Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 102
Filter
1.
BMC Nephrol ; 25(1): 55, 2024 Feb 14.
Article in English | MEDLINE | ID: mdl-38355500

ABSTRACT

BACKGROUND: The International Classification of Diseases (ICD) coding system is the industry standard tool for billing, disease classification, and epidemiology purposes. Prior research has demonstrated ICD codes to have poor accuracy, particularly in relation to rapidly progressing chronic kidney disease (CKD) patients. In 2016, the ICD system moved to revision 10. This study examines subjects in a large insurer database to determine the accuracy of ICD-10 CKD-staging codes to diagnose patients rapidly progressing towards end-stage kidney disease (ESKD). PATIENTS AND METHODS: Serial observations of outpatient serum creatinine measurements from 2016 to 2021 of 315,903 patients were transformed to estimated glomerular filtration rate (eGFR) to identify CKD stage-3 and advanced patients diagnosed clinically (eGFR-CKD). CKD-staging codes from the same time period of 59,386 patients and used to identify stage-3 and advanced patients diagnosed by ICD-code (ICD-CKD). eGFR-CKD and ICD-CKD diagnostic accuracy was compared between a total of 334,610 patients. RESULTS: 5,618 patients qualified for the progression analysis; 72 were identified as eGFR rapid progressors; 718 had multiple codes to qualify as ICD rapid progressors. Sensitivity was 5.56%, with positive predictive value (PPV) 5.6%. 34,858 patients were diagnosed as eGFR-CKD stage-3 patients; 17,549 were also diagnosed as ICD-CKD stage-3 patients, for a sensitivity of 50.34%, with PPV of 58.71%. 4,069 patients reached eGFR-CKD stage-4 with 2,750 ICD-CKD stage-4 patients, giving a sensitivity of 67.58%, PPV of 42.43%. 959 patients reached eGFR-CKD stage-5 with 566 ICD-CKD stage-5 patients, giving a sensitivity of 59.02%, PPV of 35.85%. CONCLUSION: This research shows that recent ICD revisions have not improved identification of rapid progressors in diagnostic accuracy, although marked increases in sensitivity for stage-3 (50.34% vs. 24.68%), and PPV in stage-3 (58.71% vs. 40.08%), stage-4 (42.43% vs. 18.52%), and stage-5 (35.85% vs. 4.51%) were observed. However, sensitivity in stage-5 compares poorly (59.02% vs. 91.05%).


Subject(s)
Kidney Failure, Chronic , Renal Insufficiency, Chronic , Humans , International Classification of Diseases , Renal Insufficiency, Chronic/diagnosis , Renal Insufficiency, Chronic/epidemiology , Kidney Failure, Chronic/diagnosis , Kidney Failure, Chronic/epidemiology , Glomerular Filtration Rate , Diagnostic Tests, Routine
2.
Immunology ; 170(2): 180-192, 2023 10.
Article in English | MEDLINE | ID: mdl-37222083

ABSTRACT

The complement system, an arm of the innate immune system plays a critical role in both health and disease. The complement system is highly complex with dual possibilities, helping or hurting the host, depending on the location and local microenvironment. The traditionally known functions of complement include surveillance, pathogen recognition, immune complex trafficking, processing and pathogen elimination. The noncanonical functions of the complement system include their roles in development, differentiation, local homeostasis and other cellular functions. Complement proteins are present in both, the plasma and on the membranes. Complement activation occurs both extra- and intracellularly, which leads to considerable pleiotropy in their activity. In order to design more desirable and effective therapies, it is important to understand the different functions of complement, and its location-based and tissue-specific responses. This manuscript will provide a brief overview into the complex nature of the complement cascade, outlining some of their complement-independent functions, their effects at different locale, and their implication in disease settings.


Subject(s)
Complement Activation , Complement System Proteins
3.
J Immunol Res ; 2022: 1737419, 2022.
Article in English | MEDLINE | ID: mdl-35097132

ABSTRACT

Complement factor H (FH) is a key regulator of the alternative pathway of complement, in man and mouse. Earlier, our studies revealed that the absence of FH causes the C57BL6 mouse to become susceptible to chronic serum sickness (CSS) along with an increase in the renal infiltration of macrophages compared to controls. To understand if the increased recruitment of macrophages (Mϕs) to the kidney was driving inflammation and propagating injury, we examined the effect of Mϕ depletion with clodronate in FH knockout mice with CSS. Eight-week-old FHKO mice were treated with apoferritin (4 mg/mouse) for 5 wks and with either vehicle (PBS) or clodronate (50 mg/kg ip, 3 times/wk for the last 3 weeks). The administration of clodronate decreased monocytes and Mϕs in the kidneys by >80%. Kidney function assessed by BUN and albumin remained closer to normal on depletion of Mϕs. Clodronate treatment prevented the alteration in cytokines, TNFα and IL-6, and increase in gene expression of connective tissue growth factor (CTGF), TGFß-1, matrix metalloproteinase-9 (MMP9), fibronectin, laminin, and collagen in FHKO mice with CSS (P < 0.05). Clodronate treatment led to relative protection from immune complex- (IC-) mediated disease pathology during CSS as assessed by the significantly reduced glomerular pathology (GN) and extracellular matrix. Our results suggest that complement activation is one of the mechanism that regulates the macrophage landscape and thereby fibrosis. The exact mechanism remains to be deciphered. In brief, our data shows that Mϕs play a critical role in FH-dependent ICGN and Mϕ depletion reduces disease progression.


