Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 88
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Cell ; 178(5): 1205-1221.e17, 2019 08 22.
Article in English | MEDLINE | ID: mdl-31442408

ABSTRACT

A hallmark feature of inflammation is the orchestrated recruitment of neutrophils from the bloodstream into inflamed tissue. Although selectins and integrins mediate recruitment in many tissues, they have a minimal role in the lungs and liver. Exploiting an unbiased in vivo functional screen, we identified a lung and liver homing peptide that functionally abrogates neutrophil recruitment to these organs. Using biochemical, genetic, and confocal intravital imaging approaches, we identified dipeptidase-1 (DPEP1) as the target and established its role as a physical adhesion receptor for neutrophil sequestration independent of its enzymatic activity. Importantly, genetic ablation or functional peptide blocking of DPEP1 significantly reduced neutrophil recruitment to the lungs and liver and provided improved survival in models of endotoxemia. Our data establish DPEP1 as a major adhesion receptor on the lung and liver endothelium and identify a therapeutic target for neutrophil-driven inflammatory diseases of the lungs.


Subject(s)
Dipeptidases/metabolism , Neutrophils/physiology , Platelet Glycoprotein GPIb-IX Complex/metabolism , Animals , Cilastatin/pharmacology , Cilastatin/therapeutic use , Dipeptidases/antagonists & inhibitors , Dipeptidases/genetics , Disease Models, Animal , Endotoxemia/mortality , Endotoxemia/pathology , Endotoxemia/prevention & control , GPI-Linked Proteins/antagonists & inhibitors , GPI-Linked Proteins/genetics , GPI-Linked Proteins/metabolism , Humans , Lipopolysaccharides/pharmacology , Liver/drug effects , Liver/immunology , Liver/metabolism , Lung/drug effects , Lung/immunology , Lung/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, SCID , Neutrophil Infiltration/drug effects , Peptides/chemical synthesis , Peptides/chemistry , Peptides/pharmacology , Survival Rate
2.
J Immunol ; 207(11): 2799-2812, 2021 12 01.
Article in English | MEDLINE | ID: mdl-34740957

ABSTRACT

Absent in melanoma-2 (AIM2) is an inflammasome-forming innate immune sensor for dsDNA but also exhibits inflammasome-independent functions such as restricting cellular proliferation. AIM2 is expressed in the kidney, but its localization and function are not fully characterized. In normal human glomeruli, AIM2 localized to podocytes. In patients with glomerulonephritis, AIM2 expression increased in CD44+-activated parietal epithelial cells within glomerular crescents. To explore AIM2 effects in glomerular disease, studies in Aim2 -/- mice were performed. Aim2-/- glomeruli showed reduced expression of Wilm tumor gene-1 (WT1), WT1-driven podocyte genes, and increased proliferation in outgrowth assays. In a nephrotoxic serum (NTS)-induced glomerulonephritis model, Aim2-/- (B6) mice exhibited more severe glomerular crescent formation, tubular injury, inflammation, and proteinuria compared with wild-type controls. Inflammasome activation markers were absent in both Aim2 -/- and wild-type kidneys, despite an increased inflammatory transcriptomic signature in Aim2 -/- mice. Aim2 -/- mice also demonstrated dysregulated cellular proliferation and an increase in CD44+ parietal epithelial cells during glomerulonephritis. The augmented inflammation and epithelial cell proliferation in Aim2 -/- (B6) mice was not due to genetic background, as Aim2 -/- (B6.129) mice demonstrated a similar phenotype during NTS glomerulonephritis. The AIM2-like receptor (ALR) locus was necessary for the inflammatory glomerulonephritis phenotype observed in Aim2 -/- mice, as NTS-treated ALR -/- mice displayed equal levels of injury as wild-type controls. Podocyte outgrowth from ALR -/- glomeruli was still increased, however, confirming that the ALR locus is dispensable for AIM2 effects on epithelial cell proliferation. These results identify a noncanonical role for AIM2 in suppressing inflammation and epithelial cell proliferation during glomerulonephritis.


Subject(s)
DNA-Binding Proteins/immunology , Epithelial Cells/immunology , Glomerulonephritis/immunology , Inflammation/immunology , Animals , Cell Proliferation , DNA-Binding Proteins/deficiency , Female , Glomerulonephritis/pathology , Humans , Mice , Mice, Inbred C57BL , Mice, Knockout
3.
Proc Natl Acad Sci U S A ; 117(9): 4921-4930, 2020 03 03.
Article in English | MEDLINE | ID: mdl-32071223

ABSTRACT

Antibiotic-resistant superbug bacteria represent a global health problem with no imminent solutions. Here we demonstrate that the combination (termed AB569) of acidified nitrite (A-NO2-) and Na2-EDTA (disodium ethylenediaminetetraacetic acid) inhibited all Gram-negative and Gram-positive bacteria tested. AB569 was also efficacious at killing the model organism Pseudomonas aeruginosa in biofilms and in a murine chronic lung infection model. AB569 was not toxic to human cell lines at bactericidal concentrations using a basic viability assay. RNA-Seq analyses upon treatment of P. aeruginosa with AB569 revealed a catastrophic loss of the ability to support core pathways encompassing DNA, RNA, protein, ATP biosynthesis, and iron metabolism. Electrochemical analyses elucidated that AB569 produced more stable SNO proteins, potentially explaining one mechanism of bacterial killing. Our data implicate that AB569 is a safe and effective means to kill pathogenic bacteria, suggesting that simple strategies could be applied with highly advantageous therapeutic/toxicity index ratios to pathogens associated with a myriad of periepithelial infections and related disease scenarios.


