Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 55
Filter
Add more filters

Publication year range
1.
Kidney Int ; 106(1): 85-97, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38431215

ABSTRACT

Despite the recent advances in our understanding of the role of lipids, metabolites, and related enzymes in mediating kidney injury, there is limited integrated multi-omics data identifying potential metabolic pathways driving impaired kidney function. The limited availability of kidney biopsies from living donors with acute kidney injury has remained a major constraint. Here, we validated the use of deceased transplant donor kidneys as a good model to study acute kidney injury in humans and characterized these kidneys using imaging and multi-omics approaches. We noted consistent changes in kidney injury and inflammatory markers in donors with reduced kidney function. Neighborhood and correlation analyses of imaging mass cytometry data showed that subsets of kidney cells (proximal tubular cells and fibroblasts) are associated with the expression profile of kidney immune cells, potentially linking these cells to kidney inflammation. Integrated transcriptomic and metabolomic analysis of human kidneys showed that kidney arachidonic acid metabolism and seven other metabolic pathways were upregulated following diminished kidney function. To validate the arachidonic acid pathway in impaired kidney function we demonstrated increased levels of cytosolic phospholipase A2 protein and related lipid mediators (prostaglandin E2) in the injured kidneys. Further, inhibition of cytosolic phospholipase A2 reduced injury and inflammation in human kidney proximal tubular epithelial cells in vitro. Thus, our study identified cell types and metabolic pathways that may be critical for controlling inflammation associated with impaired kidney function in humans.


Subject(s)
Acute Kidney Injury , Phenotype , Humans , Acute Kidney Injury/metabolism , Acute Kidney Injury/pathology , Acute Kidney Injury/etiology , Male , Middle Aged , Metabolomics/methods , Female , Kidney Transplantation/adverse effects , Adult , Image Cytometry/methods , Kidney/pathology , Kidney/metabolism , Phospholipases A2/metabolism , Arachidonic Acid/metabolism , Kidney Tubules, Proximal/metabolism , Kidney Tubules, Proximal/pathology , Transcriptome , Dinoprostone/metabolism , Dinoprostone/analysis , Fibroblasts/metabolism , Gene Expression Profiling , Epithelial Cells/metabolism , Epithelial Cells/pathology , Biopsy , Multiomics
2.
J Pathol ; 257(3): 300-313, 2022 07.
Article in English | MEDLINE | ID: mdl-35239186

ABSTRACT

P2RX7, an ionotropic receptor for extracellular adenosine triphosphate (ATP), is expressed on immune cells, including macrophages, monocytes, and dendritic cells and is upregulated on nonimmune cells following injury. P2RX7 plays a role in many biological processes, including production of proinflammatory cytokines such as interleukin (IL)-1ß via the canonical inflammasome pathway. P2RX7 has been shown to be important in inflammation and fibrosis and may also play a role in autoimmunity. We have developed and phenotyped a novel P2RX7 knockout (KO) inbred rat strain and, taking advantage of the human-resembling unique histopathological features of rat models of glomerulonephritis, we induced three models of disease: nephrotoxic nephritis, experimental autoimmune glomerulonephritis, and experimental autoimmune vasculitis. We found that deletion of P2RX7 does not protect rats from models of experimental glomerulonephritis or the development of autoimmunity. Notably, treatment with A-438079, a P2RX7 antagonist, was equally protective in WKY WT and P2RX7 KO rats, revealing its 'off-target' properties. We identified a novel ATP/P2RX7/K+ efflux-independent and caspase-1/8-dependent pathway for the production of IL-1ß in rat dendritic cells, which was absent in macrophages. Taken together, these results comprehensively establish that inflammation and autoimmunity in glomerulonephritis is independent of P2RX7 and reveals the off-target properties of drugs previously known as selective P2RX7 antagonists. Rat mononuclear phagocytes may be able to utilise an 'alternative inflammasome' pathway to produce IL-1ß independently of P2RX7, which may account for the susceptibility of P2RX7 KO rats to inflammation and autoimmunity in glomerulonephritis. © 2022 The Authors. The Journal of Pathology published by John Wiley & Sons Ltd on behalf of The Pathological Society of Great Britain and Ireland.


Subject(s)
Glomerulonephritis , Receptors, Purinergic P2X7 , Vasculitis , Adenosine Triphosphate/metabolism , Animals , Caspase 1/metabolism , Caspases , Inflammasomes/metabolism , Inflammation/metabolism , Inflammation/pathology , Interleukin-1beta/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Rats , Rats, Inbred WKY , Receptors, Purinergic P2X7/metabolism , Vasculitis/metabolism , Vasculitis/pathology
3.
Haematologica ; 107(4): 933-946, 2022 04 01.
Article in English | MEDLINE | ID: mdl-34134470