Subject(s)
Glomerulonephritis/immunology , Immune Complex Diseases/immunology , Kidney/metabolism , Macrophages/immunology , Animals , Apoferritins/administration & dosage , Cell Movement , Clodronic Acid/administration & dosage , Complement Factor H/metabolism , Disease Progression , Fibrosis , Kidney/immunology , Kidney/pathology , Matrix Metalloproteinase 9/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout
4.
Kidney Int ; 100(4): 824-836, 2021 10.
Article in English | MEDLINE | ID: mdl-34139209

ABSTRACT

Factor H (FH) is a critical regulator of the alternative complement pathway and its deficiency or mutation underlie kidney diseases such as dense deposit disease. Since vascular dysfunction is an important facet of kidney disease, maintaining optimal function of the lining endothelial cells is important for vascular health. To investigate the molecular mechanisms that are regulated by FH in endothelial cells, FH deficient and sufficient mouse kidney endothelial cell cultures were established. Endothelial FH deficiency resulted in cytoskeletal remodeling, increased angiogenic potential, loss of cellular layer integrity and increased cell proliferation. FH reconstitution prevented these FH-dependent proliferative changes. Respiratory flux analysis showed reduced basal mitochondrial respiration, ATP production and maximal respiratory capacity in FH deficient endothelial cells, while proton leak remained unaltered. Similar changes were observed in FH deficient human glomerular endothelial cells indicating the translational potential of these studies. Gene expression analysis revealed that the FH-dependent gene changes in mouse kidney endothelial cells include significant upregulation of genes involved in inflammation and the complement system. The transcription factor nuclear factor-kB, that regulates many biological processes, was translocated from the cytoplasm to the nucleus in the absence of FH. Thus, our studies show the functional relevance of intrinsic FH in kidney endothelial cells in man and mouse.


Subject(s)
Complement Factor H , Kidney Diseases , Animals , Complement Factor H/genetics , Complement Pathway, Alternative , Endothelial Cells , Humans , Kidney , Mice
5.
JCI Insight ; 6(11)2021 06 08.
Article in English | MEDLINE | ID: mdl-33914709

ABSTRACT

Patients with chronic kidney disease (CKD) and end-stage renal disease suffer from increased cardiovascular events and cardiac mortality. Prior studies have demonstrated that a portion of this enhanced risk can be attributed to the accumulation of microbiota-derived toxic metabolites, with most studies focusing on the sulfonated form of p-cresol (PCS). However, unconjugated p-cresol (uPC) itself was never assessed due to rapid and extensive first-pass metabolism that results in negligible serum concentrations of uPC. These reports thus failed to consider the host exposure to uPC prior to hepatic metabolism. In the current study, not only did we measure the effect of altering the intestinal microbiota on lipid accumulation in coronary arteries, but we also examined macrophage lipid uptake and handling pathways in response to uPC. We found that atherosclerosis-prone mice fed a high-fat diet exhibited significantly higher coronary artery lipid deposits upon receiving fecal material from CKD mice. Furthermore, treatment with uPC increased total cholesterol, triglycerides, and hepatic and aortic fatty deposits in non-CKD mice. Studies employing an in vitro macrophage model demonstrated that uPC exposure increased apoptosis whereas PCS did not. Additionally, uPC exhibited higher potency than PCS to stimulate LDL uptake and only uPC induced endocytosis- and pinocytosis-related genes. Pharmacological inhibition of varying cholesterol influx and efflux systems indicated that uPC increased macrophage LDL uptake by activating macropinocytosis. Overall, these findings indicate that uPC itself had a distinct effect on macrophage biology that might have contributed to increased cardiovascular risk in patients with CKD.


Subject(s)
Aorta/metabolism , Cholesterol, LDL/metabolism , Coronary Artery Disease/metabolism , Cresols/metabolism , Gastrointestinal Microbiome , Liver/metabolism , Macrophages/metabolism , Pinocytosis/physiology , Renal Insufficiency, Chronic/metabolism , Animals , Aorta/drug effects , Aorta/pathology , Cholesterol/metabolism , Cholesterol, LDL/drug effects , Coronary Artery Disease/pathology , Coronary Vessels/drug effects , Coronary Vessels/metabolism , Coronary Vessels/pathology , Cresols/pharmacology , Diet, High-Fat , Fecal Microbiota Transplantation , Kidney Failure, Chronic/metabolism , Kidney Failure, Chronic/microbiology , Liver/drug effects , Liver/pathology , Macrophages/drug effects , Mice , Pinocytosis/drug effects , Renal Insufficiency, Chronic/microbiology , Triglycerides/metabolism
6.
Immunobiology ; 225(5): 152003, 2020 09.
Article in English | MEDLINE | ID: mdl-32962822