Subject(s)
Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/pharmacology , Bacteria/drug effects , Edetic Acid/pharmacology , Sodium Nitrite/pharmacology , Animals , Anti-Bacterial Agents/therapeutic use , Biofilms/drug effects , Disease Models, Animal , Down-Regulation , Drug Resistance, Bacterial/drug effects , Edetic Acid/chemistry , Lung Diseases/drug therapy , Lung Diseases/microbiology , Metabolic Networks and Pathways , Mice , Nitrites/chemistry , Nitrites/pharmacology , Pseudomonas aeruginosa/drug effects
4.
Am J Transplant ; 21(7): 2590-2595, 2021 07.
Article in English | MEDLINE | ID: mdl-33624432

ABSTRACT

Solid organ transplant recipients are vulnerable to severe infection during induction therapy. We report a case of a 67-year-old male who died unexpectedly 10 days after receiving a kidney transplant on February 10, 2020. There was no clear cause of death, but COVID-19 was considered retrospectively, as the death occurred shortly after the first confirmed case of COVID-19 in Canada. We confirmed the presence of SARS-CoV-2 components in the renal allograft and native lung tissue using immunohistochemistry for SARS-CoV-2 spike protein and RNA scope in situ hybridization for SARS-CoV-2 RNA. Results were reaffirmed with the Food and Drug Administration Emergency Use Authorization approved Bio-Rad SARS-CoV-2 digital droplet PCR for the kidney specimen. Our case highlights the importance of patient autopsies in an unfolding global pandemic and demonstrates the utility of molecular assays to diagnose SARS-CoV-2 post-mortem. SARS-CoV-2 infection during induction therapy may portend a fatal clinical outcome. We also suggest COVID-19 may be transmittable via renal transplant.


Subject(s)
COVID-19 , Kidney Transplantation , Aged , Autopsy , Canada , Humans , Kidney Transplantation/adverse effects , Male , RNA, Viral/genetics , Retrospective Studies , SARS-CoV-2 , Spike Glycoprotein, Coronavirus , Transplant Recipients
5.
Kidney Blood Press Res ; 45(2): 233-248, 2020.
Article in English | MEDLINE | ID: mdl-32062660

ABSTRACT

INTRODUCTION: Diabetic nephropathy (DN) and hypertensive nephrosclerosis (HN) represent the most common causes of chronic kidney disease (CKD) and many patients progress to -end-stage renal disease. Patients are treated primarily through the management of cardiovas-cular risk factors and hypertension; however patients with HN have a more favorable outcome. A noninvasive clinical approach to separate these two entities, especially in hypertensive patients who also have diabetes, would allow for targeted treatment and more appropriate resource allocation to those patients at the highest risk of CKD progression. Meth-ods: In this preliminary study, high-spatial-resolution matrix-assisted laser desorption/ion-ization (MALDI) mass spectrometry imaging (MSI) was integrated with high-mass accuracy MALDI-FTICR-MS and nLC-ESI-MS/MS analysis in order to detect tissue proteins within kidney biopsies to discriminate cases of DN (n = 9) from cases of HN (n = 9). RESULTS: Differences in the tryptic peptide profiles of the 2 groups could clearly be detected, with these becoming even more evident in the more severe histological classes, even if this was not evident with routine histology. In particular, 4 putative proteins were detected and had a higher signal intensity within regions of DN tissue with extensive sclerosis or fibrosis. Among these, 2 proteins (PGRMC1 and CO3) had a signal intensity that increased at the latter stages of the disease and may be associated with progression. DISCUSSION/CONCLUSION: This preliminary study represents a valuable starting point for a future study employing a larger cohort of patients to develop sensitive and specific protein biomarkers that could reliably differentiate between diabetic and hypertensive causes of CKD to allow for improved diagnosis, fewer biopsy procedures, and refined treatment approaches for clinicians.


