis and biochemistry studies. We unexpectedly discovered that all expressed GNB isoforms are required for LFA-1 activation. Their downregulation led to a significant impairment of LFA-1 activation, which was demonstrated in vitro and in vivo. Furthermore, we showed that GPCR activation leads to Ras-related C3 botulinum toxin substrate 1 (Rac1)-dependent activation of both phospholipase C ß2 (Plcß2) and Plcß3. They act nonredundantly to produce inositol triphosphate-mediated intracellular Ca(2+) flux and LFA-1 activation that support chemokine-induced arrest in vivo. In a complex inflammatory disease model, Plcß2-, Plcß3-, or Rac1-deficient mice were protected from lipopolysaccharide-induced lung injury. Taken together, we demonstrated that all Gnb isoforms are required for chemokine-induced downstream signaling, and Rac1, Plcß2, and Plcß3 are critically involved in integrin activation and leukocyte arrest.


Subject(s)
Cell Cycle Checkpoints , GTP-Binding Protein beta Subunits/metabolism , Lymphocyte Function-Associated Antigen-1/metabolism , Neutrophils/metabolism , Phospholipase C beta/metabolism , Signal Transduction , rac1 GTP-Binding Protein/metabolism , Animals , Antigens, Surface/genetics , Antigens, Surface/metabolism , Calcium/metabolism , Cell Cycle Checkpoints/drug effects , Cell Cycle Checkpoints/genetics , Cell Line , Chemokines/pharmacology , Chemotaxis/drug effects , Chemotaxis/genetics , Chemotaxis/immunology , Disease Models, Animal , Down-Regulation , GTP-Binding Protein alpha Subunit, Gi2/genetics , GTP-Binding Protein alpha Subunit, Gi2/metabolism , GTP-Binding Protein beta Subunits/genetics , Gene Expression Regulation , Gene Knockdown Techniques , Humans , Lipopolysaccharides/adverse effects , Mice , Models, Biological , Neutrophils/drug effects , Neutrophils/immunology , Phospholipase C beta/genetics , Pneumonia/chemically induced , Pneumonia/genetics , Pneumonia/immunology , Pneumonia/metabolism , Protein Binding , Protein Isoforms , RNA, Small Interfering/genetics , Receptors, G-Protein-Coupled/metabolism , rac1 GTP-Binding Protein/genetics
11.
Eur J Clin Invest ; 48 Suppl 2: e12964, 2018 Nov.
Article in English | MEDLINE | ID: mdl-29873837

ABSTRACT

The recruitment of neutrophils to sites of inflammation, their battle against invading microorganisms through phagocytosis and the release of antimicrobial agents is a highly coordinated and tightly regulated process that involves the interplay of many different receptors, ion channels and signalling pathways. Changes in intracellular calcium levels, caused by cytosolic Ca2+ store depletion and the influx of extracellular Ca2+ via ion channels, play a critical role in synchronizing neutrophil activation and function. In this review, we provide an overview of how Ca2+ signalling is initiated in neutrophils and how changes in intracellular Ca2+ levels modulate neutrophil function.


Subject(s)
Calcium Signaling/physiology , Neutrophil Infiltration/physiology , Animals , Calcium Channels/physiology , Humans , Ion Channels/physiology , Mice , Neutrophils/physiology , Potassium Channels/physiology , Receptors, Purinergic P2X/physiology , Sodium Channels/physiology , Transient Receptor Potential Channels/physiology
12.
Stem Cells ; 35(6): 1493-1504, 2017 06.
Article in English | MEDLINE | ID: mdl-28276602