ABSTRACT

Complement (C) system is a double edge sword acting as the first line of defense on the one hand and causing aggravation of disease on the other. C activation when unregulated affects different organs including muscle regeneration. However, the effect of factor H (FH), a critical regulator of the alternative C pathway in muscle remains to be studied. FH deficiency results in excessive C activation and generates proinflammatory fragments C5a and C3a as byproducts. C3a and C5a signal through their respective receptors, C5aR and C3aR. In this study, we investigated the role of FH and downstream C5a/C5aR signaling in muscle architecture and function. Using the FH knockout (fh-/-) and fh-/-/C5aR-/double knockout mice we explored the role of C, specifically the alternative C pathway in muscle dysfunction. Substantial C3 and C9 deposits occur along the walls of the fh-/- muscle fibers indicative of unrestricted C activation. Physical performance assessments of the fh-/- mice show reduced grip endurance (76 %), grip strength (14 %) and rotarod balance (36 %) compared to controls. Histological analysis revealed a shift in muscle fiber populations indicated by an increase in glycolytic MHC IIB fibers and reduction in oxidative MHC IIA fibers. Consistent with this finding, mitochondrial DNA (mtDNA) and citrate synthase (CS) expression were both reduced indicating possible reduction in mitochondrial biomass. In addition, our results showed a significant increase in TGFß expression and altered TGFß localization in this setting. The architecture of cytoskeletal proteins actin and vimentin in the fh-/- muscle was changed that could lead to contractile weakness and loss of skeletal muscle elasticity. The muscle pathology in fh-/- mice was reduced in fh-/-/C5aR-/- double knockout (DKO) mice, highlighting partial C5aR dependence. Our results for the first time demonstrate an important role of FH in physical performance and skeletal muscle health.


Subject(s)
Complement C5a/metabolism , Complement Factor H/genetics , Muscle, Skeletal/metabolism , Physical Endurance/genetics , Receptor, Anaphylatoxin C5a/metabolism , Actins/metabolism , Animals , Complement C3/analysis , Complement C3/genetics , Complement C5a/analysis , Complement Factor H/metabolism , DNA, Mitochondrial , Gene Expression , Male , Mice, Inbred C57BL , Mice, Knockout , Muscle Fatigue/genetics , Muscle Strength/genetics , Receptor, Anaphylatoxin C5a/genetics , Rotarod Performance Test , Vimentin/metabolism
7.
Precis Clin Med ; 3(1): 34-43, 2020 Mar.
Article in English | MEDLINE | ID: mdl-32257532

ABSTRACT

Systemic lupus erythematosus (SLE) is an autoimmune disease that is a challenge to diagnose and treat. There is an urgent need for biomarkers to help define organ involvement, and more effective therapies. A unique population of T cells, the CD3+CD4-CD8- (DNeg) cells, is significantly increased in lupus patients. Twenty-seven cases (53%) of pediatric SLE patients had elevated DNeg cells in their peripheral blood, which correlated with kidney function (R2  = 0.54). Significant infiltration of DNeg cells was observed in both adult and pediatric lupus kidneys by immunofluorescence. For the first time, this study provides direct evidence that DNeg cells facilitate kidney injury in preclinical 8-week-old MRL/lpr lupus mice. In lupus mice, the increase in DNeg cells tracked with worsening disease and correlated with kidney function (R2  = 0.85). Our results show that DNeg cells per se can cause kidney dysfunction, increase in number with increase in disease pathology, and could serve as a potential biomarker.

8.
Clin Immunol ; 198: 102-105, 2019 01.
Article in English | MEDLINE | ID: mdl-30472267

ABSTRACT

Owing to an increasing shortage of donor organs, the majority of patients with end-stage kidney disease remains reliant on extracorporeal hemodialysis (HD) in order to counter the lifelong complications of a failing kidney. While HD remains a life-saving option for these patients, mounting evidence suggests that it also fuels a vicious cycle of thromboinflammation that can increase the risk of cardiovascular disease. During HD, blood-borne innate immune systems become inappropriately activated on the biomaterial surface, instigating proinflammatory reactions that can alter endothelial and vascular homeostasis. Complement activation, early during the HD process, has been shown to fuel a multitude of detrimental thromboinflammatory reactions that collectively contribute to patient morbidity. Here we discuss emerging aspects of complement's involvement in HD-induced inflammation and put forth the concept that targeted intervention at the level of C3 might constitute a promising therapeutic approach in HD patients.


Subject(s)
Complement C3/antagonists & inhibitors , Complement Inactivating Agents/therapeutic use , Inflammation/drug therapy , Renal Dialysis/adverse effects , Humans
9.
Immunobiology ; 223(12): 761-771, 2018 12.
Article in English | MEDLINE | ID: mdl-30107932

ABSTRACT

Complement system is an important arm of the immune system that promotes inflammation. Complement Factor H (FH) is a critical regulator of the alternative complement pathway. Its absence causes pathology in different organs resulting in diseases such as age related macular degeneration and dense deposit disease. Recent studies suggest that the complement system plays a role in bone development and homeostasis. To determine the role of FH in bone architecture, we studied the FH knockout (fh-/-) mice. 3D reconstructions of femur from 16 week old fh-/- mice reveal significant changes, such as decreased BV/TV (4.5%, p < 0.02), trabecular number (22%, p < 0.01), tissue mineral density (16%, p < 0.04), and increased marrow area (16% p < 0.01), compared to their wild type (WT) counterparts. Kidney function and histology remained normal indicating that bone changes occurred prior to kidney dysfunction. Next we examined cultured osteoblasts and osteoclasts isolated from bone marrow. FH is expressed ubiquitously in the osteoblasts and in the cytoplasm of osteoclasts. The changes caused by absence of FH include: increase in number of osteoblasts (362%) and osteoclasts (342%), increase in RNA (180%) and protein expression of cathepsin K and increased osteoclast function (pit formation, 233%). Actin rearrangement in both osteoblasts and osteoclasts was altered, with a loss of integrity of the F-actin ring at the periphery of the osteoclasts. For the first time our studies demonstrate a direct role of FH in the maintenance of bone structure and function and is highlighted as a promising therapeutic target in bone diseases.