ABSTRACT

The GPIbT-VWF A1 domain interaction is essential for platelet tethering under high shear. Synergy between GPIbα and GPVI signaling machineries has been suggested previously, however its molecular mechanism remains unclear. We generated a novel GPIbα transgenic mouse (GpIbαΔsig/Δsig) by CRISPR-Cas9 technology to delete the last 24 residues of the GPIbα intracellular tail that harbors the 14-3-3 and phosphoinositide-3 kinase binding sites. GPIbαΔsig/Δsig platelets bound VWF normally under flow. However, they formed fewer filopodia on VWF/botrocetin in the presence of a oIIbI3 blocker, demonstrating that despite normal ligand binding, VWF-dependent signaling is diminished. Activation of GpIbαΔsig/Δsig platelets with ADP and thrombin was normal, but GpIbαΔsig/Δsig platelets stimulated with collagen-related-peptide (CRP) exhibited markedly decreased P-selectin exposure and eIIbI3 activation, suggesting a role for the GpIbaaintracellular tail in GPVI-mediated signaling. Consistent with this, while haemostasis was normal in GPIbαΔsig/Δsig mice, diminished tyrosine-phosphorylation, (particularly pSYK) was detected in CRP-stimulated GpIbαΔsig/Δsig platelets as well as reduced platelet spreading on CRP. Platelet responses to rhodocytin were also affected in GpIbαΔsig/Δsig platelets but to a lesser extent than those with CRP. GpIbαΔsig/Δsig platelets formed smaller aggregates than wild-type platelets on collagen-coated microchannels at low, medium and high shear. In response to both VWF and collagen binding, flow assays performed with plasma-free blood or in the presence of bIIbI3- or GPVI-blockers suggested reduced bIIbI3 activation contributes to the phenotype of the GpIbαΔsig/Δsig platelets. Together, these results reveal a new role for the intracellular tail of GPIbiiin transducing both VWF-GPIbGGand collagen-GPVI signaling events in platelets.


Subject(s)
Blood Platelets , von Willebrand Factor , Animals , Blood Platelets/metabolism , Collagen/metabolism , Hemostasis , Humans , Mice , Signal Transduction , von Willebrand Factor/genetics , von Willebrand Factor/metabolism
4.
BMC Nephrol ; 23(1): 208, 2022 06 13.
Article in English | MEDLINE | ID: mdl-35698028

ABSTRACT

Diabetic kidney disease (DKD) is the leading cause of kidney failure worldwide. Mortality and morbidity associated with DKD are increasing with the global prevalence of type 2 diabetes. Chronic, sub-clinical, non-resolving inflammation contributes to the pathophysiology of renal and cardiovascular disease associated with diabetes. Inflammatory biomarkers correlate with poor renal outcomes and mortality in patients with DKD. Targeting chronic inflammation may therefore offer a route to novel therapeutics for DKD. However, the DKD patient population is highly heterogeneous, with varying etiology, presentation and disease progression. This heterogeneity is a challenge for clinical trials of novel anti-inflammatory therapies. Here, we present a conceptual model of how chronic inflammation affects kidney function in five compartments: immune cell recruitment and activation; filtration; resorption and secretion; extracellular matrix regulation; and perfusion. We believe that the rigorous alignment of pathophysiological insights, appropriate animal models and pathology-specific biomarkers may facilitate a mechanism-based shift from recruiting 'all comers' with DKD to stratification of patients based on the principal compartments of inflammatory disease activity.


Subject(s)
Diabetes Mellitus, Type 2 , Diabetic Nephropathies , Animals , Biomarkers , Diabetes Mellitus, Type 2/complications , Diabetes Mellitus, Type 2/drug therapy , Diabetic Nephropathies/etiology , Humans , Inflammation/complications , Inflammation/drug therapy , Kidney
5.
J Hepatol ; 75(1): 177-189, 2021 07.
Article in English | MEDLINE | ID: mdl-33631227

ABSTRACT

BACKGROUND & AIMS: Checkpoint inhibitor-related hepatitis (CPI-Hep) is an emerging clinical challenge. We aimed to gain insights into the immunopathology of CPI-Hep by comprehensively characterising myeloid and lymphoid subsets. METHODS: CPI-treated patients with or without related hepatitis (CPI-Hep; n = 22 and CPI-noHep; n = 7) were recruited. Phenotypic and transcriptional profiling of peripheral immune subsets was performed and compared with 19 healthy controls (HCs). In vitro monocyte-derived macrophages (MoMFs) were assessed for activation and cytokine production. CD163, CCR2, CD68, CD3, CD8 and granzyme B expression was assessed using immunohistochemistry/immunofluorescence (n = 4). RESULTS: A significant total monocyte depletion was observed in CPI-Hep compared with HCs (p = 0.04), along with a proportionate increase in the classical monocyte population (p = 0.0002) and significant upregulation of CCR2, CD163 and downregulation of CCR7. Soluble CD163 levels were significantly elevated in CPI-Hep compared with HCs (p <0.0001). In vitro MoMFs from CPI-Hep showed enhanced production of pro-inflammatory cytokines. CD8+ T cells demonstrated increased perforin, granzyme B, ICOS and HLA-DR expression in CPI-Hep. Transcriptional profiling indicated the presence of activated monocyte and enhanced effector CD8+ T cell populations in CPI-Hep. Immunohistochemistry demonstrated co-localisation of CD8+/granzyme B+ T cells with CD68+CCR2+/CD68+CD163+ macrophages in CPI-Hep liver tissue. CONCLUSIONS: CPI-Hep is associated with activation of peripheral monocytes and an enhanced cytotoxic, effector CD8+ T cell phenotype. These changes were reflected by liver inflammation composed of CD163+/CCR2+ macrophages and CD8+ T cells. LAY SUMMARY: Some patients who receive immunotherapy for cancer develop liver inflammation, which requires cessation of cancer treatment. Herein, we describe ways in which the white blood cells of patients who develop liver inflammation differ from those of patients who receive the same immunotherapy but do not experience liver-related side effects. Targeting some of the pathways we identify may help to prevent or manage this side effect and facilitate cancer treatment.