Subject(s)
Diabetic Nephropathies/diagnostic imaging , Hypertension, Renal/diagnostic imaging , Nephritis/diagnostic imaging , Proteomics/methods , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization/methods , Aged , Female , Humans , Male , Middle Aged
6.
J Am Soc Nephrol ; 2024 Jun 14.
Article in English | MEDLINE | ID: mdl-38875013
7.
Mol Pharmacol ; 96(2): 259-271, 2019 08.
Article in English | MEDLINE | ID: mdl-31182542

ABSTRACT

Abnormal cardiac electrical activity is a common side effect caused by unintended block of the promiscuous drug target human ether-à-go-go-related gene (hERG1), the pore-forming domain of the delayed rectifier K+ channel in the heart. hERG1 block leads to a prolongation of the QT interval, a phase of the cardiac cycle that underlies myocyte repolarization detectable on the electrocardiogram. Even newly released drugs such as heart-rate lowering agent ivabradine block the rapid delayed rectifier current IKr, prolong action potential duration, and induce potentially lethal arrhythmia known as torsades de pointes. In this study, we describe a critical drug-binding pocket located at the lateral pore surface facing the cellular membrane. Mutations of the conserved M651 residue alter ivabradine-induced block but not by the common hERG1 blocker dofetilide. As revealed by molecular dynamics simulations, binding of ivabradine to a lipophilic pore access site is coupled to a state-dependent reorientation of aromatic residues F557 and F656 in the S5 and S6 helices. We show that the M651 mutation impedes state-dependent dynamics of F557 and F656 aromatic cassettes at the protein-lipid interface, which has a potential to disrupt drug-induced block of the channel. This fundamentally new mechanism coupling the channel dynamics and small-molecule access from the membrane into the hERG1 intracavitary site provides a simple rationale for the well established state-dependence of drug blockade. SIGNIFICANCE STATEMENT: The drug interference with the function of the cardiac hERG channels represents one of the major sources of drug-induced heart disturbances. We found a novel and a critical drug-binding pocket adjacent to a lipid-facing surface of the hERG1 channel, which furthers our molecular understanding of drug-induced QT syndrome.


Subject(s)
Ether-A-Go-Go Potassium Channels/chemistry , Ether-A-Go-Go Potassium Channels/metabolism , Ivabradine/pharmacology , Membrane Lipids/metabolism , Binding Sites , Ether-A-Go-Go Potassium Channels/genetics , Humans , Ivabradine/chemistry , Models, Molecular , Molecular Docking Simulation , Molecular Dynamics Simulation , Mutagenesis, Site-Directed , Phenethylamines/pharmacology , Protein Binding , Protein Structure, Tertiary , Sulfonamides/pharmacology
8.
J Pharmacol Exp Ther ; 370(1): 44-53, 2019 07.
Article in English | MEDLINE | ID: mdl-31004077

ABSTRACT

The pregnane X receptor (PXR) is a ligand-activated nuclear receptor that acts as a xenobiotic sensor, responding to compounds of foreign origin, including pharmaceutical compounds, environmental contaminants, and natural products, to induce transcriptional events that regulate drug detoxification and efflux pathways. As such, the PXR is thought to play a key role in protecting the host from xenobiotic exposure. More recently, the PXR has been reported to regulate the expression of innate immune receptors in the intestine and modulate inflammasome activation in the vasculature. In the current study, we report that activation of the PXR in primed macrophages triggers caspase-1 activation and interleukin-1ß release. Mechanistically, we show that this response is nucleotide-binding oligomerization domain, leucine-rich repeat, and pyrin domain-containing 3-dependent and is driven by the rapid efflux of ATP and P2X purinoceptor 7 activation following PXR stimulation, an event that involves pannexin-1 gating, and is sensitive to inhibition of Src-family kinases. Our findings identify a mechanism whereby the PXR drives innate immune signaling, providing a potential link between xenobiotic exposure and the induction of innate inflammatory responses.


Subject(s)
Adenosine Triphosphate/metabolism , Inflammasomes/metabolism , Macrophages/drug effects , Macrophages/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Pregnane X Receptor/metabolism , Animals , Caspase 1/metabolism , Cell Line, Tumor , Connexins/metabolism , Enzyme Activation/drug effects , Humans , Interleukin-1beta/metabolism , Kinetics , Ligands , Mice , Nerve Tissue Proteins/metabolism , Pregnane X Receptor/agonists , Receptors, Purinergic P2X7/metabolism , src-Family Kinases/metabolism
9.
Nat Rev Neurosci ; 15(2): 84-97, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24399084

ABSTRACT

Microglia and macrophages in the CNS contain multimolecular complexes termed inflammasomes. Inflammasomes function as intracellular sensors for infectious agents as well as for host-derived danger signals that are associated with neurological diseases, including meningitis, stroke and Alzheimer's disease. Assembly of an inflammasome activates caspase 1 and, subsequently, the proteolysis and release of the cytokines interleukin-1ß and interleukin-18, as well as pyroptotic cell death. Since the discovery of inflammasomes in 2002, there has been burgeoning recognition of their complexities and functions. Here, we review the current understanding of the functions of different inflammasomes in the CNS and their roles in neurological diseases.