Subject(s)
Bone and Bones/immunology , Bone and Bones/metabolism , Complement Factor H/immunology , Actins/metabolism , Animals , Biomarkers , Bone Resorption/genetics , Bone Resorption/metabolism , Bone Resorption/pathology , Bone and Bones/diagnostic imaging , Bone and Bones/pathology , Complement Factor H/genetics , Complement Factor H/metabolism , Fluorescent Antibody Technique , Immunohistochemistry , Mice , Mice, Knockout , Osteoblasts/immunology , Osteoblasts/metabolism , Osteoclasts/immunology , Osteoclasts/metabolism , Phenotype , X-Ray Microtomography
11.
Am J Physiol Renal Physiol ; 315(3): F487-F502, 2018 09 01.
Article in English | MEDLINE | ID: mdl-29693447

ABSTRACT

Several lines of evidence suggest that gut bacterial microbiota is altered in patients with chronic kidney disease (CKD), though the mechanism of which this dysbiosis takes place is not well understood. Recent studies delineated changes in gut microbiota in both CKD patients and experimental animal models using microarray chips. We present 16S ribosomal RNA gene sequencing of both stool pellets and small bowel contents of C57BL/6J mice that underwent a remnant kidney model and establish that changes in microbiota take place in the early gastrointestinal tract. Increased intestinal urea concentration has been hypothesized as a leading contributor to dysbiotic changes in CKD. We show that urea transporters (UT)-A and UT-B mRNA are both expressed throughout the whole gastrointestinal tract. The noted increase in intestinal urea concentration appears to be independent of UTs' expression. Urea supplementation in drinking water resulted in alteration in bacterial gut microbiota that is quite different than that seen in CKD. This indicates that increased intestinal urea concentration might not fully explain the CKD- associated dysbiosis.


Subject(s)
Bacteria/metabolism , Dysbiosis , Gastrointestinal Microbiome , Intestine, Small/microbiology , Renal Insufficiency, Chronic/microbiology , Urea/metabolism , Uremia/microbiology , Administration, Oral , Animals , Bacteria/classification , Bacteria/genetics , Disease Models, Animal , Feces/microbiology , Host-Pathogen Interactions , Hydrolysis , Intestine, Small/metabolism , Male , Membrane Transport Proteins/metabolism , Mice, Inbred C57BL , Renal Insufficiency, Chronic/metabolism , Ribotyping , Urea/administration & dosage , Urease/metabolism , Uremia/metabolism
12.
Immunobiology ; 223(6-7): 460-465, 2018.
Article in English | MEDLINE | ID: mdl-29478734

ABSTRACT

Microbiota consists of more than 1014 microorganisms that inhabit different areas of the body including the gastrointestinal tract, mainly the mouth and gut. It includes viruses, fungi, protozoa, archaea and bacteria. The microbiota interacts closely with host leading to a dynamic relationship that results in the biological effects observed. Its diverse genetic material (microbiome) interacts closely with the host immune system and cells, and therefore is closely associated with inflammation, immune tolerance, adaptive immunity and autoimmune diseases. Bacterial microbiota, which is the mostly studied lives in harmony with the host and maintains a symbiotic relationship. Therefore it plays an important role in immunological, metabolic, and neurological aspects and thereby the well-being of the host. Alteration of the homeostatic environment or the dynamic balance of microorganisms can result in dysbiosis or disease. However, does dysbiosis cause disease, aggravate disease or is the result of the disease remains to be defined, it could be a bit of all three factors. More recently, a number of studies demonstrate that these microorganisms could contribute to disease. Alteration of the tightly balanced composition of bacterial microbiota (dysbiosis) leads to exacerbation, rapid progression and worsening of disease states. It is important to identify the 'healthy' microbes that maintain a healthy environment, the 'sensitive' microbes that go awry with disease, the 'bad' microbes that cause disease and the 'therapeutic' microbes that can help rectify the changes. Increased relative abundance of certain bacterial species has been linked to triggering autoimmune diseases. Despite the burgeoning literature in the field, the molecular mechanisms by which the microbiota impacts the body in health and disease remain largely unknown. In this review, we will discuss recent advancements in our understanding of the gut bacterial microbiota associated with inflammatory and immunological processes and the role they play in the autoimmune disease, systemic lupus erythematosus.