ABSTRACT

Mesenchymal stem cell therapies promote wound healing by manipulating the local environment to enhance the function of host cells. Aggregation of mesenchymal stem cells (MSCs) into three-dimensional spheroids increases cell survival and augments their anti-inflammatory and proangiogenic potential, yet there is no consensus on the preferred conditions for maximizing spheroid function in this application. The objective of this study was to optimize conditions for forming MSC spheroids that simultaneously enhance their anti-inflammatory and proangiogenic nature. We applied a design of experiments (DOE) approach to determine the interaction between three input variables (number of cells per spheroid, oxygen tension, and inflammatory stimulus) on MSC spheroids by quantifying secretion of prostaglandin E2 (PGE2 ) and vascular endothelial growth factor (VEGF), two potent molecules in the MSC secretome. DOE results revealed that MSC spheroids formed with 40,000 cells per spheroid in 1% oxygen with an inflammatory stimulus (Spheroid 1) would exhibit enhanced PGE2 and VEGF production versus those formed with 10,000 cells per spheroid in 21% oxygen with no inflammatory stimulus (Spheroid 2). Compared to Spheroid 2, Spheroid 1 produced fivefold more PGE2 and fourfold more VEGF, providing the opportunity to simultaneously upregulate the secretion of these factors from the same spheroid. The spheroids induced macrophage polarization, sprout formation with endothelial cells, and keratinocyte migration in a human skin equivalent model-demonstrating efficacy on three key cell types that are dysfunctional in chronic non-healing wounds. We conclude that DOE-based analysis effectively identifies optimal culture conditions to enhance the anti-inflammatory and proangiogenic potential of MSC spheroids. Stem Cells 2017;35:1493-1504.


Subject(s)
Anti-Inflammatory Agents/metabolism , Mesenchymal Stem Cells/metabolism , Neovascularization, Physiologic , Spheroids, Cellular/metabolism , Cell Polarity/drug effects , Cell Size/drug effects , Cellular Microenvironment/drug effects , Colony-Forming Units Assay , Culture Media, Conditioned/pharmacology , Humans , Macrophage Activation/drug effects , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/drug effects , NF-kappa B/metabolism , Neovascularization, Physiologic/drug effects , Phenotype , Reproducibility of Results , Research Design , Signal Transduction/drug effects , Skin, Artificial , Spheroids, Cellular/cytology , Spheroids, Cellular/drug effects , Tumor Necrosis Factor-alpha/metabolism , Wound Healing/drug effects
13.
Blood ; 125(12): 1855-6, 2015 Mar 19.
Article in English | MEDLINE | ID: mdl-25792733

ABSTRACT

In this issue of Blood, Liao et al report that kindlin-2 is necessary for angiogenic sprouting in vitro and for developmental and tumor angiogenesis in vivo. The process of blood vessel sprouting is known to involve the αVß3 vitronectin integrin on endothelium. Kindlin-2 linkage to the C-terminal tail of ß3 completed the outside-in circuit necessary for integrin signaling that is critical for navigation of new vessel sprouts.


Subject(s)
Cytoskeletal Proteins/metabolism , Integrin alphaVbeta3/chemistry , Integrin beta3/metabolism , Muscle Proteins/metabolism , Animals , Humans
14.
J Immunol ; 195(11): 5380-92, 2015 Dec 01.
Article in English | MEDLINE | ID: mdl-26519532

ABSTRACT

Recruitment of foamy monocytes to inflamed endothelium expressing VCAM-1 contributes to the development of plaque during atherogenesis. Foamy CD11c(+) monocytes arise in the circulation during the onset of hypercholesterolemia and recruit to nascent plaque, but the mechanism of CD11c/CD18 and very late Ag-4 (VLA-4) activation and cooperation in shear-resistant cell arrest on VCAM-1 are ill defined. Within 1 wk of the onset of a Western high-fat diet (WD) in apolipoprotein E-deficient mice, an inflammatory subset of foamy monocytes emerged that made up one fourth of the circulating population. These cells expressed ∼3-fold more CD11c/CD18 and 50% higher chemokine receptors than nonfoamy monocytes. Recruitment from blood to a VCAM-1 substrate under shear stress was assessed ex vivo using a unique artery-on-a-chip microfluidic assay. It revealed that foamy monocytes from mice on a WD increased their adhesiveness over 5 wk, rising to twice that of mice on a normal diet or CD11c(-/-) mice fed a WD. Shear-resistant capture of foamy human or mouse monocytes was initiated by high-affinity CD11c, which directly activated VLA-4 adhesion via phosphorylated spleen tyrosine kinase and paxillin within focal adhesion complexes. Lipid uptake and activation of CD11c are early and critical events in signaling VLA-4 adhesive function on foamy monocytes competent to recruit to VCAM-1 on inflamed arterial endothelium.