Subject(s)
Central Nervous System/metabolism , Inflammasomes/metabolism , Animals , Central Nervous System/cytology , Humans , Macrophages/metabolism , Nervous System Diseases/metabolism , Nervous System Diseases/pathology , Neuroglia/metabolism
10.
Arch Biochem Biophys ; 670: 4-14, 2019 07 30.
Article in English | MEDLINE | ID: mdl-30772258

ABSTRACT

The NOD-like receptor (NLR) family of proteins is a group of pattern recognition receptors (PRRs) known to mediate the initial innate immune response to cellular injury and stress. The NLRP proteins represent a fourteen-member subset of the NLR family that contains an N-terminal pyrin domain. Some NLRs are known to form multi-protein complexes known as inflammasomes. Inflammasomes consist of an NLR, the adaptor protein ASC, and the effector molecule pro-caspase-1. Once activated, these inflammasomes facilitate the cleavage and activation of caspase-1, which in turn mediates the cleavage of the pro-inflammatory cytokines IL-1ß and IL-18 into their active and secreted forms. Activated caspase-1 also drives the cleavage of gasdermin D, which triggers an inflammatory form of cell death known as pyroptosis. Several NLRs are also known to possess non-canonical, inflammasome-independent functions, regulating a variety of signaling pathways. In this review, a thorough overview of both inflammasome-dependent and -independent NLR signaling will be presented, with highlights from the field as well as promising future directions and postulates based on the known science.


Subject(s)
Inflammasomes/metabolism , NLR Proteins/metabolism , Signal Transduction , Humans , Pyroptosis
11.
Arch Biochem Biophys ; 670: 104-115, 2019 07 30.
Article in English | MEDLINE | ID: mdl-30641048

ABSTRACT

The NLRP proteins are a subfamily of the NOD-like receptor (NLR) innate immune sensors that possess an ATP-binding NACHT domain. As the most well studied member, NLRP3 can initiate the assembly process of a multiprotein complex, termed the inflammasome, upon detection of a wide range of microbial products and endogenous danger signals and results in the activation of pro-caspase-1, a cysteine protease that regulates multiple host defense pathways including cytokine maturation. Dysregulated NLRP3 activation contributes to inflammation and the pathogenesis of several chronic diseases, and the ATP-binding properties of NLRPs are thought to be critical for inflammasome activation. In light of this, we examined the utility of immobilized ATP matrices in the study of NLRP inflammasomes. Using NLRP3 as the prototypical member of the family, P-linked ATP Sepharose was determined to be a highly-effective capture agent. In subsequent examinations, P-linked ATP Sepharose was used as an enrichment tool to enable the effective profiling of NLRP3-biomarker signatures with selected reaction monitoring-mass spectrometry (SRM-MS). Finally, ATP Sepharose was used in combination with a fluorescence-linked enzyme chemoproteomic strategy (FLECS) screen to identify potential competitive inhibitors of NLRP3. The identification of a novel benzo[d]imidazol-2-one inhibitor that specifically targets the ATP-binding and hydrolysis properties of the NLRP3 protein implies that ATP Sepharose and FLECS could be applied other NLRPs as well.


Subject(s)
Adenosine Triphosphate/metabolism , Inflammasomes/metabolism , NLR Proteins/metabolism , HEK293 Cells , Humans , Phosphorylation , Protein Processing, Post-Translational , Ubiquitination
12.
BMC Nephrol ; 20(1): 29, 2019 01 31.
Article in English | MEDLINE | ID: mdl-30704432

ABSTRACT

BACKGROUND: Anti-glomerular basement membrane (anti-GBM) disease is characterized by circulating IgG glomerular basement membrane antibodies and is clinically expressed as a rapidly progressive crescentic glomerulonephritis (GN), with 30-60% of patients also developing pulmonary hemorrhage. Classically, the renal biopsy shows glomerular crescent formation, bright linear staining of glomerular basement membranes (GBM) for IgG on direct immunofluorescence (IF), and the serologic presence of circulating anti-GBM antibodies. Recently, patients with linear IgG IF staining, undetectable circulating anti-GBM antibodies and glomerular changes atypical for anti-GBM disease have been described as "atypical anti-GBM disease", with a distinctly more benign clinical course than typical anti-GBM disease. We present a case report of a patient with negative anti-GBM serology but positive linear IgG staining by IF, severe diffuse crescentic and endocapillary proliferative glomerulonephritis, and renal failure, complicated by severe pulmonary hemorrhage after immunosuppression, likely due to cytomegalovirus (CMV) pneumonitis. CASE PRESENTATION: A 24-year-old man was admitted to hospital with hemoptysis and renal failure. Investigations for anti-GBM serology by addressable laser bead immunoassay (ALBIA) was negative for anti-GBM antibodies. Renal biopsy showed diffuse endocapillary proliferative glomerulonephritis with membranoproliferative features and diffuse circumferential crescents. Direct IF showed strong linear staining for IgG along GBMs. The patient's hemoptysis improved with immunosuppression, but 1 month later he was readmitted with gross hemoptysis, which was refractory to further cyclophosphamide, plasma exchange and rituximab. Bronchoalveolar lavage (BAL) and blood work confirmed CMV pneumonitis, and the patient's hemoptysis resolved with ganciclovir, though he became dialysis dependent. CONCLUSIONS: This case demonstrates an atypical presentation of anti-GBM disease with both crescents and endocapillary hypercellularity and negative serology. The patient is dialysis dependent, unlike most previously described patients with atypical anti-GBM disease. The course was complicated by CMV pneumonitis, which contributed to the severity of the pulmonary manifestations and added diagnostic difficulty.