Subject(s)
Bacteria , Dysbiosis/immunology , Intestinal Mucosa/microbiology , Lupus Erythematosus, Systemic/microbiology , Microbiota/immunology , Animals , Autoimmunity , Homeostasis , Humans , Immune Tolerance , Intestinal Mucosa/immunology , Lupus Erythematosus, Systemic/immunology
13.
Immunol Invest ; 46(8): 816-832, 2017 Nov.
Article in English | MEDLINE | ID: mdl-29058550

ABSTRACT

The complement system which is a critical mediator of innate immunity plays diverse roles in the neuropathogenesis of HIV-1 infection such as clearing HIV-1 and promoting productive HIV-1 replication. In the development of HIV-1 associated neurological disorders (HAND), there may be an imbalance between complement activation and regulation, which may contribute to the neuronal damage as a consequence of HIV-1 infection. It is well recognized that opiate abuse exacerbates HIV-1 neuropathology, however, little is known about the role of complement proteins in opiate induced neuromodulation, specifically in the presence of co-morbidity such as HIV-1 infection. Complement levels are significantly increased in the HIV-1-infected brain, thus HIV-induced complement synthesis may represent an important mechanism for the pathogenesis of AIDS in the brain, but remains underexplored. Anti-HIV-1 antibodies are able to initiate complement activation in HIV-1 infected CNS cells such as microglia and astrocytes during the course of disease progression; however, this complement activation fails to clear and eradicate HIV-1 from infected cells. In addition, the antiretroviral agents used for HIV therapy cause dysregulation of lipid metabolism, endothelial, and adipocyte cell function, and activation of pro-inflammatory cytokines. We speculate that both HIV-1 and opiates trigger a cytokine-mediated pro-inflammatory stimulus that modulates the complement cascade to exacerbate the virus-induced neurological damage. We examined the expression levels of C1q, SC5b-9, C5L2, C5aR, C3aR, and C9 key members of the complement cascade both in vivo in post mortem brain frontal cortex tissue from patients with HAND who used/did not use heroin, and in vitro using human microglial cultures treated with HIV tat and/or heroin. We observed significant expression of C1q and SC5b-9 by immunofluorescence staining in both the brain cortical and hippocampal region in HAND patients who abused heroin. Additionally, we observed increased gene expression of C5aR, C3aR, and C9 in the brain tissue of both HIV-1 infected patients with HAND who abused and did not abuse heroin, as compared to HIV negative controls. Our results show a significant increase in the expression of complement proteins C9, C5L2, C5aR, and C3aR in HIV transfected microglia and an additional increase in the levels of these complement proteins in heroin-treated HIV transfected microglia. This study highlights the a) potential roles of complement proteins in the pathogenesis of HIV-1-related neurodegenerative disorders; b) the combined effect of an opiate, like heroin, and HIV viral protein like HIV tat on complement proteins in normal human microglial cells and HIV transfected microglial cells. In the context of HAND, targeting selective steps in the complement cascade could help ameliorating the HIV burden in the CNS, thus investigations of complement-related therapeutic approaches for the treatment of HAND are warranted.


Subject(s)
AIDS-Associated Nephropathy/immunology , Complement System Proteins/metabolism , Frontal Lobe/metabolism , HIV Infections/immunology , HIV-1/physiology , Heroin Dependence/immunology , Inflammation Mediators/metabolism , Microglia/metabolism , AIDS-Associated Nephropathy/epidemiology , Cadaver , Cells, Cultured , Comorbidity , Complement Activation , Cytokines/metabolism , HIV Infections/epidemiology , Heroin Dependence/epidemiology , Humans , Immunomodulation , Microglia/pathology , Microglia/virology , Up-Regulation , tat Gene Products, Human Immunodeficiency Virus/metabolism
14.
PLoS One ; 12(9): e0184789, 2017.
Article in English | MEDLINE | ID: mdl-28931089

ABSTRACT

The modern Western diet is rich in advanced glycation end products (AGEs). We have previously shown an association between dietary AGEs and markers of inflammation and oxidative stress in a population of end stage renal disease (ESRD) patients undergoing peritoneal dialysis (PD). In the current pilot study we explored the effects of dietary AGEs on the gut bacterial microbiota composition in similar patients. AGEs play an important role in the development and progression of cardiovascular (CVD) disease. Plasma concentrations of different bacterial products have been shown to predict the risk of incident major adverse CVD events independently of traditional CVD risk factors, and experimental animal models indicates a possible role AGEs might have on the gut microbiota population. In this pilot randomized open label controlled trial, twenty PD patients habitually consuming a high AGE diet were recruited and randomized into either continuing the same diet (HAGE, n = 10) or a one-month dietary AGE restriction (LAGE, n = 10). Blood and stool samples were collected at baseline and after intervention. Variable regions V3-V4 of 16s rDNA were sequenced and taxa was identified on the phyla, genus, and species levels. Dietary AGE restriction resulted in a significant decrease in serum Nε-(carboxymethyl) lysine (CML) and methylglyoxal-derivatives (MG). At baseline, our total cohort exhibited a lower relative abundance of Bacteroides and Alistipes genus and a higher abundance of Prevotella genus when compared to the published data of healthy population. Dietary AGE restriction altered the bacterial gut microbiota with a significant reduction in Prevotella copri and Bifidobacterium animalis relative abundance and increased Alistipes indistinctus, Clostridium citroniae, Clostridium hathewayi, and Ruminococcus gauvreauii relative abundance. We show in this pilot study significant microbiota differences in peritoneal dialysis patients' population, as well as the effects of dietary AGEs on gut microbiota, which might play a role in the increased cardiovascular events in this population and warrants further studies.