Subject(s)
CD11c Antigen/biosynthesis , CD18 Antigens/biosynthesis , Hypercholesterolemia/immunology , Inflammation/immunology , Integrin alpha4beta1/metabolism , Monocytes/immunology , Animals , Antigens, Ly/metabolism , Apolipoproteins E/genetics , Cell Adhesion , Cell Movement/immunology , Diet, High-Fat/adverse effects , Endothelium, Vascular/immunology , Endothelium, Vascular/pathology , Enzyme Activation , Focal Adhesions , Membrane Microdomains/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Microfluidics , Paxillin/metabolism , Vascular Cell Adhesion Molecule-1/metabolism
15.
J Mol Cell Cardiol ; 94: 180-188, 2016 05.
Article in English | MEDLINE | ID: mdl-27079253

ABSTRACT

OBJECTIVE: Endothelial dysfunction, including upregulation of inflammatory adhesion molecules and impaired vasodilatation, is a key element in cardiovascular disease. Aging and estrogen withdrawal in women are associated with endothelial inflammation, vascular stiffness and increased cardiovascular disease. Epoxyecosatrienoic acids (EETs), the products of arachidonic acid metabolism mediated by cytochrome P450 (CYP) 2J, 2C and other isoforms, are regulated by soluble epoxide hydrolase (sEH)-catalyzed conversion into less active diols. We hypothesized that 11,12-EETs would reduce the endothelial dysfunction associated with aging and estrogen loss. APPROACH/RESULTS: When stabilized by an sEH inhibitor (seHi), 11,12-EET at a physiologically low dose (0.1nM) reduced cytokine-stimulated upregulation of adhesion molecules on human aorta endothelial cells (HAEC) and monocyte adhesion under shear flow through marked depolarization of the HAEC when combined with TNFα. Mechanistically, neither 11,12-EETs nor 17ß-estradiol (E2) at physiologic concentrations prevented activation of NFκB by TNFα. E2 at physiological concentrations reduced sEH expression in HAEC, but did not alter CYP expression, and when combined with TNFα depolarized the cell. We also examined vascular dysfunction in adult and aged ovariectomized Norway brown rats (with and without E2 replacement) using an ex-vivo model to analyze endothelial function in an intact segment of artery. sEHi and 11,12-EET with or without E2 attenuated phenylephrine induced constriction and increased endothelial-dependent dilation of aortic rings from ovariectomized rats. CONCLUSIONS: Increasing 11,12-EETs through sEH inhibition effectively attenuates inflammation and may provide an effective strategy to preserve endothelial function and prevent atherosclerotic heart disease in postmenopausal women.


Subject(s)
8,11,14-Eicosatrienoic Acid/analogs & derivatives , Aging/metabolism , Endothelium, Vascular/metabolism , Estrogens/metabolism , 8,11,14-Eicosatrienoic Acid/metabolism , 8,11,14-Eicosatrienoic Acid/pharmacology , Animals , Cell Adhesion/drug effects , Cell Membrane/metabolism , Endothelium, Vascular/drug effects , Female , Humans , Membrane Potentials/drug effects , Monocytes/drug effects , Monocytes/metabolism , NF-kappa B/metabolism , Rats , Stress, Mechanical , Tumor Necrosis Factor-alpha/metabolism , Vascular Stiffness
16.
Arterioscler Thromb Vasc Biol ; 35(8): 1787-97, 2015 Aug.
Article in English | MEDLINE | ID: mdl-26112011