Subject(s)
Antigens, CD/immunology , Antigens, Differentiation, Myelomonocytic/immunology , Antineoplastic Agents/adverse effects , CD8-Positive T-Lymphocytes/immunology , Chemical and Drug Induced Liver Injury , Immune Checkpoint Inhibitors/adverse effects , Macrophages/immunology , Receptors, CCR2/immunology , Receptors, CCR7/immunology , Receptors, Cell Surface/immunology , Antineoplastic Agents/administration & dosage , Chemical and Drug Induced Liver Injury/blood , Chemical and Drug Induced Liver Injury/etiology , Chemical and Drug Induced Liver Injury/immunology , Chemical and Drug Induced Liver Injury/prevention & control , Drug Discovery , Female , Humans , Immune Checkpoint Inhibitors/administration & dosage , Immune Checkpoint Inhibitors/immunology , Macrophage Activation/immunology , Male , Middle Aged , T-Lymphocytes, Cytotoxic/immunology
6.
J Am Soc Nephrol ; 31(11): 2523-2542, 2020 11.
Article in English | MEDLINE | ID: mdl-32868399

ABSTRACT

BACKGROUND: Immune complexes within glomerular capillary walls cause crescentic GN (CrGN). Monocytes and macrophages are important in mediating CrGN, but little work has been done to phenotype the subpopulations involved and determine their respective contributions to glomerular inflammation. METHODS: Live glomerular imaging using confocal microscopy monitored intravascular monocyte subset behavior during nephrotoxic nephritis (NTN) in a novel WKY-hCD68-GFP monocyte/macrophage reporter rat strain. Flow cytometry and qPCR further analyzed ex vivo the glomerular leukocyte infiltrate during NTN. RESULTS: Non-classical monocytes surveyed the glomerular endothelium via lymphocyte function-associated antigen 1 (LFA-1) in the steady state. During NTN, non-classical monocytes were recruited first, but subsequent recruitment and retention of classical monocytes was associated with glomerular damage. Monocytes recruited to the glomerular vasculature did not undergo transendothelial migration. This finding suggests that inflammation in immune complex-mediated CrGN is predominantly intravascular, driven by dynamic interactions between intravascular blood monocytes and the endothelium. Glomerular endothelium and non-classical monocytes overexpressed a distinct chemokine axis, which may orchestrate inflammatory myeloid cell recruitment and expression of damage mediators. Reduced classical monocyte recruitment in Lewis rats during NTN confirmed a role for CD16 in mediating glomerular damage. CONCLUSIONS: Monocyte subsets with distinct phenotypes and effector functions may be important in driving inflammation in experimental CrGN resulting from immune complexes formed within the glomerular capillary wall. LFA-1-dependent endothelial surveillance by non-classical monocytes may detect immune complexes through CD16, orchestrating the inflammatory response through intravascular retention of classical monocytes, which results in glomerular damage and proteinuria.


Subject(s)
Endothelium/pathology , Glomerulonephritis/immunology , Glomerulonephritis/pathology , Kidney Glomerulus/immunology , Monocytes/pathology , Monocytes/physiology , Animals , Antigens, CD/metabolism , Antigens, Differentiation, Myelomonocytic/metabolism , Capillaries , Cell Movement , Endothelium/immunology , Flow Cytometry , Intravital Microscopy , Kidney Glomerulus/diagnostic imaging , Male , Microscopy, Confocal , Monocytes/metabolism , Phenotype , Rats , Receptors, IgG/metabolism
7.
Gut ; 67(2): 333-347, 2018 02.
Article in English | MEDLINE | ID: mdl-28450389