Subject(s)
Cardiovascular Diseases/etiology , Diet/adverse effects , Gastrointestinal Microbiome/drug effects , Glycation End Products, Advanced/adverse effects , Kidney Failure, Chronic/complications , Peritoneal Dialysis/adverse effects , Feces/microbiology , Female , Glycation End Products, Advanced/analysis , Humans , Male , Middle Aged , Pilot Projects
15.
Immunology ; 148(4): 407-19, 2016 08.
Article in English | MEDLINE | ID: mdl-27213693

ABSTRACT

Blood-brain barrier (BBB) dysfunction complicates central nervous system lupus, an important aspect of systemic lupus erythematosus. To gain insight into the underlying mechanism, vascular corrosion casts of brain were generated from the lupus mouse model, MRL/lpr mice and the MRL/MpJ congenic controls. Scanning electron microscopy of the casts showed loss of vascular endothelial cells in lupus mice compared with controls. Immunostaining revealed a significant increase in caspase 3 expression in the brain vascular endothelial cells, which suggests that apoptosis could be an important mechanism causing cell loss, and thereby loss of BBB integrity. Complement activation occurs in lupus resulting in increased generation of circulating C5a, which caused the endothelial layer to become 'leaky'. In this study, we show that C5a and lupus serum induced apoptosis in cultured human brain microvascular endothelial cells (HBMVECs), whereas selective C5a receptor 1 (C5aR1) antagonist reduced apoptosis in these cells, demonstrating C5a/C5aR1-dependence. Gene expression of initiator caspases, caspase 1 and caspase 8, and pro-apoptotic proteins death-associated protein kinase 1, Fas-associated protein (FADD), cell death-inducing DNA fragmentation factor 45 000 MW subunit A-like effector B (CIDEB) and BCL2-associated X protein were increased in HBMVECs treated with lupus serum or C5a, indicating that both the intrinsic and extrinsic apoptotic pathways could be critical mediators of brain endothelial cell apoptosis in this setting. Overall, our findings suggest that C5a/C5aR1 signalling induces apoptosis through activation of FADD, caspase 8/3 and CIDEB in brain endothelial cells in lupus. Further elucidation of the underlying apoptotic mechanisms mediating the reduced endothelial cell number is important in establishing the potential therapeutic effectiveness of C5aR1 inhibition that could prevent and/or reduce BBB alterations and preserve the physiological function of BBB in central nervous system lupus.


Subject(s)
Apoptosis Regulatory Proteins/metabolism , Apoptosis , Brain/pathology , Complement C5a/metabolism , Endothelium, Vascular/immunology , Fas-Associated Death Domain Protein/metabolism , Lupus Vasculitis, Central Nervous System/immunology , Animals , Apoptosis/drug effects , Apoptosis/immunology , Brain/metabolism , Caspase 3/metabolism , Cells, Cultured , Disease Models, Animal , Endothelium, Vascular/drug effects , Endothelium, Vascular/pathology , Humans , Mice , Mice, Inbred MRL lpr , Peptides, Cyclic/pharmacology , Receptor, Anaphylatoxin C5a/antagonists & inhibitors , Signal Transduction/drug effects
16.
Am J Physiol Renal Physiol ; 310(9): F895-908, 2016 05 01.
Article in English | MEDLINE | ID: mdl-26887830

ABSTRACT

Thrombotic microangiopathy (TMA) is a disorder characterized by microvascular occlusion that can lead to thrombocytopenia, hemolytic anemia, and glomerular damage. Complement activation is the central event in most cases of TMA. Primary forms of TMA are caused by mutations in genes encoding components of the complement or regulators of the complement cascade. Recently, we and others have described a genetic form of TMA caused by mutations in the gene diacylglycerol kinase-ε (DGKE) that encodes the lipid kinase DGKε (Lemaire M, Fremeaux-Bacchi V, Schaefer F, Choi MR, Tang WH, Le Quintrec M, Fakhouri F, Taque S, Nobili F, Martinez F, Ji WZ, Overton JD, Mane SM, Nurnberg G, Altmuller J, Thiele H, Morin D, Deschenes G, Baudouin V, Llanas B, Collard L, Majid MA, Simkova E, Nurnberg P, Rioux-Leclerc N, Moeckel GW, Gubler MC, Hwa J, Loirat C, Lifton RP. Nat Genet 45: 531-536, 2013; Ozaltin F, Li BH, Rauhauser A, An SW, Soylemezoglu O, Gonul II, Taskiran EZ, Ibsirlioglu T, Korkmaz E, Bilginer Y, Duzova A, Ozen S, Topaloglu R, Besbas N, Ashraf S, Du Y, Liang CY, Chen P, Lu DM, Vadnagara K, Arbuckle S, Lewis D, Wakeland B, Quigg RJ, Ransom RF, Wakeland EK, Topham MK, Bazan NG, Mohan C, Hildebrandt F, Bakkaloglu A, Huang CL, Attanasio M. J Am Soc Nephrol 24: 377-384, 2013). DGKε is unrelated to the complement pathway, which suggests that unidentified pathogenic mechanisms independent of complement dysregulation may result in TMA. Studying Dgke knockout mice may help to understand the pathogenesis of this disease, but no glomerular phenotype has been described in these animals so far. Here we report that Dgke null mice present subclinical microscopic anomalies of the glomerular endothelium and basal membrane that worsen with age and develop glomerular capillary occlusion when exposed to nephrotoxic serum. We found that induction of cyclooxygenase-2 and of the proangiogenic prostaglandin E2 are impaired in Dgke null kidneys and are associated with reduced expression of the antithrombotic cell adhesion molecule platelet endothelial cell adhesion molecule-1/CD31 in the glomerular endothelium. Notably, prostaglandin E2 supplementation was able to rescue motility defects of Dgke knockdown cells in vitro and to restore angiogenesis in a test in vivo. Our results unveil an unexpected role of Dgke in the induction of cyclooxygenase-2 and in the regulation of glomerular prostanoids synthesis under stress.