Subject(s)
Anti-Glomerular Basement Membrane Disease/complications , Cytomegalovirus Infections/complications , Hemoptysis/etiology , Pneumonia, Viral/complications , Viremia/complications , Anti-Glomerular Basement Membrane Disease/therapy , Antiviral Agents/therapeutic use , Autoantibodies/analysis , Combined Modality Therapy , Cyclophosphamide/therapeutic use , Cytomegalovirus Infections/diagnosis , Cytomegalovirus Infections/drug therapy , Delayed Diagnosis , Disease Progression , Ganciclovir/therapeutic use , Hemorrhage/etiology , Humans , Immunoglobulin G/analysis , Kidney Glomerulus/chemistry , Kidney Glomerulus/immunology , Lung Diseases/etiology , Male , Plasma , Plasma Exchange , Pneumonia, Viral/drug therapy , Recurrence , Viremia/diagnosis , Viremia/drug therapy , Young Adult
13.
J Am Soc Nephrol ; 29(4): 1165-1181, 2018 04.
Article in English | MEDLINE | ID: mdl-29439156

ABSTRACT

Nonmicrobial inflammation contributes to CKD progression and fibrosis. Absent in melanoma 2 (AIM2) is an inflammasome-forming receptor for double-stranded DNA. AIM2 is expressed in the kidney and activated mainly by macrophages. We investigated the potential pathogenic role of the AIM2 inflammasome in kidney disease. In kidneys from patients with diabetic or nondiabetic CKD, immunofluorescence showed AIM2 expression in glomeruli, tubules, and infiltrating leukocytes. In a mouse model of unilateral ureteral obstruction (UUO), Aim2 deficiency attenuated the renal injury, fibrosis, and inflammation observed in wild-type (WT) littermates. In bone marrow chimera studies, UUO induced substantially more tubular injury and IL-1ß cleavage in Aim2-/- or WT mice that received WT bone marrow than in WT mice that received Aim2-/- bone marrow. Intravital microscopy of the kidney in LysM(gfp/gfp) mice 5-6 days after UUO demonstrated the significant recruitment of GFP+ proinflammatory macrophages that crawled along injured tubules, engulfed DNA from necrotic cells, and expressed active caspase-1. DNA uptake occurred in large vacuolar structures within recruited macrophages but not resident CX3CR1+ renal phagocytes. In vitro, macrophages that engulfed necrotic debris showed AIM2-dependent activation of caspase-1 and IL-1ß, as well as the formation of AIM2+ ASC specks. ASC specks are a hallmark of inflammasome activation. Cotreatment with DNaseI attenuated the increase in IL-1ß levels, confirming that DNA was the principal damage-associated molecular pattern in this process. Therefore, the activation of the AIM2 inflammasome by DNA from necrotic cells drives a proinflammatory phenotype that contributes to chronic injury in the kidney.


Subject(s)
DNA-Binding Proteins/physiology , DNA/metabolism , Inflammasomes/physiology , Macrophages/physiology , Renal Insufficiency, Chronic/metabolism , Animals , Bone Marrow Transplantation , Caspase 1/metabolism , DNA-Binding Proteins/deficiency , DNA-Binding Proteins/genetics , Diabetic Nephropathies/metabolism , Enzyme Activation , Fibrosis , Humans , Interleukin-1beta/metabolism , Kidney Glomerulus/metabolism , Kidney Tubules/metabolism , Leukocytes/metabolism , Male , Mice, Inbred C57BL , Mice, Knockout , Necrosis , Nephrosclerosis/metabolism , Phagocytosis , Phenotype , Radiation Chimera , THP-1 Cells , Ureteral Obstruction/metabolism , Ureteral Obstruction/pathology
14.
BMC Nephrol ; 18(1): 252, 2017 Jul 26.
Article in English | MEDLINE | ID: mdl-28747168

ABSTRACT

BACKGROUND: Advances in technology and the ability to interrogate disease pathogenesis using systems biology approaches are exploding. As exemplified by the substantial progress in the personalized diagnosis and treatment of cancer, the application of systems biology to enable precision medicine in other disciplines such as Nephrology is well underway. Infrastructure that permits the integration of clinical data, patient biospecimens and advanced technologies is required for institutions to contribute to, and benefit from research in molecular disease classification and to devise specific and patient-oriented treatments. METHODS AND RESULTS: We describe the establishment of the Biobank for the Molecular Classification of Kidney Disease (BMCKD) at the University of Calgary, Alberta, Canada. The BMCKD consists of a fully equipped wet laboratory, an information technology infrastructure, and a formal operational, ethical and legal framework for banking human biospecimens and storing clinical data. The BMCKD first consolidated a large retrospective cohort of kidney biopsy specimens to create a population-based renal pathology database and tissue inventory of glomerular and other kidney diseases. The BMCKD will continue to prospectively bank all kidney biopsies performed in Southern Alberta. The BMCKD is equipped to perform molecular, clinical and epidemiologic studies in renal pathology. The BMCKD also developed formal biobanking procedures for human specimens such as blood, urine and nucleic acids collected for basic and clinical research studies or for advanced diagnostic technologies in clinical care. The BMCKD is guided by standard operating procedures, an ethics framework and legal agreements with stakeholders that include researchers, data custodians and patients. The design and structure of the BMCKD permits its inclusion in a wide variety of research and clinical activities. CONCLUSION: The BMCKD is a core multidisciplinary facility that will bridge basic and clinical research and integrate precision medicine into renal pathology and nephrology.