ABSTRACT

OBJECTIVE: Acute liver failure (ALF) is characterised by overwhelming hepatocyte death and liver inflammation with massive infiltration of myeloid cells in necrotic areas. The mechanisms underlying resolution of acute hepatic inflammation are largely unknown. Here, we aimed to investigate the impact of Mer tyrosine kinase (MerTK) during ALF and also examine how the microenvironmental mediator, secretory leucocyte protease inhibitor (SLPI), governs this response. DESIGN: Flow cytometry, immunohistochemistry, confocal imaging and gene expression analyses determined the phenotype, functional/transcriptomic profile and tissue topography of MerTK+ monocytes/macrophages in ALF, healthy and disease controls. The temporal evolution of macrophage MerTK expression and its impact on resolution was examined in APAP-induced acute liver injury using wild-type (WT) and Mer-deficient (Mer-/-) mice. SLPI effects on hepatic myeloid cells were determined in vitro and in vivo using APAP-treated WT mice. RESULTS: We demonstrate a significant expansion of resolution-like MerTK+HLA-DRhigh cells in circulatory and tissue compartments of patients with ALF. Compared with WT mice which show an increase of MerTK+MHCIIhigh macrophages during the resolution phase in ALF, APAP-treated Mer-/- mice exhibit persistent liver injury and inflammation, characterised by a decreased proportion of resident Kupffer cells and increased number of neutrophils. Both in vitro and in APAP-treated mice, SLPI reprogrammes myeloid cells towards resolution responses through induction of a MerTK+HLA-DRhigh phenotype which promotes neutrophil apoptosis and their subsequent clearance. CONCLUSIONS: We identify a hepatoprotective, MerTK+, macrophage phenotype that evolves during the resolution phase following ALF and represents a novel immunotherapeutic target to promote resolution responses following acute liver injury.


Subject(s)
Liver Failure, Acute/immunology , Liver Failure, Acute/metabolism , Macrophages/metabolism , Secretory Leukocyte Peptidase Inhibitor/pharmacology , c-Mer Tyrosine Kinase/metabolism , Acetaminophen , Adult , Aged , Animals , Case-Control Studies , Female , Gene Expression , Genes, MHC Class II , HLA-DR Antigens/metabolism , Humans , Kupffer Cells/immunology , Kupffer Cells/metabolism , Liver Failure, Acute/chemically induced , Liver Failure, Acute/pathology , Macrophages/immunology , Male , Mice , Middle Aged , Monocytes/immunology , Monocytes/metabolism , Neutrophils/physiology , Phenotype , Secretory Leukocyte Peptidase Inhibitor/metabolism , Secretory Leukocyte Peptidase Inhibitor/therapeutic use , Transcriptome , c-Mer Tyrosine Kinase/deficiency , c-Mer Tyrosine Kinase/genetics
8.
Blood ; 124(24): 3646-55, 2014 Dec 04.
Article in English | MEDLINE | ID: mdl-25293769

ABSTRACT

Tissue factor (TF) (CD142) is a 47 kDa transmembrane cell surface glycoprotein that triggers the extrinsic coagulation cascade and links thrombosis with inflammation. Although macrophage TF expression is known to be regulated at the RNA level, very little is known about the mechanisms involved. Poly(adenosine 5'-diphosphate [ADP]-ribose)-polymerase (PARP)-14 belongs to a family of intracellular proteins that generate ADP-ribose posttranslational adducts. Functional screening of PARP-14-deficient macrophages mice revealed that PARP-14 deficiency leads to increased TF expression and functional activity in macrophages after challenge with bacterial lipopolysaccharide. This was related to an increase in TF messenger RNA (mRNA) stability. Ribonucleoprotein complex immunoprecipitation and biotinylated RNA pull-down assays demonstrated that PARP-14 forms a complex with the mRNA-destabilizing protein tristetraprolin (TTP) and a conserved adenylate-uridylate-rich element in the TF mRNA 3' untranslated region. TF mRNA regulation by PARP-14 was selective, as tumor necrosis factor (TNF)α mRNA, which is also regulated by TTP, was not altered in PARP-14 deficient macrophages. Consistent with the in vitro data, TF expression and TF activity, but not TNFα expression, were increased in Parp14(-/-) mice in vivo. Our study provides a novel mechanism for the posttranscriptional regulation of TF expression, indicating that this is selectively regulated by PARP-14.


Subject(s)
Gene Expression Regulation , Macrophages/metabolism , Poly(ADP-ribose) Polymerases/metabolism , Thromboplastin/biosynthesis , Tristetraprolin/metabolism , 3' Untranslated Regions/physiology , Animals , Lipopolysaccharides/pharmacology , Mice , Mice, Knockout , Poly(ADP-ribose) Polymerases/genetics , RNA Stability/drug effects , RNA Stability/physiology , Thromboplastin/genetics , Tristetraprolin/genetics , Tumor Necrosis Factor-alpha/biosynthesis , Tumor Necrosis Factor-alpha/genetics
9.
Circulation ; 130(1): 35-50, 2014 Jul 01.
Article in English | MEDLINE | ID: mdl-24982116