ABSTRACT

OBJECTIVE: To examine infiltration of blood foamy monocytes, containing intracellular lipid droplets, into early atherosclerotic lesions and its contribution to development of nascent atherosclerosis. APPROACH AND RESULTS: In apoE(-/-) mice fed Western high-fat diet (WD), >10% of circulating monocytes became foamy monocytes at 3 days on WD and >20% of monocytes at 1 week. Foamy monocytes also formed early in blood of Ldlr(-/-)Apobec1(-/-) (LDb) mice on WD. Based on CD11c and CD36, mouse monocytes were categorized as CD11c(-)CD36(-), CD11c(-)CD36(+), and CD11c(+)CD36(+). The majority of foamy monocytes were CD11c(+)CD36(+), whereas most nonfoamy monocytes were CD11c(-)CD36(-) or CD11c(-)CD36(+) in apoE(-/-) mice on WD. In wild-type mice, CD11c(+)CD36(+) and CD11c(-)CD36(+), but few CD11c(-)CD36(-), monocytes took up cholesteryl ester-rich very low-density lipoproteins (CE-VLDLs) isolated from apoE(-/-) mice on WD, and CE-VLDL uptake accelerated CD11c(-)CD36(+) to CD11c(+)CD36(+) monocyte differentiation. Ablation of CD36 decreased monocyte uptake of CE-VLDLs. Intravenous injection of DiI-CE-VLDLs in apoE(-/-) mice on WD specifically labeled CD11c(+)CD36(+) foamy monocytes, which infiltrated into nascent atherosclerotic lesions and became CD11c(+) cells that were selectively localized in atherosclerotic lesions. CD11c deficiency reduced foamy monocyte infiltration into atherosclerotic lesions. Specific and consistent depletion of foamy monocytes (for 3 weeks) by daily intravenous injections of low-dose clodrosome reduced development of nascent atherosclerosis. CONCLUSIONS: Foamy monocytes, which form early in blood of mice with hypercholesterolemia, infiltrate into early atherosclerotic lesions in a CD11c-dependent manner and play crucial roles in nascent atherosclerosis development.


Subject(s)
Aorta/pathology , Aortic Diseases/metabolism , Atherosclerosis/metabolism , Cholesterol, VLDL/metabolism , Hypercholesterolemia/metabolism , Monocytes/metabolism , Animals , Aorta/drug effects , Aorta/immunology , Aorta/metabolism , Aortic Diseases/genetics , Aortic Diseases/immunology , Aortic Diseases/pathology , Aortic Diseases/prevention & control , Apolipoproteins E/deficiency , Apolipoproteins E/genetics , Atherosclerosis/genetics , Atherosclerosis/immunology , Atherosclerosis/pathology , Atherosclerosis/prevention & control , CD11c Antigen/genetics , CD36 Antigens/deficiency , CD36 Antigens/genetics , Cell Line , Cholesterol, Dietary , Diet, High-Fat , Disease Models, Animal , Humans , Hypercholesterolemia/drug therapy , Hypercholesterolemia/genetics , Hypercholesterolemia/immunology , Male , Mice, Inbred C57BL , Mice, Knockout , Monocytes/drug effects , Monocytes/immunology , Receptors, LDL/deficiency , Receptors, LDL/genetics , Time Factors , Toll-Like Receptor 4/deficiency , Toll-Like Receptor 4/genetics
18.
Proc Natl Acad Sci U S A ; 110(34): 13944-9, 2013 Aug 20.
Article in English | MEDLINE | ID: mdl-23918401

ABSTRACT

Monocyte recruitment to inflamed arterial endothelium initiates plaque formation and drives progression of atherosclerosis. Three distinct monocyte subsets are detected in circulation (CD14(++)CD16(-), CD14(++)CD16(+), and CD14(+)CD16(++)), and each may play distinct roles during atherogenesis and myocardial infarction. We studied a range of subjects that included otherwise healthy patients with elevated serum triglyceride levels to patients presenting with acute myocardial infarction. Our objective was to correlate an individual's risk with the activation state of each monocyte subset as a function of changes in adhesion receptor expression using flow cytometric quantitation of integrins and l-selectin membrane expression. A microfluidic-based laboratory-on-a-chip was developed to quantify the adhesion efficiency of monocytes sheared in whole blood on vascular cell adhesion molecule-1, while characterizing adhesion receptor expression and topography on captured monocytes. CD14(++)CD16(+) monocytes adhered with sevenfold higher efficiency than other subsets, and in patients with myocardial infarction the capture efficiency of this subset was double that for healthy subjects. In patients with hypertriglyceridemia, this increase in monocyte adhesion was attributable to CD14(++)CD16(+) uptake of triglyceride-rich lipoproteins and subsequent signaling via a Phospholipase C-dependent mechanism to increase CD11c expression, very late antigen-4 function, and integrin coclustering within focal adhesive sites on vascular cell adhesion molecule-1. In summary, we introduce a unique laboratory-on-a-chip method for quantifying the activation state of monocyte subsets. These experiments reveal that CD11c/CD18 is an inducible integrin whose expression correlates with a monocyte inflammatory state in subjects at risk for atherogenesis and in patients with myocardial infarction.