Subject(s)
Biological Specimen Banks/standards , Kidney Diseases/pathology , Kidney/pathology , Nephrology/standards , Precision Medicine/standards , Translational Research, Biomedical/standards , Cohort Studies , Female , Humans , Kidney Diseases/classification , Male , Nephrology/methods , Precision Medicine/methods , Retrospective Studies , Translational Research, Biomedical/methods
15.
Mediators Inflamm ; 2016: 5637685, 2016.
Article in English | MEDLINE | ID: mdl-27610005

ABSTRACT

The pathogenesis of Crohn's disease (CD) involves defects in the innate immune system, impairing responses to microbes. Studies have revealed that mutations NLRP3 are associated with CD. We reported previously that Nlrp3-/- mice were more susceptible to colitis and exhibited reduced colonic IL-10 expression. In the current study, we sought to determine how the loss of NLRP3 might be altering the function of regulatory T cells, a major source of IL-10. Colitis was induced in wild-type (WT) and Nlrp3-/- mice by treatment with dextran sulphate sodium (DSS). Lamina propria (LP) cells were assessed by flow cytometry and cytokine expression was assessed. DSS-treated Nlrp3-/- mice exhibited increased numbers of colonic foxp3+ T cells that expressed significantly lower levels of IL-10 but increased IL-17. This was associated with increased expression of colonic IL-15 and increased surface expression of IL-15 on LP dendritic cells. Neutralizing IL-15 in Nlrp3-/- mice attenuated the severity of colitis, decreased the number of colonic foxp3+ cells, and reduced the colonic expression of IL-12p40 and IL-17. These data suggest that the NLRP3 inflammasome can regulate intestinal inflammation through noncanonical mechanisms, providing additional insight as to how NLRP3 variants may contribute to the pathogenesis of CD.


Subject(s)
Colitis/metabolism , Cytokines/metabolism , Forkhead Transcription Factors/metabolism , Interleukin-15/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/deficiency , Animals , Colitis/immunology , Colitis/pathology , Dendritic Cells/metabolism , Flow Cytometry , Inflammasomes/metabolism , Interleukin-10/metabolism , Interleukin-17/metabolism , Male , Mice , Mice, Knockout , NLR Family, Pyrin Domain-Containing 3 Protein/genetics , Peroxidase/metabolism
16.
BMC Med Ethics ; 17(1): 48, 2016 08 15.
Article in English | MEDLINE | ID: mdl-27527514

ABSTRACT

BACKGROUND: Personal health information and biospecimens are valuable research resources essential for the advancement of medicine and protected by national standards and provincial statutes. Research ethics and privacy standards attempt to balance individual interests with societal interests. However these standards may not reflect public opinion or preferences. The purpose of this study was to assess the opinions and preferences of patients with kidney disease about the use of their health information and biospecimens for medical research. METHODS: A 45-item survey was distributed to a convenience sample of patients at an outpatient clinic in a large urban centre. The survey briefly addressed sociodemographic and illness characteristics. Opinions were sought on the research use of health information and biospecimens including consent preferences. RESULTS: Two hundred eleven of 400 distributed surveys were completed (response rate 52.8 %). Respondents were generally supportive of medical research and trusting of researchers. Many respondents supported the use of their information and biospecimens for health research and also preferred consent be sought for use of health information and biospecimens. Some supported the use of their information and biospecimens for research without consent. There were significant differences in the opinions people offered regarding the research use of biospecimens compared to health information. Some respondent perspectives about consent were at odds with current regulatory and legal standards. CONCLUSIONS: Clinical health data and biospecimens are valuable research resources, critical to the advancement of medicine. Use of these data for research requires balancing respect for individual autonomy, privacy and the societal interest in the greater good. Incongruence between some respondent perspectives and the regulatory standards suggest both a need for public education and review of legislation to increase understanding and ensure the public's trust is maintained.