ABSTRACT

BACKGROUND: The relevance of the dissociation of circulating pentameric C-reactive protein (pCRP) to its monomeric subunits (mCRP) is poorly understood. We investigated the role of conformational C-reactive protein changes in vivo. METHODS AND RESULTS: We identified mCRP in inflamed human striated muscle, human atherosclerotic plaque, and infarcted myocardium (rat and human) and its colocalization with inflammatory cells, which suggests a general causal role of mCRP in inflammation. This was confirmed in rat intravital microscopy of lipopolysaccharide-induced cremasteric muscle inflammation. Intravenous pCRP administration significantly enhanced leukocyte rolling, adhesion, and transmigration via localized dissociation to mCRP in inflamed but not noninflamed cremaster muscle. This was confirmed in a rat model of myocardial infarction. Mechanistically, this process was dependent on exposure of lysophosphatidylcholine on activated cell membranes, which is generated after phospholipase A2 activation. These membrane changes could be visualized intravitally on endothelial cells, as could the colocalized mCRP generation. Blocking of phospholipase A2 abrogated C-reactive protein dissociation and thereby blunted the proinflammatory effects of C-reactive protein. Identifying the dissociation process as a therapeutic target, we stabilized pCRP using 1,6-bis(phosphocholine)-hexane, which prevented dissociation in vitro and in vivo and consequently inhibited the generation and proinflammatory activity of mCRP; notably, it also inhibited mCRP deposition and inflammation in rat myocardial infarction. CONCLUSIONS: These results provide in vivo evidence for a novel mechanism that localizes and aggravates inflammation via phospholipase A2-dependent dissociation of circulating pCRP to mCRP. mCRP is proposed as a pathogenic factor in atherosclerosis and myocardial infarction. Most importantly, the inhibition of pCRP dissociation represents a promising, novel anti-inflammatory therapeutic strategy.


Subject(s)
C-Reactive Protein/chemistry , Carrier Proteins/chemistry , Inflammation/metabolism , Muscle, Skeletal/metabolism , Myocardial Infarction/metabolism , Myositis/metabolism , Animals , Anti-Inflammatory Agents/pharmacology , Anti-Inflammatory Agents/therapeutic use , Biopolymers , C-Reactive Protein/physiology , Carrier Proteins/physiology , Cell Adhesion/drug effects , Cell Membrane/drug effects , Cell Membrane/metabolism , Chemotaxis, Leukocyte , Complement Activation , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Hexanes/pharmacology , Hexanes/therapeutic use , Humans , Inflammation/drug therapy , Inflammation/etiology , Leukocyte Rolling/drug effects , Lipopolysaccharides/toxicity , Lysophosphatidylcholines/metabolism , Male , Membrane Lipids/metabolism , Muscle, Skeletal/blood supply , Myocardial Infarction/pathology , Myositis/chemically induced , Myositis/pathology , Phospholipase A2 Inhibitors/pharmacology , Phospholipase A2 Inhibitors/therapeutic use , Phospholipases A2/metabolism , Phosphorylcholine/analogs & derivatives , Phosphorylcholine/pharmacology , Phosphorylcholine/therapeutic use , Protein Structure, Quaternary , Random Allocation , Rats , Rats, Wistar , Receptors, IgG/physiology , Reperfusion Injury/metabolism , Reperfusion Injury/pathology
10.
Arterioscler Thromb Vasc Biol ; 34(9): 2120-7, 2014 Sep.
Article in English | MEDLINE | ID: mdl-25060791

ABSTRACT

OBJECTIVE: Patients with chronic kidney disease (CKD) display impaired cholesterol efflux capacity and elevated CD14(++)CD16(+) monocyte counts. In mice, dysfunctional cholesterol efflux causes monocytosis. It is unknown whether cholesterol efflux capacity and monocyte subsets are associated in CKD. APPROACH AND RESULTS: In 438 patients with CKD, mediators of cholesterol efflux capacity (high-density lipoprotein cholesterol/apolipoprotein A-I) and monocyte subsets were analyzed as predictors of cardiovascular events. Monocyte subset-specific intracellular lipid content, CD36, CD68, and ABCA1 were measured in a subgroup. Experimentally, we analyzed subset-specific cholesterol efflux capacity and response to oxidized low-density lipoprotein cholesterol stimulation in CKD. Epidemiologically, both low Apo-I and low high-density lipoprotein cholesterol were associated with high CD14(++)CD16(+) monocyte counts in linear regression analyses (apolipoprotein A-I: ß=-0.171; P<0.001; high-density lipoprotein cholesterol: ß=-0.138; P=0.005), but not with counts of other monocyte subsets. In contrast to apolipoprotein A-I or high-density lipoprotein cholesterol, higher CD14(++)CD16(+) monocyte counts independently predicted cardiovascular events (hazard ratio per increase of 1 cell/µL: 1.011 [1.003-1.020]; P=0.007). Experimentally, CD14(++)CD16(+) monocytes demonstrated preferential lipid accumulation, high CD36, CD68, and low ABCA1 expression and, consequently, displayed low cholesterol efflux capacity, avid oxidized low-density lipoprotein cholesterol uptake, and potent intracellular interleukin-6, interleukin-1ß, and tumor necrosis factor-α production. CONCLUSIONS: Taken together, mediators of cholesterol efflux are associated with CD14(++)CD16(+) monocyte counts, which independently predict adverse outcome in CKD.