Subject(s)
Atherosclerosis/metabolism , Endothelium, Vascular/pathology , Hypertriglyceridemia/complications , Monocytes/metabolism , Myocardial Infarction/metabolism , Phenotype , Adult , Analysis of Variance , Atherosclerosis/etiology , CD11c Antigen/metabolism , CD18 Antigens/metabolism , Cell Adhesion/physiology , Endothelium, Vascular/cytology , Female , Flow Cytometry , Humans , Male , Membrane Glycoproteins/metabolism , Microfluidic Analytical Techniques/methods , Monocytes/cytology , Myocardial Infarction/etiology , Platelet Glycoprotein GPIb-IX Complex , Vascular Cell Adhesion Molecule-1/metabolism
19.
Annu Rev Biomed Eng ; 16: 505-32, 2014 Jul 11.
Article in English | MEDLINE | ID: mdl-24905872

ABSTRACT

Mammals are endowed with a complex set of mechanisms that sense mechanical forces imparted by blood flow to endothelial cells (ECs), smooth muscle cells, and circulating blood cells to elicit biochemical responses through a process referred to as mechanotransduction. These biochemical responses are critical for a host of other responses, including regulation of blood pressure, control of vascular permeability for maintaining adequate perfusion of tissues, and control of leukocyte recruitment during immunosurveillance and inflammation. This review focuses on the role of the endothelial surface proteoglycan/glycoprotein layer-the glycocalyx (GCX)-that lines all blood vessel walls and is an agent in mechanotransduction and the modulation of blood cell interactions with the EC surface. We first discuss the biochemical composition and ultrastructure of the GCX, highlighting recent developments that reveal gaps in our understanding of the relationship between composition and spatial organization. We then consider the roles of the GCX in mechanotransduction and in vascular permeability control and review the prominent interaction of plasma-borne sphingosine-1 phosphate (S1P), which has been shown to regulate both the composition of the GCX and the endothelial junctions. Finally, we consider the association of GCX degradation with inflammation and vascular disease and end with a final section on future research directions.


Subject(s)
Capillary Permeability , Endothelium, Vascular/pathology , Mechanotransduction, Cellular/physiology , Animals , Blood Cells/cytology , Caveolae/metabolism , Cell Communication , Cell Membrane/metabolism , Endothelial Cells/cytology , Glycocalyx/metabolism , Glycoproteins/chemistry , Humans , Inflammation , Lysophospholipids/chemistry , Membrane Microdomains/chemistry , Mice , Microcirculation , Microscopy , Nitric Oxide/chemistry , Phospholipids/chemistry , Proteoglycans/chemistry , Signal Transduction , Sphingosine/analogs & derivatives , Sphingosine/chemistry
20.
Blood ; 121(20): 4017-8, 2013 May 16.
Article in English | MEDLINE | ID: mdl-23682030

ABSTRACT

In this issue of Blood, Jakob et al report that hematopoietic progenitor kinase 1 (HPK1) participates during signaling of neutrophil recruitment by acting as a regulator of the adhesiveness of the b2-integrin lymphocyte function-associated antigen 1 (LFA-1) during acute inflammation.


Subject(s)
Acute-Phase Reaction/genetics , Inflammation/genetics , Lymphocyte Function-Associated Antigen-1/physiology , Neutrophil Infiltration/genetics , Protein Serine-Threonine Kinases/physiology , Animals , Humans
SELECTION OF CITATIONS
SEARCH DETAIL