Subject(s)
Attitude to Health , Biomedical Research , Ethics, Research , Informed Consent , Privacy , Research Design , Adult , Aged , Biomedical Research/ethics , Biomedical Research/methods , Cells , Female , Health Services Needs and Demand , Humans , Male , Medical Records , Middle Aged , Patients , Personal Autonomy , Specimen Handling , Trust
17.
J Biol Chem ; 289(28): 19571-84, 2014 Jul 11.
Article in English | MEDLINE | ID: mdl-24841199

ABSTRACT

Nucleotide-binding domain and leucine-rich repeat containing PYD-3 (NLRP3) is a pattern recognition receptor that is implicated in the pathogenesis of inflammation and chronic diseases. Although much is known regarding the NLRP3 inflammasome that regulates proinflammatory cytokine production in innate immune cells, the role of NLRP3 in non-professional immune cells is unclear. Here we report that NLRP3 is expressed in cardiac fibroblasts and increased during TGFß stimulation. NLRP3-deficient cardiac fibroblasts displayed impaired differentiation and R-Smad activation in response to TGFß. Only the central nucleotide binding domain of NLRP3 was required to augment R-Smad signaling because the N-terminal Pyrin or C-terminal leucine-rich repeat domains were dispensable. Interestingly, NLRP3 regulation of myofibroblast differentiation proceeded independently from the inflammasome, IL-1ß/IL-18, or caspase 1. Instead, mitochondrially localized NLRP3 potentiated reactive oxygen species to augment R-Smad activation. In vivo, NLRP3-deficient mice were protected against angiotensin II-induced cardiac fibrosis with preserved cardiac architecture and reduced collagen 1. Together, these results support a distinct role for NLRP3 in non-professional immune cells independent from the inflammasome to regulate differential aspects of wound healing and chronic disease.


Subject(s)
Carrier Proteins/metabolism , Inflammasomes , Mitochondrial Proteins/metabolism , Myocardium/metabolism , Myofibroblasts/metabolism , Reactive Oxygen Species/metabolism , Signal Transduction , Smad Proteins, Receptor-Regulated/metabolism , Angiotensin II/adverse effects , Angiotensin II/pharmacology , Animals , Carrier Proteins/genetics , Collagen Type I/biosynthesis , Collagen Type I/genetics , Fibrosis , Heart Diseases/chemically induced , Heart Diseases/genetics , Heart Diseases/metabolism , Heart Diseases/pathology , Interleukin-18/genetics , Interleukin-18/metabolism , Interleukin-1beta/genetics , Interleukin-1beta/metabolism , Mice , Mice, Knockout , Mitochondrial Proteins/genetics , Myocardium/pathology , Myofibroblasts/pathology , NLR Family, Pyrin Domain-Containing 3 Protein , Smad Proteins, Receptor-Regulated/genetics , Transforming Growth Factor beta/genetics , Transforming Growth Factor beta/metabolism , Vasoconstrictor Agents/adverse effects , Vasoconstrictor Agents/pharmacology
18.
J Cell Biochem ; 116(5): 711-20, 2015 May.
Article in English | MEDLINE | ID: mdl-25639477

ABSTRACT

Vitamin D3 has emerged as an important regulator of the immune system. With metabolic enzymes for vitamin D3 activation and vitamin D receptors (VDR) now identified in a variety of immune cells, the active vitamin D3 metabolite 1,25(OH)2D3, is thought to possess immunomodulatory properties. We examined whether 1,25(OH)2D3 might also enhance the NLRP3-dependent release of mature IL-1ß from macrophages. PMA-differentiated THP-1 cells were stimulated with vitamin D3 metabolites and assessed for CYP27, CYP24, NLRP3, ASC, pro-caspase-1 expression by western blot and real-time qPCR as well as inflammasome activation with pro-inflammatory cytokine IL-1ß release measured by ELISA. Exposure to 1,25(OH)2D3 had no effect on the basal expression levels of VDR; however, CYP27A1 transcript was suppressed and CYP24A1 transcript was substantively elevated. Both 1,25(OH)2D3 - and 25(OH)D3 induced IL-1ß release from THP-1 cells, and these effects were blocked with application of the caspase-1 inhibitor YVAD and the NLRP3 inhibitors glyburide and Bay 11-7082. Interestingly, 1,25 (OH)2D3 exposure reduced NLRP3 protein expression but had no effect on ASC or pro-caspase-1 protein levels. The increase in mature IL-1ß elicited by 1,25(OH)2D3 was modest compared to that found for ATP or C. difficile toxins. However, co-treatment of THP-1 cells with ATP and 1,25(OH)2D3 resulted in more IL-1ß secretion than ATP or 1,25(OH)2D3 alone.


Subject(s)
Cholecalciferol/metabolism , Interleukin-1beta/metabolism , Monocytes/metabolism , Amino Acid Chloromethyl Ketones/pharmacology , Carrier Proteins/antagonists & inhibitors , Carrier Proteins/metabolism , Caspase 1/metabolism , Caspase Inhibitors/pharmacology , Cholecalciferol/chemistry , Cholestanetriol 26-Monooxygenase/metabolism , Glyburide/pharmacology , Humans , Monocytes/drug effects , NLR Family, Pyrin Domain-Containing 3 Protein , Nitriles/pharmacology , Receptors, Calcitriol/metabolism , Sulfones/pharmacology
19.
J Immunol ; 190(3): 1239-49, 2013 Feb 01.
Article in English | MEDLINE | ID: mdl-23264657