Subject(s)
Apolipoprotein A-I/analysis , Cardiovascular Diseases/epidemiology , Cholesterol, HDL/blood , Monocytes , Renal Insufficiency, Chronic/blood , ATP Binding Cassette Transporter 1/blood , Aged , Cardiovascular Diseases/etiology , Female , GPI-Linked Proteins/analysis , Humans , Immunophenotyping , Interleukin-1beta/blood , Interleukin-6/biosynthesis , Interleukin-6/blood , Leukocyte Count , Lipids/blood , Lipopolysaccharide Receptors/analysis , Lipoproteins, LDL/blood , Male , Middle Aged , Monocytes/classification , Prospective Studies , Receptors, IgG/analysis , Renal Insufficiency, Chronic/complications , Single-Blind Method , Tumor Necrosis Factor-alpha/analysis
11.
Kidney Int ; 85(5): 1011-4, 2014 May.
Article in English | MEDLINE | ID: mdl-24786874

ABSTRACT

The kidney has a diverse repertoire of cells that make up the mononuclear phagocyte system (MPS). Wu et al. identify a population of CD8αα+CD11c+MHC-II+ blood precursors that display dendritic cell-like characteristics in the glomeruli. These cells show increased recruitment in a rat anti-glomerular basement membrane glomerulonephritis model and were able to attenuate disease. This study highlights the importance of the MPS in kidney disease and the need to better understand it to develop immunotherapeutics translatable to the renal patient.


Subject(s)
Adoptive Transfer , Autoimmunity , CD11c Antigen/blood , CD3 Complex/blood , CD8 Antigens/blood , CD8-Positive T-Lymphocytes/transplantation , Glomerulonephritis/prevention & control , Histocompatibility Antigens Class II/blood , Kidney Glomerulus/immunology , Animals , Female
13.
ACS Chem Biol ; 19(4): 962-972, 2024 04 19.
Article in English | MEDLINE | ID: mdl-38509779

ABSTRACT

Inhibition of the NLRP3 inflammasome is a promising strategy for the development of new treatments for inflammatory diseases. MCC950 is a potent and selective small-molecule inhibitor of the NLRP3 pathway and has been validated in numerous species and disease models. Although the capacity of MCC950 to block NLRP3 signaling is well-established, it is still critical to identify the mechanism of action and molecular targets of MCC950 to inform and derisk drug development. Quantitative proteomics performed in disease-relevant systems provides a powerful method to study both direct and indirect pharmacological responses to small molecules to elucidate the mechanism of action and confirm target engagement. A comprehensive target deconvolution campaign requires the use of complementary chemical biology techniques. Here we applied two orthogonal chemical biology techniques: compressed Cellular Thermal Shift Assay (CETSA) and photoaffinity labeling chemoproteomics, performed under biologically relevant conditions with LPS-primed THP-1 cells, thereby deconvoluting, for the first time, the molecular targets of MCC950 using chemical biology techniques. In-cell chemoproteomics with inlysate CETSA confirmed the suspected mechanism as the disruption of inflammasome formation via NLRP3. Further cCETSA (c indicates compressed) in live cells mapped the stabilization of NLRP3 inflammasome pathway proteins, highlighting modulation of the targeted pathway. This is the first evidence of direct MCC950 engagement with endogenous NLRP3 in a human macrophage cellular system using discovery proteomics chemical biology techniques, providing critical information for inflammasome studies.


Subject(s)
Inflammasomes , NLR Family, Pyrin Domain-Containing 3 Protein , Animals , Humans , Cell Line , Disease Models, Animal , Furans/pharmacology , Inflammasomes/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Proteomics , Sulfonamides/pharmacology , Sulfones/pharmacology
14.
J Immunother Cancer ; 12(4)2024 Apr 04.
Article in English | MEDLINE | ID: mdl-38580334

ABSTRACT

BACKGROUND: Checkpoint inhibitor-induced hepatitis (CPI-hepatitis) is an emerging problem with the widening use of CPIs in cancer immunotherapy. Here, we developed a mouse model to characterize the mechanism of CPI-hepatitis and to therapeutically target key pathways driving this pathology. METHODS: C57BL/6 wild-type (WT) mice were dosed with toll-like receptor (TLR)9 agonist (TLR9-L) for hepatic priming combined with anti-cytotoxic T lymphocyte antigen-4 (CTLA-4) plus anti-programmed cell death 1 (PD-1) ("CPI") or phosphate buffered saline (PBS) control for up to 7 days. Flow cytometry, histology/immunofluorescence and messenger RNA sequencing were used to characterize liver myeloid/lymphoid subsets and inflammation. Hepatocyte damage was assessed by plasma alanine transaminase (ALT) and cytokeratin-18 (CK-18) measurements. In vivo investigations of CPI-hepatitis were carried out in Rag2-/- and Ccr2rfp/rfp transgenic mice, as well as following anti-CD4, anti-CD8 or cenicriviroc (CVC; CCR2/CCR5 antagonist) treatment. RESULTS: Co-administration of combination CPIs with TLR9-L induced liver pathology closely resembling human disease, with increased infiltration and clustering of granzyme B+perforin+CD8+ T cells and CCR2+ monocytes, 7 days post treatment. This was accompanied by apoptotic hepatocytes surrounding these clusters and elevated ALT and CK-18 plasma levels. Liver RNA sequencing identified key signaling pathways (JAK-STAT, NF-ΚB) and cytokine/chemokine networks (Ifnγ, Cxcl9, Ccl2/Ccr2) as drivers of CPI-hepatitis. Using this model, we show that CD8+ T cells mediate hepatocyte damage in experimental CPI-hepatitis. However, their liver recruitment, clustering, and cytotoxic activity is dependent on the presence of CCR2+ monocytes. The absence of hepatic monocyte recruitment in Ccr2rfp/rfp mice and CCR2 inhibition by CVC treatment in WT mice was able to prevent the development and reverse established experimental CPI-hepatitis. CONCLUSION: This newly established mouse model provides a platform for in vivo mechanistic studies of CPI-hepatitis. Using this model, we demonstrate the central role of liver infiltrating CCR2+ monocyte interaction with tissue-destructive CD8+ T cells in the pathogenesis of CPI-hepatitis and highlight CCR2 inhibition as a novel therapeutic target.


Subject(s)
Hepatitis , Monocytes , Humans , Mice , Animals , CD8-Positive T-Lymphocytes , Toll-Like Receptor 9 , Mice, Inbred C57BL , Hepatitis/drug therapy , Hepatitis/etiology
15.
Cell Rep ; 43(2): 113602, 2024 Feb 27.
Article in English | MEDLINE | ID: mdl-38237600

ABSTRACT

Recent regenerative studies using human pluripotent stem cells (hPSCs) have developed multiple kidney-lineage cells and organoids. However, to further form functional segments of the kidney, interactions of epithelial and interstitial cells are required. Here we describe a selective differentiation of renal interstitial progenitor-like cells (IPLCs) from human induced pluripotent stem cells (hiPSCs) by modifying our previous induction method for nephron progenitor cells (NPCs) and analyzing mouse embryonic interstitial progenitor cell (IPC) development. Our IPLCs combined with hiPSC-derived NPCs and nephric duct cells form nephrogenic niche- and mesangium-like structures in vitro. Furthermore, we successfully induce hiPSC-derived IPLCs to differentiate into mesangial and erythropoietin-producing cell lineages in vitro by screening differentiation-inducing factors and confirm that p38 MAPK, hypoxia, and VEGF signaling pathways are involved in the differentiation of mesangial-lineage cells. These findings indicate that our IPC-lineage induction method contributes to kidney regeneration and developmental research.


Subject(s)
Erythropoietin , Induced Pluripotent Stem Cells , Humans , Animals , Mice , Kidney , Cell Lineage , Regeneration
16.
J Adv Res ; 2024 Jul 09.
Article in English | MEDLINE | ID: mdl-38992424

ABSTRACT

INTRODUCTION: Despite recent substantial progress in vascularized composite allotransplantation (VCA), such as face transplantations, short- and long-term allograft survival is severely limited by allograft rejection. The acute-phase response, directly after allogeneic transplantation, represents an immune-inflammatory reaction to ischemia/reperfusion and acts as an early initiator of graft rejection. Acute-phase reactants mediate this immune response via crosstalk with the mononuclear phagocyte system. OBJECTIVE: C-reactive protein (CRP), a well-known marker of inflammation, has pro-inflammatory properties and aggravates ischemia/reperfusion injury. Thus, we investigated how CRP impacts acute allograft rejection. METHODS: Based on clinical observations in facial VCAs, we applied a complex hindlimb transplantation model in rats to investigate whether CRP directly affects transplant rejection. We further analyzed subset-specific infiltration and tissue distribution of recipient-derived monocytes in the early phase of acute rejection and assessed their differential regulation by CRP using intravital imaging. RESULTS: We demonstrate that CRP accelerates allograft rejection and reduces allograft survival via selectively activating non-classical monocytes. The therapeutic stabilization of CRP abrogates this activating effect on monocytes, consequently attenuating acute allograft rejection. Intravital imaging of graft-infiltrating, recipient-derived monocytes during the early phase of acute rejection confirmed their differential regulation by CRP and their crucial role in driving the early stage of graft rejection. CONCLUSION: Differential activation of recipient-derived monocytes by CRP aggravates innate immune response and accelerates clinical allograft rejection Thus, therapeutic targeting of CRP represents a novel promising strategy for preventing acute allograft rejection and potentially reducing chronic allograft rejection.

17.
Clin Sci (Lond) ; 125(5): 221-35, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23668229

ABSTRACT

Cardiovascular disease is the leading cause of death in several countries. The underlying process is atherosclerosis, a slowly progressing chronic disorder that can lead to intravascular thrombosis. There is overwhelming evidence for the underlying importance of our immune system in atherosclerosis. Monocytes, which comprise part of the innate immune system, can be recruited to inflamed endothelium and this recruitment has been shown to be proportional to the extent of atherosclerotic disease. Monocytes undergo migration into the vasculature, they differentiate into macrophage phenotypes, which are highly phagocytic and can scavenge modified lipids, leading to foam cell formation and development of the lipid-rich atheroma core. This increased influx leads to a highly inflammatory environment and along with other immune cells can increase the risk in the development of the unstable atherosclerotic plaque phenotype. The present review provides an overview and description of the immunological aspect of innate and adaptive immune cell subsets in atherosclerosis, by defining their interaction with the vascular environment, modified lipids and other cellular exchanges. There is a particular focus on monocytes and macrophages, but shorter descriptions of dendritic cells, lymphocyte populations, neutrophils, mast cells and platelets are also included.


Subject(s)
Atherosclerosis/immunology , Mononuclear Phagocyte System/physiology , Animals , Blood Platelets/physiology , Humans
18.
Elife ; 122023 08 11.
Article in English | MEDLINE | ID: mdl-37566453

ABSTRACT

Monocytes are heterogeneous innate effector leukocytes generated in the bone marrow and released into circulation in a CCR2-dependent manner. During infection or inflammation, myelopoiesis is modulated to rapidly meet the demand for more effector cells. Danger signals from peripheral tissues can influence this process. Herein we demonstrate that repetitive TLR7 stimulation via the epithelial barriers drove a potent emergency bone marrow monocyte response in mice. This process was unique to TLR7 activation and occurred independently of the canonical CCR2 and CX3CR1 axes or prototypical cytokines. The monocytes egressing the bone marrow had an immature Ly6C-high profile and differentiated into vascular Ly6C-low monocytes and tissue macrophages in multiple organs. They displayed a blunted cytokine response to further TLR7 stimulation and reduced lung viral load after RSV and influenza virus infection. These data provide insights into the emergency myelopoiesis likely to occur in response to the encounter of single-stranded RNA viruses at barrier sites.


Subject(s)
Myelopoiesis , Toll-Like Receptor 7 , Virus Diseases , Animals , Mice , Cytokines , Lung , Mice, Inbred C57BL , Monocytes , Toll-Like Receptor 7/genetics , Virus Diseases/immunology
19.
Kidney Int Rep ; 8(1): 75-80, 2023 Jan.
Article in English | MEDLINE | ID: mdl-36644346

ABSTRACT

Introduction: Inflammation is highly prevalent among patients with end-stage kidney disease and is associated with adverse outcomes. We aimed to investigate longitudinal changes in inflammatory markers in a diverse international incident hemodialysis patient population. Methods: The MONitoring Dialysis Outcomes (MONDO) Consortium encompasses hemodialysis databases from 31 countries in Europe, North America, South America, and Asia. The MONDO database was queried for inflammatory markers (total white blood cell count [WBC], neutrophil count, lymphocyte count, serum albumin, and C-reactive protein [CRP]) and hemoglobin levels in incident hemodialysis patients. Laboratory parameters were measured every month. Patients were stratified by survival time (≤6 months, >6 to 12 months, >12 to 18 months, >18 to 24 months, >24 to 30 months, >30 to 36 months, and >36 months) following dialysis initiation. We used cubic B-spline basis function to evaluate temporal changes in inflammatory parameters in relationship with patient survival. Results: We studied 18,726 incident hemodialysis patients. Their age at dialysis initiation was 71.3 ± 11.9 years; 10,802 (58%) were males. Within the first 6 months, 2068 (11%) patients died, and 12,295 patients (67%) survived >36 months (survivor cohort). Hemodialysis patients who died showed a distinct biphasic pattern of change in inflammatory markers where an initial decline of inflammation was followed by a rapid rise that was consistently evident approximately 6 months before death. This pattern was similar in all patients who died and was consistent across the survival time intervals. In contrast, in the survivor cohort, we observed initial decline of inflammation followed by sustained low levels of inflammatory biomarkers. Conclusion: Our international study of incident hemodialysis patients highlights a temporal relationship between serial measurements of inflammatory markers and patient survival. This finding may inform the development of prognostic models, such as the integration of dynamic changes in inflammatory markers for individual risk profiling and guiding preventive and therapeutic interventions.

20.
EMBO Mol Med ; 15(1): e16236, 2023 01 11.
Article in English | MEDLINE | ID: mdl-36468184

ABSTRACT

C-reactive protein (CRP) is an early-stage acute phase protein and highly upregulated in response to inflammatory reactions. We recently identified a novel mechanism that leads to a conformational change from the native, functionally relatively inert, pentameric CRP (pCRP) structure to a pentameric CRP intermediate (pCRP*) and ultimately to the monomeric CRP (mCRP) form, both exhibiting highly pro-inflammatory effects. This transition in the inflammatory profile of CRP is mediated by binding of pCRP to activated/damaged cell membranes via exposed phosphocholine lipid head groups. We designed a tool compound as a low molecular weight CRP inhibitor using the structure of phosphocholine as a template. X-ray crystallography revealed specific binding to the phosphocholine binding pockets of pCRP. We provide in vitro and in vivo proof-of-concept data demonstrating that the low molecular weight tool compound inhibits CRP-driven exacerbation of local inflammatory responses, while potentially preserving pathogen-defense functions of CRP. The inhibition of the conformational change generating pro-inflammatory CRP isoforms via phosphocholine-mimicking compounds represents a promising, potentially broadly applicable anti-inflammatory therapy.


Subject(s)
C-Reactive Protein , Phosphorylcholine , Humans , Phosphorylcholine/pharmacology , Inflammation/drug therapy , Inflammation/metabolism , Cell Membrane/metabolism , Anti-Inflammatory Agents
SELECTION OF CITATIONS
SEARCH DETAIL