Subject(s)
Cyclooxygenase 2/biosynthesis , Diacylglycerol Kinase/genetics , Dinoprostone/biosynthesis , Endothelium/pathology , Glomerulonephritis/pathology , Kidney Glomerulus/metabolism , Kidney Glomerulus/pathology , Aging/pathology , Animals , Cell Movement , Glomerulonephritis/enzymology , Glomerulonephritis/metabolism , Kidney Function Tests , Kidney Glomerulus/enzymology , Macrophages/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Neovascularization, Physiologic , Wound Healing
17.
Cell Mol Immunol ; 13(5): 651-7, 2016 09.
Article in English | MEDLINE | ID: mdl-26166765

ABSTRACT

Activation of complement generates C5a which leads to signaling through C5aR1. This is tightly controlled, including by the plasma proteins factor H (FH) and carboxypeptidase N. Here we studied a chronic serum sickness (CSS) model of glomerulonephritis (GN) in which there is an active humoral immune response, formation of glomerular immune complexes (ICs), and resulting glomerular inflammation. The antibody response, glomerular IC deposition, the degree of GN, and consequent renal functional insufficiency in CSS were all worse in FH-/- mice compared to wild-type FH+/+ animals. This was ameliorated in the former by giving a C5aR1 antagonist for the final 3 weeks of the 5-week protocol. In contrast, blocking CP-mediated inactivation of C5a increased these disease measures. Thus, complement regulation by both plasma FH and CP to limit the quantity of active C5a is important in conditions where the humoral immune response is directed to a continuously present foreign antigen. Signaling through C5aR1 enhances the humoral immune response as well as the inflammatory response to ICs that have formed in glomeruli. Both effects are relevant even after disease has begun. Thus, pharmacological targeting of C5a in IC-mediated GN has potential clinical relevance.


Subject(s)
Antigen-Antibody Complex/metabolism , Carboxypeptidases/metabolism , Glomerulonephritis/immunology , Receptor, Anaphylatoxin C5a/antagonists & inhibitors , Animals , Apoferritins/metabolism , Complement C5a/metabolism , Complement Factor H/deficiency , Complement Factor H/metabolism , Glomerulonephritis/pathology , Immunity, Humoral , Immunoglobulin G/metabolism , Kidney/pathology , Kidney/physiopathology , Kidney Function Tests , Male , Mice, Inbred C57BL , Receptor, Anaphylatoxin C5a/metabolism
18.
Adv Chronic Kidney Dis ; 22(5): 343-51, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26311595

ABSTRACT

Infections with hepatitis B virus (HBV) and hepatitis C virus (HCV) are prevalent worldwide. In this review, we discuss the epidemiology, pathogenesis, clinical manifestations, and treatment of HBV- and HCV-related glomerulonephritis (GN). The most common histopathologic presentation of HBV-GN is HBV-associated membranous nephropathy, which usually manifests clinically with varying grades of proteinuria and microscopic hematuria. The pathogenesis is likely to be immune complex mediated; however, other host and viral factors have been implicated. The treatment of HBV-GN revolves around antiviral therapy. Various histologic types of glomerular diseases are reported in association with HCV infection, the most frequent being Type 1 membranoproliferative glomerulonephritis, usually in the context of Type 2 mixed cryoglobulinemia. The pathogenesis of HCV-GN can be attributed to glomerular deposition of cryoglobulins or noncryoglobulin-immune complexes. Cryoglobulins typically comprised immunoglobulin Mκ with rheumatoid factor activity. Clinically, patients may present with proteinuria, microscopic hematuria, hypertension, and acute nephritic and/or nephrotic syndrome. The treatment of HCV-GN, especially cryoglobulinemic membranoproliferative glomerulonephritis, encompasses various options including contemporary antiviral therapy with or without conventional and novel immunomodulatory agents.


Subject(s)
Glomerulonephritis/epidemiology , Glomerulonephritis/virology , Hepatitis B/epidemiology , Hepatitis C/epidemiology , Antiviral Agents/therapeutic use , Comorbidity , Disease Progression , Female , Glomerular Filtration Rate , Glomerulonephritis/drug therapy , Glomerulonephritis/pathology , Hepatitis B/drug therapy , Hepatitis B/physiopathology , Hepatitis C/drug therapy , Hepatitis C/physiopathology , Humans , Kidney Function Tests , Male , Prognosis , Risk Assessment , Severity of Illness Index , Treatment Outcome
19.
Immunology ; 146(1): 130-43, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26059553

ABSTRACT

The blood-brain barrier (BBB) plays a crucial role in brain homeostasis, thereby maintaining the brain environment precise for optimal neuronal function. Its dysfunction is an intriguing complication of systemic lupus erythematosus (SLE). SLE is a systemic autoimmune disorder where neurological complications occur in 5-50% of cases and is associated with impaired BBB integrity. Complement activation occurs in SLE and is an important part of the clinical profile. Our earlier studies demonstrated that C5a generated by complement activation caused the loss of brain endothelial layer integrity in rodents. The goal of the current study was to determine the translational potential of these studies to a human system. To assess this, we used a two dimensional in vitro BBB model constructed using primary human brain microvascular endothelial cells and astroglial cells, which closely emulates the in vivo BBB allowing the assessment of BBB integrity. Increased permeability monitored by changes in transendothelial electrical resistance and cytoskeletal remodelling caused by actin fiber rearrangement were observed when the cells were exposed to lupus serum and C5a, similar to the observations in mice. In addition, our data show that C5a/C5aR1 signalling alters nuclear factor-κB translocation into nucleus and regulates the expression of the tight junction proteins, claudin-5 and zonula occludens 1 in this setting. Our results demonstrate for the first time that C5a regulates BBB integrity in a neuroinflammatory setting where it affects both endothelial and astroglial cells. In addition, we also demonstrate that our previous findings in a mouse model, were emulated in human cells in vitro, bringing the studies one step closer to understanding the translational potential of C5a/C5aR1 blockade as a promising therapeutic strategy in SLE and other neurodegenerative diseases.


Subject(s)
Blood-Brain Barrier/metabolism , Complement C5a/metabolism , Lupus Erythematosus, Systemic/pathology , Receptor, Anaphylatoxin C5a/metabolism , Actin Cytoskeleton/metabolism , Active Transport, Cell Nucleus , Adolescent , Astrocytes/immunology , Brain/blood supply , Cells, Cultured , Child , Claudin-5/biosynthesis , Complement Activation/immunology , Complement C5a/immunology , Cyclic AMP Response Element-Binding Protein/metabolism , Electric Impedance , Endothelial Cells/immunology , Extracellular Signal-Regulated MAP Kinases/metabolism , Female , Humans , Junctional Adhesion Molecules/biosynthesis , Lupus Erythematosus, Systemic/immunology , Male , NF-kappa B/metabolism , Protein Transport , Receptor, Anaphylatoxin C5a/antagonists & inhibitors , Receptor, Anaphylatoxin C5a/immunology , Tight Junctions/metabolism , Zonula Occludens-1 Protein/biosynthesis
20.
Kidney Int ; 87(5): 930-9, 2015 May.
Article in English | MEDLINE | ID: mdl-25565310

ABSTRACT

In chronic serum sickness, glomerular immune complexes form, yet C57BL/6 mice do not develop glomerulonephritis unless complement factor H (CfH) is absent, indicating the relevance of complement regulation. Complement receptor 3 (CD11b) and Fcγ receptors on leukocytes, and CfH on platelets, can bind immune complexes. Here we induced immune complex-mediated glomerulonephritis in CfH(-/-) mice chimeric for wild-type, CfH(-/-), CD11b(-/-), or FcRγ(-/-) bone marrow stem cells. Glomerulonephritis was worse in CD11b(-/-) chimeras compared with all others, whereas disease in FcRγ(-/-) and wild-type chimeras was comparable. Disease tracked strongly with humoral immune responses, but not glomerular immune complex deposits. Interstitial inflammation with M1 macrophages strongly correlated with glomerulonephritis scores. CD11b(-/-) chimeras had significantly more M1 macrophages and CD4(+) T cells. The renal dendritic cell populations originating from bone marrow-derived CD11c(+) cells were similar in all experimental groups. CD11b(+) cells bearing colony-stimulating factor 1 receptor were present in kidneys, including CD11b(-/-) chimeras; these cells correlated negatively with glomerulonephritis scores. Thus, experimental immune complex-mediated glomerulonephritis is associated with accumulation of M1 macrophages and CD4(+) T cells in kidneys and functional renal insufficiency. Hence, CD11b on mononuclear cells is instrumental in generating an anti-inflammatory response in the inflamed kidney.


Subject(s)
CD11b Antigen/metabolism , Complement Factor H/metabolism , Glomerulonephritis/immunology , Leukocytes/metabolism , Receptors, IgG/metabolism , Animals , Antigen-Antibody Complex/metabolism , Apoferritins/immunology , Bone Marrow/metabolism , CD11b Antigen/genetics , CD18 Antigens/metabolism , Complement Factor H/genetics , Glomerulonephritis/metabolism , Immunoglobulin G/metabolism , Mice, Inbred C57BL , Receptor, Anaphylatoxin C5a/metabolism , Receptors, IgG/genetics , Serum Sickness/complications
SELECTION OF CITATIONS
SEARCH DETAIL
...