ABSTRACT

Tubulointerstitial inflammation and fibrosis are strongly associated with the outcome of chronic kidney disease. We recently demonstrated that the NOD-like receptor, pyrin domain containing-3 (NLRP3) contributes to renal inflammation, injury, and fibrosis following unilateral ureteric obstruction in mice. NLRP3 expression in renal tubular epithelial cells (TECs) was found to be an important component of experimental disease pathogenesis, although the biology of NLRP3 in epithelial cells is unknown. In human and mouse primary renal TECs, NLRP3 expression was increased in response to TGF-ß1 stimulation and associated with epithelial-mesenchymal transition (EMT) and the expression of α-smooth muscle actin (αSMA) and matrix metalloproteinase (MMP) 9. TGF-ß1-induced EMT and the induction of MMP-9 and αSMA were significantly decreased in mouse Nlrp3(-/-) renal TECs, suggesting a role for Nlrp3 in TGF-ß-dependent signaling. Although apoptosis-associated speck-like protein containing a CARD domain(-/-) TECs demonstrated a phenotype similar to that of Nlrp3(-/-) cells in response to TGF-ß1, the effect of Nlrp3 on MMP-9 and αSMA expression was inflammasome independent, as IL-1ß, IL-18, MyD88, and caspase-1 were dispensable. Smad2 and Smad3 phosphorylation in response to TGF-ß1 was attenuated in Nlrp3(-/-) and apoptosis-associated speck-like protein containing a CARD domain(-/-) cells, accounting for the dampened EMT and TGF-ß1 responsiveness in these cells. Consistent with these findings, overexpression of NLRP3 in 293T cells resulted in increased Smad3 phosphorylation and activity. Taken together, these data support a novel and direct role for NLRP3 in promoting TGF-ß signaling and R-Smad activation in epithelial cells independent of the inflammasome.


Subject(s)
Carrier Proteins/physiology , Epithelial Cells/immunology , Epithelial-Mesenchymal Transition/physiology , Inflammasomes/physiology , Kidney Tubules, Proximal/immunology , Signal Transduction/immunology , Transforming Growth Factor beta1/physiology , Animals , Caspase 1/metabolism , Cells, Cultured/drug effects , Cells, Cultured/metabolism , Cytokines/metabolism , Gene Expression Regulation , Humans , Interleukin-1beta/pharmacology , Kidney Tubules, Proximal/metabolism , Matrix Metalloproteinases/biosynthesis , Matrix Metalloproteinases/genetics , Mice , Mice, Inbred C57BL , NLR Family, Pyrin Domain-Containing 3 Protein , Nephritis, Interstitial/immunology , Nephritis, Interstitial/pathology , Real-Time Polymerase Chain Reaction , Smad Proteins/metabolism , Transforming Growth Factor beta1/pharmacology
20.
J Biol Chem ; 288(52): 37319-31, 2013 Dec 27.
Article in English | MEDLINE | ID: mdl-24253040

ABSTRACT

Chronic kidney diseases cause significant morbidity and mortality in the population. During renal injury, kidney-localized proteinases can signal by cleaving and activating proteinase-activated receptor-2 (PAR2), a G-protein-coupled receptor involved in inflammation and fibrosis that is highly expressed in renal tubular cells. Following unilateral ureteric obstruction, PAR2-deficient mice displayed reduced renal tubular injury, fibrosis, collagen synthesis, connective tissue growth factor (CTGF), and α-smooth muscle actin gene expression at 7 days, compared with wild-type controls. In human proximal tubular epithelial cells in vitro, PAR2 stimulation with PAR2-activating peptide (PAR2-AP) alone significantly up-regulated the expression of CTGF, a potent profibrotic cytokine. The induction of CTGF by PAR2-AP was synergistically increased when combined with transforming growth factor-ß (TGF-ß). Consistent with these findings, treating human proximal tubular epithelial cells with PAR2-AP induced Smad2/3 phosphorylation in the canonical TGF-ß signaling pathway. The Smad2 phosphorylation and CTGF induction required signaling via both the TGFß-receptor and EGF receptor suggesting that PAR2 utilizes transactivation mechanisms to initiate fibrogenic signaling. Taken together, our data support the hypothesis that PAR2 synergizes with the TGFß signaling pathway to contribute to renal injury and fibrosis.


Subject(s)
ErbB Receptors/biosynthesis , Kidney Diseases/metabolism , Receptor, PAR-2/metabolism , Receptors, Transforming Growth Factor beta/biosynthesis , Signal Transduction , Transcriptional Activation , Animals , Cells, Cultured , Connective Tissue Growth Factor/genetics , Connective Tissue Growth Factor/metabolism , ErbB Receptors/genetics , Female , Fibrosis/metabolism , Fibrosis/pathology , Humans , Kidney Diseases/pathology , Kidney Tubules, Proximal/metabolism , Kidney Tubules, Proximal/pathology , Male , Mice , Mice, Mutant Strains , Oligopeptides/pharmacology , Receptor, PAR-2/agonists , Receptor, PAR-2/genetics , Receptors, Transforming Growth Factor beta/genetics , Smad2 Protein/genetics , Smad2 Protein/metabolism , Smad3 Protein/genetics , Smad3 Protein/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL