ABSTRACT
The aetiology and progression of systemic lupus erythematosus (SLE) resulted from a complex sequence of events generated both from genetic and epigenetic processes. In the current research, the effect of methyl-supplemented nutrition on the development of SLE was studied in the pristane-induced mouse model of the disease. The results clearly demonstrated decreased anti-dsDNA antibody and proteinuria levels, modulation of cytokines and protected renal structures in the group of treated mice. An additional increase in the DNA methylation of mouse B lymphocytes was also observed. The beneficial effect of the diet is due to the methyl-containing micronutrients with possible anti-inflammatory and immunomodulating effects on cell proliferation and gene expression. Since these components are responsible for maintaining the physiological methylation level of DNA, the results point to the central role of methylation processes in environmentally triggered lupus. As nutrition represents one of the major epigenetic factors, these micronutrients may be considered novel agents with significant therapeutic outcomes.
Subject(s)
Antibodies, Antinuclear , B-Lymphocytes , DNA Methylation , Dietary Supplements , Disease Models, Animal , Lupus Erythematosus, Systemic , Terpenes , Animals , Lupus Erythematosus, Systemic/immunology , Lupus Erythematosus, Systemic/chemically induced , Mice , Antibodies, Antinuclear/immunology , Antibodies, Antinuclear/blood , Female , B-Lymphocytes/immunology , B-Lymphocytes/metabolism , Cytokines/metabolism , Epigenesis, Genetic , Micronutrients/administration & dosage , Proteinuria/immunology , Kidney/immunology , Kidney/metabolism , Kidney/pathology , Kidney/drug effectsABSTRACT
BACKGROUND AND PURPOSE: Reports of patients who have autoimmune nodopathies concurrent with nephrotic syndrome are increasing. We investigated whether proteinuria could be a biomarker of autoimmune nodopathies. METHODS: Qualitative urinalysis results were retrospectively obtained from 69 patients who were diagnosed with chronic inflammatory demyelinating polyneuropathy (CIDP) at a hospital in Japan. Proteinuria was graded as mild to severe (i.e., mild, 30-99; moderate, 100-299; severe, 300 mg/dL or more) according to the results of the urine dipstick test. Autoantibodies against the paranodal proteins contactin 1 (CNTN1), neurofascin 155 (NF155), and contactin-associated protein 1 (Caspr1) and the nodal protein neurofascin 186 (NF186) were measured, and the predominant IgG subclass was determined by enzyme-linked immunosorbent assay in sera from the 69 patients. RESULTS: Four patients (6%), five patients (7%), and one (1%) patient were positive for anti-CNTN1, anti-NF155, and anti-Caspr1 IgG4 antibodies, respectively. No patients had IgG4 antibodies against NF186. Proteinuria of mild or greater levels was found in three patients with anti-CNTN1 IgG4 and two patients with anti-NF155 IgG4 antibodies. The autoantibody-positive patients more frequently had proteinuria of mild or greater levels than the seronegative patients (p = 0.01). CONCLUSIONS: Proteinuria is a possible biomarker of autoimmune nodopathies associated with autoantibodies targeting CNTN1 or NF155. Urinalysis results should be carefully checked for quick differentiation of autoimmune nodopathies from CIDP. Patients who present with nephrotic syndrome should be tested for anti-CNTN1 IgG4 antibodies, and patients who exhibit mild proteinuria should be tested for anti-NF155 IgG4 antibodies.
Subject(s)
Autoantibodies , Proteinuria , Humans , Female , Male , Middle Aged , Adult , Proteinuria/immunology , Aged , Autoantibodies/blood , Retrospective Studies , Polyradiculoneuropathy, Chronic Inflammatory Demyelinating/immunology , Polyradiculoneuropathy, Chronic Inflammatory Demyelinating/diagnosis , Polyradiculoneuropathy, Chronic Inflammatory Demyelinating/blood , Biomarkers/urine , Biomarkers/blood , Young Adult , Adolescent , Immunoglobulin G/blood , Immunoglobulin G/urine , Contactin 1/immunology , Nerve Growth Factors/immunology , Aged, 80 and overABSTRACT
Anti-phospholipase A2 receptor autoantibody (PLA2R-Ab) plays a critical role in the pathogenesis of primary membranous nephropathy (PMN), an autoimmune kidney disease characterized by immune deposits in the glomerular subepithelial spaces and proteinuria. However, the mechanism of how PLA2R-Abs interact with the conformational epitope(s) of PLA2R has remained elusive. PLA2R is a single transmembrane helix receptor containing ten extracellular domains that begin with a CysR domain followed by a FnII and eight CTLD domains. Here, we examined the interactions of PLA2R-Ab with the full PLA2R protein, N-terminal domain truncations, and C-terminal domain deletions under either denaturing or physiological conditions. Our data demonstrate that the PLA2R-Abs against the dominant epitope (the N-terminal CysR-CTLD1 triple domain) possess weak cross-reactivities to the C-terminal domains beyond CTLD1. Moreover, both the CysR and CTLD1 domains are required to form a conformational epitope for PLA2R-Ab interaction, with FnII serving as a linker domain. Upon close examination, we also observed that patients with newly diagnosed PMN carry two populations of PLA2R-Abs in sera that react to the denatured CysR-CTLD3 (the PLA2R-Ab1) and denatured CysR-CTLD1 (the PLA2R-Ab2) domain complexes on Western blots, respectively. Furthermore, the PLA2R-Ab1 appeared at an earlier time point than PLA2R-Ab2 in patients, whereas the increased levels of PLA2R-Ab2 coincided with the worsening of proteinuria. In summary, our data support that an integrated folding of the three PLA2R N-terminal domains, CysR, FnII, and CTLD1, is a prerequisite to forming the PLA2R conformational epitope and that the dominant epitope-reactive PLA2R-Ab2 plays a critical role in PMN clinical progression.
Subject(s)
Glomerulonephritis, Membranous , Receptors, Phospholipase A2 , Autoantibodies/immunology , Autoantibodies/metabolism , Epitopes , Female , Glomerulonephritis, Membranous/genetics , Glomerulonephritis, Membranous/immunology , Glomerulonephritis, Membranous/urine , Humans , Male , Proteinuria/genetics , Proteinuria/immunology , Receptors, Phospholipase A2/chemistry , Receptors, Phospholipase A2/immunologyABSTRACT
The novel coronavirus (SARS-CoV-2) has turned into a life-threatening pandemic disease (Covid-19). About 5% of patients with Covid-19 have severe symptoms including septic shock, acute respiratory distress syndrome, and the failure of several organs, while most of them have mild symptoms. Frequently, the kidneys are involved through direct or indirect mechanisms. Kidney involvement mainly manifests itself as proteinuria and acute kidney injury (AKI). The SARS-CoV-2-induced kidney damage is expected to be multifactorial; directly it can infect the kidney podocytes and proximal tubular cells and based on an angiotensin-converting enzyme 2 (ACE2) pathway it can lead to acute tubular necrosis, protein leakage in Bowman's capsule, collapsing glomerulopathy and mitochondrial impairment. The SARS-CoV-2-driven dysregulation of the immune responses including cytokine storm, macrophage activation syndrome, and lymphopenia can be other causes of the AKI. Organ interactions, endothelial dysfunction, hypercoagulability, rhabdomyolysis, and sepsis are other potential mechanisms of AKI. Moreover, lower oxygen delivery to kidney may cause an ischaemic injury. Understanding the fundamental molecular pathways and pathophysiology of kidney injury and AKI in Covid-19 is necessary to develop management strategies and design effective therapies.
Subject(s)
Acute Kidney Injury/pathology , COVID-19/physiopathology , Cytokine Release Syndrome/pathology , Disseminated Intravascular Coagulation/pathology , Lymphopenia/pathology , Necrosis/pathology , Proteinuria/pathology , Sepsis/pathology , Acute Kidney Injury/immunology , Acute Kidney Injury/virology , Angiotensin-Converting Enzyme 2/genetics , Angiotensin-Converting Enzyme 2/immunology , COVID-19/immunology , COVID-19/virology , Cytokine Release Syndrome/immunology , Cytokine Release Syndrome/virology , Cytokines/genetics , Cytokines/immunology , Disseminated Intravascular Coagulation/immunology , Disseminated Intravascular Coagulation/virology , Host-Pathogen Interactions/genetics , Host-Pathogen Interactions/immunology , Humans , Kidney Tubules, Proximal/immunology , Kidney Tubules, Proximal/physiopathology , Lymphopenia/immunology , Lymphopenia/virology , Necrosis/immunology , Necrosis/virology , Podocytes/immunology , Podocytes/pathology , Proteinuria/immunology , Proteinuria/virology , SARS-CoV-2/immunology , SARS-CoV-2/pathogenicity , Sepsis/immunology , Sepsis/virology , Serine Endopeptidases/genetics , Serine Endopeptidases/immunology , Spike Glycoprotein, Coronavirus/genetics , Spike Glycoprotein, Coronavirus/immunologyABSTRACT
Children with idiopathic nephrotic syndrome (INS) usually have podocyte injury, and recent studies suggest a B cell dysfunction in INS. Therefore, this study attempts to screen and identify the podocyte autoantibodies in patients. Two-dimensional electrophoresis and mass spectrometry were used to screen and identify the pathogenic podocyte autoantibodies in children with INS. The positive rate, expression pattern, and clinical correlation of these podocyte autoantibodies in children with INS were determined by clinical study. At least 66% of INS children have podocyte autoantibodies. Seven podocyte autoantibodies closely related to INS were screened and identified for the first time in this study. These podocyte autoantibodies are positively correlated with proteinuria, and its titer will decrease rapidly after effective treatment. In this study, a group of new disease subgroup-"autoimmune podocytes" were identified by podocyte autoantibodies.
Subject(s)
Autoantibodies/immunology , Autoantigens/immunology , Autoimmune Diseases/immunology , Nephrotic Syndrome/immunology , Podocytes/immunology , Adolescent , Child , Child, Preschool , Female , Humans , Male , Proteinuria/immunologyABSTRACT
BACKGROUND: Systemic-lupus-nephritis is a chronic autoimmune disease characterized by immune complex deposition and a flare of autoantibodies and leading to renal injury. OBJECTIVES: To expose anti-Dense-Fine-Speckled-70 (DFS70)-antibodies to genetically-prone-lupus-mice. METHODS: NZBXW/F1 female mice were monitored for the onset of glomerulonephritis by proteinuria upon infusion of anti-DFS70 (40 µg/mouse), commercial-human-IgG (cIgG) or phosphate-buffered-saline (PBS) as controls. The survival time was detected by mice death. Circulating anti-dsDNA were tested by ELISA. Proteinuria, was defined by a standard semi-quantitative-Bayer-Multistix-dipstick. Kidney histology was analyzed by periodic-acid-Schiff-PAS staining. RESULTS: A significantly higher percentage of anti-DFS70-infused mice exhibited prolonged survival time as compared with cIgG and PBS-subjected mice (p < 0.022). One mouse out of 10 mice injected with anti-DFS70-antibodies died at week 36, whereas, 6 out of 10 mice subjected with PBS found dead at this time. Eighty percent of anti-DFS70 injected mice did not show severe glomerulonephritis by histology. CONCLUSIONS: anti-DFS70 attenuated the progression of glomerulonephritis and prolonged the survival time. Circulating anti-DFS70-autoantibodies may confer a protective role against renal injury in murine-lupus-nephritis. Our data may propose a novel therapy approach for lupus patients.
Subject(s)
Antibodies, Antinuclear/immunology , Glomerulonephritis/immunology , Glomerulonephritis/pathology , Lupus Nephritis/immunology , Lupus Nephritis/pathology , Adaptor Proteins, Signal Transducing/immunology , Animals , Antibodies, Antinuclear/pharmacology , Disease Models, Animal , Female , Kidney/pathology , Mice , Mice, Inbred NZB , Proteinuria/immunology , Proteinuria/pathology , Survival Rate , Transcription Factors/immunologyABSTRACT
Immune checkpoint inhibitors have dramatically improved cancer therapy for many patients. These humanized monoclonal antibodies against various immune checkpoints (receptors and ligands) effectively treat a number of malignancies by unleashing the immune system to destroy cancer cells. These drugs are not excreted by the kidneys or liver, have a long half-life, and undergo receptor-mediated clearance. Although these agents have greatly improved the prognosis of many cancers, immune-related end organ injury is a complication that has come to light in clinical practice. Although less common than other organ involvement, kidney lesions resulting in acute kidney injury and/or proteinuria are being described. Acute tubulointerstitial nephritis is the most common lesion seen on kidney biopsy, while acute tubular injury and glomerular lesions occur less commonly. Clinical findings and laboratory tests are suboptimal in predicting the underlying renal lesion, making kidney biopsy necessary in the majority of cases to definitely diagnose the lesion and potentially guide therapy. Immune checkpoint inhibitor discontinuation and corticosteroid therapy are recommended for acute tubulointerstitial nephritis. Based on a handful of cases, re-exposure to these drugs in patients who previously developed acute tubulointerstitial nephritis has been mixed. Although it is unclear whether re-exposure is appropriate, it should perhaps be considered in patients with limited options. When this approach is taken, patients should be closely monitored for recurrence of acute kidney injury. Treatment of cancer in patients with a kidney transplant with immune checkpoint inhibitors risks the development of acute rejection in some patients and requires close surveillance.
Subject(s)
Acute Kidney Injury/prevention & control , Antineoplastic Agents, Immunological/adverse effects , Graft Rejection/prevention & control , Immune Checkpoint Inhibitors/adverse effects , Proteinuria/prevention & control , Acute Kidney Injury/chemically induced , Acute Kidney Injury/immunology , Acute Kidney Injury/physiopathology , Graft Rejection/chemically induced , Graft Rejection/immunology , Humans , Kidney Glomerulus/drug effects , Kidney Glomerulus/immunology , Kidney Glomerulus/physiopathology , Kidney Transplantation/adverse effects , Neoplasms/drug therapy , Neoplasms/immunology , Proteinuria/chemically induced , Proteinuria/immunology , Proteinuria/physiopathologyABSTRACT
PURPOSE OF REVIEW: Immune dysfunction, including severe combined immunodeficiency, has been described in genetic disorders affecting the metabolism of the vitamins cobalamin (vitamin B12) and folate. We have reviewed reports of clinical findings in patients with a number of inborn errors of cobalamin or folate metabolism, specifically looking for immune problems. RECENT FINDINGS: There is little evidence that immune function is affected in most of the disorders. Exceptions are Imerslund-Gräsbeck syndrome and hereditary folate malabsorption (affecting intestinal absorption of cobalamin and folate, respectively), transcobalamin deficiency (affecting transport of cobalamin in blood and cellular cobalamin uptake), and methylenetetrahydrofolate dehydrogenase 1 deficiency (catalyzing cytoplasmic interconversion of reduced folate coenzyme derivatives). SUMMARY: Although some inborn errors of cobalamin or folate can be associated with immune dysfunction, the degree and type of immune dysfunction vary with no obvious pattern.
Subject(s)
Folic Acid Deficiency/immunology , Malabsorption Syndromes/immunology , Nutritional Physiological Phenomena/immunology , Primary Immunodeficiency Diseases/immunology , Vitamin B 12 Deficiency/immunology , Anemia, Megaloblastic/congenital , Anemia, Megaloblastic/immunology , Folic Acid/genetics , Folic Acid/immunology , Folic Acid Deficiency/congenital , Humans , Malabsorption Syndromes/congenital , Methylenetetrahydrofolate Dehydrogenase (NADP)/deficiency , Methylenetetrahydrofolate Dehydrogenase (NADP)/immunology , Minor Histocompatibility Antigens/immunology , Proteinuria/congenital , Proteinuria/immunology , Transcobalamins/deficiency , Transcobalamins/immunology , Vitamin B 12/genetics , Vitamin B 12/immunology , Vitamin B 12 Deficiency/congenitalABSTRACT
BACKGROUND: Previous reports suggest initial presentation of IgA nephropathy (IgAN) in children is different from adults. No systematic comparison of clinical, biological, and histological childhood- and adult-onset IgAN is currently available. METHODS: We compared pediatric and adult clinical and histological characteristics at IgAN diagnosis. Data on 211 consecutive patients from two different centers in Paris (82 children, 129 adults) were reviewed. Kidney biopsies were scored for Oxford classification and podocytopathic (P1) features. RESULTS: We report higher eGFR at diagnosis in children compared to adults (89.5 vs. 64 ml/min/1.73 m2; p = 0.0001) but no difference in proteinuria. Histological analysis of kidney biopsy found higher proportions of mesangial (M1) and endocapillary (E1) hypercellularity in children compared with adults (M1 [80.7% vs. 27.9%, p = 0.0001]; E1 [71.3% vs. 30%, p = 0.0001]). Focal glomerulosclerosis (S1), tubular atrophy/interstitial fibrosis ≥ 25% (T1), and P1 were more frequent in adults (S1 [81.5% vs. 61.3%, p = 0.0012], T1 [49.5% vs. 1.35%, p = 0.0001], P1 [33.8% vs. 16.4%, p = 0.008). Proteinuria associated with M1, E1, and C1 in children (M1, p = 0.0001; E1, p = 0.0005; C1, p = 0.0014) but S1, P1, and T1 in adults (S1, p = 0.0001; P1, p = 0.0001; T1, p = 0.001). After steroid treatment (41 children and 28 adults), proteinuria decreased in children (p < 0.001, follow-up 38 months) and adults (p < 0.001, follow-up 76.9 months), whereas eGFR remained stable in adults but increased significantly in children (90.6 to 110 ml/min/1.73m2). CONCLUSION: Proteinuria in children with IgAN is a marker of glomerular proliferative lesions whereas its presence in adults often reflects the presence of chronic lesions. This suggests the need for histological assessment.
Subject(s)
Glomerulonephritis, IGA/diagnosis , Glomerulosclerosis, Focal Segmental/epidemiology , Glucocorticoids/administration & dosage , Kidney Glomerulus/pathology , Proteinuria/diagnosis , Adult , Age Factors , Biopsy , Child , Female , Follow-Up Studies , Glomerular Filtration Rate/drug effects , Glomerular Filtration Rate/physiology , Glomerulonephritis, IGA/complications , Glomerulonephritis, IGA/drug therapy , Glomerulonephritis, IGA/immunology , Glomerulosclerosis, Focal Segmental/diagnosis , Glomerulosclerosis, Focal Segmental/drug therapy , Glomerulosclerosis, Focal Segmental/immunology , Humans , Kidney Glomerulus/drug effects , Kidney Glomerulus/immunology , Male , Proteinuria/drug therapy , Proteinuria/immunology , Proteinuria/urine , Retrospective Studies , Severity of Illness Index , Treatment OutcomeABSTRACT
BACKGROUND: Crescent formation in immunoglobulin A nephropathy (IgAN) has been demonstrated to be a risk factor for worse outcomes. For IgAN patients with 0-25% crescentic glomeruli (C1), whether corticosteroids (CS) can improve the prognosis remains unclear. We tried to investigate the need for using CS in IgAN patients with C1 in different proteinuria levels. METHODS: A total of 120 eligible IgAN patients with C1 from two academic medical centers were retrospectively studied, and 57 (47.5%) received CS. Patients were grouped according to with or without CS. The outcomes were the rate of estimated glomerular filtration rate (eGFR) decline (ml/min per 1.73 m2/year) and a composite outcome (50% decrease in eGFR, end stage renal disease (ESRD) or death due to kidney disease). The progression of adverse outcome among them were analyzed in Kaplan-Meier curve. The independent significance of CS on renal outcome or eGFR decline rate were analyzed by multivariable Cox regression or linear regression. RESULTS: Unadjusted Kaplan-Meier showed that the outcome of treated patients was better than that of the untreated patients. Multiple Cox regression and linear regression analysis found that CS independently protected the renal outcome and decreased the eGFR decline rate. In the subgroup analysis, multivariate linear regression showed that CS decreased the eGFR decline rate both in proteinuria ≥ 1 g/day and < 1 g/day. CONCLUSIONS: CS protected the renal outcome and slowed the eGFR decline rate of IgAN patients with C1, it also decreased the eGFR decline rate even in those with initial proteinuria < 1 g/day.
Subject(s)
Adrenal Cortex Hormones/administration & dosage , Glomerular Filtration Rate/drug effects , Glomerulonephritis, IGA/drug therapy , Kidney Glomerulus/drug effects , Prednisolone/administration & dosage , Prednisone/administration & dosage , Proteinuria/drug therapy , Adrenal Cortex Hormones/adverse effects , Adult , China , Disease Progression , Female , Glomerulonephritis, IGA/immunology , Glomerulonephritis, IGA/mortality , Glomerulonephritis, IGA/physiopathology , Humans , Kidney Failure, Chronic/immunology , Kidney Failure, Chronic/mortality , Kidney Failure, Chronic/physiopathology , Kidney Failure, Chronic/prevention & control , Kidney Glomerulus/immunology , Kidney Glomerulus/pathology , Kidney Glomerulus/physiopathology , Male , Proteinuria/immunology , Proteinuria/mortality , Proteinuria/physiopathology , Retrospective Studies , Risk Factors , Time Factors , Treatment OutcomeABSTRACT
BACKGROUND: Angiotensin II type 1 receptors (AT1Rs) are expressed on podocytes, endothelial and other cells, and play an essential role in the maintenance of podocyte function and vascular homeostasis. The presence of AT1R antibodies (AT1R-Abs) leads to activation of these receptors resulting in podocyte injury and endothelial cell dysfunction. We assessed the correlation between AT1R-Abs and the risk of post-transplant FSGS. METHODS: This is a retrospective study, which included all kidney transplant recipients with positive AT1R-Abs (≥ 9 units/ml), who were transplanted and followed at our center between 2006 and 2016. We assessed the development of biopsy proven FSGS and proteinuria by urine protein to creatinine ratio of ≥1 g/g and reviewed short and long term outcomes. RESULTS: We identified 100 patients with positive AT1R-Abs at the time of kidney transplant biopsy or proteinuria. 49% recipients (FSGS group) had biopsy-proven FSGS and/or proteinuria and 51% did not (non-FSGS group). Pre-transplant hypertension was present in 89% of the FSGS group compared to 72% in the non-FSGS group, p = 0.027. Of the FSGS group, 43% were on angiotensin converting enzyme inhibitors or angiotensin receptor blockers prior to transplantation, compared to 25.5% in the non-FSGS group, p = 0.06. Primary idiopathic FSGS was the cause of ESRD in 20% of the FSGS group, compared to 6% in the non-FSGS group, p = 0.03. The allograft loss was significantly higher in the FSGS group 63% compared to 39% in non-FSGS. Odds ratio and 95% confidence interval were 2.66 (1.18-5.99), p = 0.017. CONCLUSIONS: Our data suggest a potential association between AT1R-Abs and post-transplant FSGS leading to worse allograft outcome. Therefore, AT1R-Abs may be considered biomarkers for post-transplant FSGS.
Subject(s)
Antibodies/immunology , Glomerulosclerosis, Focal Segmental/epidemiology , Graft Rejection/epidemiology , Kidney Failure, Chronic/surgery , Kidney Transplantation , Postoperative Complications/epidemiology , Proteinuria/epidemiology , Receptor, Angiotensin, Type 1/immunology , Adult , Female , Glomerulosclerosis, Focal Segmental/complications , Glomerulosclerosis, Focal Segmental/immunology , Glomerulosclerosis, Focal Segmental/therapy , Graft Rejection/immunology , Graft Rejection/therapy , Humans , Kidney Failure, Chronic/etiology , Male , Middle Aged , Postoperative Complications/immunology , Postoperative Complications/therapy , Proteinuria/immunology , Proteinuria/therapy , Recurrence , Retrospective StudiesABSTRACT
Systemic lupus erythematosus (SLE) is an autoimmune disease in which the main contributors to organ damage are antibodies against autoantigens, such as double-stranded DNA (dsDNA). Calorie restriction and intermittent fasting (IF) have been shown to improve autoimmune disease symptoms in patients and animal models. Here, we tested the hypothesis that IF might improve symptoms in MRL/lpr mice, which spontaneously develop an SLE-like disease. Groups of mice were fed every other day (IF) or provided food ad libitum (controls), and various lupus-associated clinicopathological parameters were analyzed for up to 28 weeks. Contrary to expectations, anti-dsDNA antibody levels, immune complex deposition in the kidney, and glomerular injury were higher in the IF group than the control group, although there were no differences in spleen and lymph node weights between groups. Proteinuria was also worsened in the IF group. IF also increased the abundance of B cells, plasmablasts, and plasma cells and elevated autophagy in plasma cells in the spleen and lymph nodes. Secretion of anti-dsDNA antibody by splenocytes in vitro was reduced by chloroquine-induced inhibition of autophagy. These results suggest that IF exacerbates lupus nephritis in MRL/lpr mice by increasing autoantibody immune complex formation.
Subject(s)
Antibody-Producing Cells/immunology , Autophagy/immunology , Fasting/physiology , Lupus Nephritis/immunology , Lupus Nephritis/pathology , Animals , Antibodies, Antinuclear/immunology , Antibody-Producing Cells/pathology , Autoantibodies/immunology , B-Lymphocytes/immunology , B-Lymphocytes/pathology , Disease Models, Animal , Female , Kidney Glomerulus/immunology , Kidney Glomerulus/pathology , Mice , Mice, Inbred MRL lpr , Proteinuria/immunology , Proteinuria/pathology , Spleen/immunology , Spleen/pathologyABSTRACT
Growth differentiation factor 15 (GDF15) is a member of the transforming growth factor-ß (TGF-ß) cytokine family and an inflammation-associated protein. Here, we investigated the role of GDF15 in murine anti-glomerular basement membrane (GBM) glomerulonephritis. Glomerulonephritis induction in mice induced systemic expression of GDF15. Moreover, we demonstrate the protective effects for GDF15, as GDF15-deficient mice exhibited increased proteinuria with an aggravated crescent formation and mesangial expansion in anti-GBM nephritis. Herein, GDF15 was required for the regulation of T-cell chemotactic chemokines in the kidney. In addition, we found the upregulation of the CXCR3 receptor in activated T-cells in GDF15-deficient mice. These data indicate that CXCL10/CXCR3-dependent-signaling promotes the infiltration of T cells into the organ during acute inflammation controlled by GDF15. Together, these results reveal a novel mechanism limiting the migration of lymphocytes to the site of inflammation during glomerulonephritis.
Subject(s)
Cell Movement/immunology , Glomerular Basement Membrane/immunology , Glomerulonephritis, Membranous/immunology , Growth Differentiation Factor 15/immunology , Proteinuria/immunology , T-Lymphocytes/immunology , Animals , Cell Movement/genetics , Chemokine CXCL10/genetics , Chemokine CXCL10/immunology , Glomerular Basement Membrane/pathology , Glomerulonephritis, Membranous/genetics , Glomerulonephritis, Membranous/pathology , Growth Differentiation Factor 15/genetics , Mice , Mice, Knockout , Proteinuria/genetics , Proteinuria/pathology , Receptors, CXCR3/genetics , Receptors, CXCR3/immunology , T-Lymphocytes/pathologyABSTRACT
Glomerulonephritis (GN) is a common cause of end-stage kidney disease and is characterized by glomerular inflammation, hematuria, proteinuria, and progressive renal dysfunction. Transforming growth factor (TGF)-ß is involved in glomerulosclerosis and interstitial fibrosis. TGF-ß activates multiple signaling pathways, including the canonical SMAD pathway. We evaluated the role of SMAD signaling in renal injury and proteinuria in a murine model of GN. SMAD3+/+ or SMAD3-/- mice received anti-glomerular basement membrane antibodies to induce GN. We confirmed previous reports that demonstrated that SMAD3 is an important mediator of glomerulosclerosis and renal interstitial fibrosis. Proteinuria was highly SMAD3 dependent. We found differential effects of SMAD3 deletion on podocytes and glomerular endothelial cells. GN led to podocyte injury, including foot process effacement and loss of podocyte-specific markers. Interestingly, these changes were not SMAD3 dependent. Furthermore, there were significant changes to glomerular endothelial cells, including loss of fenestrations, swelling, and basement membrane reduplication, which were SMAD3 dependent. Despite ongoing markers of podocyte injury in SMAD3-/- mice, proteinuria was transient. Renal injury in the setting of GN involves TGF-ß and SMAD3 signaling. Cell populations within the glomerulus respond differently to SMAD3 deletion. Proteinuria correlated more with endothelial cell changes as opposed to podocyte injury in this model.
Subject(s)
Anti-Glomerular Basement Membrane Disease/metabolism , Kidney Glomerulus/metabolism , Smad3 Protein/metabolism , Animals , Anti-Glomerular Basement Membrane Disease/genetics , Anti-Glomerular Basement Membrane Disease/immunology , Anti-Glomerular Basement Membrane Disease/pathology , Autoantibodies , Cell Line , Disease Models, Animal , Endothelial Cells/immunology , Endothelial Cells/metabolism , Endothelial Cells/pathology , Fibrosis , Kidney Glomerulus/immunology , Kidney Glomerulus/pathology , Membrane Proteins/metabolism , Mice, Inbred C57BL , Mice, Knockout , Microfilament Proteins/metabolism , Paracrine Communication , Podocytes/immunology , Podocytes/metabolism , Podocytes/pathology , Proteinuria/immunology , Proteinuria/metabolism , Signal Transduction , Smad3 Protein/deficiency , Smad3 Protein/genetics , Transforming Growth Factor beta/metabolismABSTRACT
The discovery in 2009 of the M-type phospholipase A2 receptor (PLA2R) as the primary target in membranous nephropathy (MN) greatly advanced basic and clinical research. Primary MN is now considered a renal-limited autoimmune disease, with antibodies against PLA2R (aPLA2Rab) identified in 70-80 % of patients of various ethnic groups. Although the use of aPLA2Rab as a diagnostic and prognostic biomarker is now widely accepted, many questions related to the development of the auto-immune response, the role of IgG subclasses and antigenic epitopes, and the pathways to podocyte injury remain unresolved. PLA2R-associated MN most likely develops governed by factors such as genetic susceptibility, loss of tolerance, alterations in antigen expression with a role for environmental factors like air pollution, smoking, and infections. More detailed knowledge of genetic factors, the relevant B- and T-cell epitopes, and the mechanisms of podocyte injury is needed to identify patients at risk for disease progression and to develop optimized, targeted treatment strategies. In this review we highlight unresolved issues, addressing initiation of antibody formation, the timeline of antibody production, the role of IgG subclass, and the pathogenicity of the antibodies in concert with complement to produce glomerular pathology and proteinuria.
Subject(s)
Glomerulonephritis, Membranous/immunology , Receptors, Phospholipase A2/immunology , Antibody Formation , Glomerulonephritis, Membranous/therapy , Humans , Molecular Targeted Therapy , Proteinuria/immunologyABSTRACT
In mice, the initial stage of nephrotoxic serum-induced nephritis (NTN) mimics antibody-mediated human glomerulonephritis. Local immune deposits generate tumor necrosis factor (TNF), which activates pro-inflammatory pathways in glomerular endothelial cells (GECs) and podocytes. Because TNF receptors mediate antibacterial defense, existing anti-TNF therapies can promote infection; however, we have previously demonstrated that different functional domains of TNF may have opposing effects. The TIP peptide mimics the lectin-like domain of TNF, and has been shown to blunt inflammation in acute lung injury without impairing TNF receptor-mediated antibacterial activity. We evaluated the impact of TIP peptide in NTN. Intraperitoneal administration of TIP peptide reduced inflammation, proteinuria, and blood urea nitrogen. The protective effect was blocked by the cyclooxygenase inhibitor indomethacin, indicating involvement of prostaglandins. Targeted glomerular delivery of TIP peptide improved pathology in moderate NTN and reduced mortality in severe NTN, indicating a local protective effect. We show that TIP peptide activates the epithelial sodium channel(ENaC), which is expressed by GEC, upon binding to the channel's α subunit. In vitro, TNF treatment of GEC activated pro-inflammatory pathways and decreased the generation of prostaglandin E2 and nitric oxide, which promote recovery from NTN. TIP peptide counteracted these effects. Despite the capacity of TIP peptide to activate ENaC, it did not increase mean arterial blood pressure in mice. In the later autologous phase of NTN, TIP peptide blunted the infiltration of Th17 cells. By countering the deleterious effects of TNF through direct actions in GEC, TIP peptide could provide a novel strategy to treat glomerular inflammation.
Subject(s)
Epithelial Sodium Channels/metabolism , Glomerulonephritis/drug therapy , Kidney Glomerulus/drug effects , Peptides, Cyclic/administration & dosage , Proteinuria/drug therapy , Animals , Blood Urea Nitrogen , Cell Line , Dinoprostone/metabolism , Disease Models, Animal , Endothelial Cells/drug effects , Endothelial Cells/pathology , Female , Glomerulonephritis/blood , Glomerulonephritis/immunology , Glomerulonephritis/pathology , Humans , Injections, Intraperitoneal , Kidney Glomerulus/cytology , Kidney Glomerulus/pathology , Mice , Nitric Oxide/metabolism , Patch-Clamp Techniques , Primary Cell Culture , Proteinuria/blood , Proteinuria/immunology , Proteinuria/pathology , Signal Transduction/drug effects , Signal Transduction/immunology , Th17 Cells/drug effects , Th17 Cells/immunology , Tumor Necrosis Factor-alpha/immunology , Tumor Necrosis Factor-alpha/metabolismABSTRACT
Antibodies against THSD7A (thrombospondin type 1 domain-containing protein 7A) have been proposed to play a causal role in the development of nephrotic syndrome in patients with THSD7A antibody-positive membranous nephropathy. We hypothesized that removal of these antibodies from plasma could lead to a rapid reduction in proteinuria. Using immunoadsorption to reduce THSD7A antibodies led to a rapid reduction in proteinuria in 2 patients with THSD7A antibody-positive membranous nephropathy. Moreover, our findings support and strengthen the pathogenic role of the antibodies in the development of nephrotic syndrome in patients with THSD7A antibody-positive membranous nephropathy. Taken together, these 2 cases suggest that immunoadsorption could be a useful tool in the treatment of patients with THSD7A antibody-positive membranous nephropathy.
Subject(s)
Autoantibodies/immunology , Glomerulonephritis, Membranous/immunology , Glomerulonephritis, Membranous/therapy , Nephrotic Syndrome/pathology , Thrombospondins/immunology , Adult , Aged , Biopsy, Needle , Disease Progression , Glomerulonephritis, Membranous/pathology , Humans , Immunohistochemistry , Male , Nephrotic Syndrome/physiopathology , Plasmapheresis , Prognosis , Proteinuria/immunology , Proteinuria/physiopathology , Risk Assessment , Sampling Studies , Treatment OutcomeABSTRACT
Ox40 ligand (Ox40L) locus genetic variants are associated with the risk for systemic lupus erythematosus (SLE); however, it is unclear how Ox40L contributes to SLE pathogenesis. In this study, we evaluated the contribution of Ox40L and its cognate receptor, Ox40, using in vivo agonist and antagonist approaches in the NZB × NZW (NZB/W) F1 mouse model of SLE. Ox40 was highly expressed on several CD4 Th cell subsets in the spleen and kidney of diseased mice, and expression correlated with disease severity. Treatment of aged NZB/W F1 mice with agonist anti-Ox40 mAbs potently exacerbated renal disease, which was accompanied by activation of kidney-infiltrating T cells and cytokine production. The agonist mAbs also induced activation and inflammatory gene expression in splenic CD4 T cells, including IFN-regulated genes, increased the number of follicular helper T cells and plasmablasts in the spleen, and led to elevated levels of serum IgM and enhanced renal glomerular IgM deposition. In a type I IFN-accelerated lupus model, treatment with an antagonist Ox40:Fc fusion protein significantly delayed the onset of severe proteinuria and improved survival. These data support the hypothesis that the Ox40/Ox40L pathway drives cellular and humoral autoimmune responses during lupus nephritis in NZB/W F1 mice and emphasize the potential clinical value of targeting this pathway in human lupus.
Subject(s)
Lupus Nephritis/immunology , Membrane Glycoproteins/metabolism , Plasma Cells/immunology , Receptors, OX40/metabolism , T-Lymphocytes, Helper-Inducer/immunology , Tumor Necrosis Factors/metabolism , Animals , Antibodies, Antinuclear/immunology , Antibodies, Monoclonal/immunology , Autoimmunity , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , Disease Models, Animal , Female , Humans , Kidney/immunology , Kidney/pathology , Kidney Glomerulus/immunology , Kidney Glomerulus/pathology , Lupus Nephritis/physiopathology , Mice , Mice, Inbred NZB , OX40 Ligand , Proteinuria/immunology , Signal Transduction , T-Lymphocytes, Helper-Inducer/metabolismABSTRACT
Properdin is known as the only positive regulator of the complement system. Properdin promotes the activity of this defense system by stabilizing its key enzymatic complexes: the complement alternative pathway (AP) convertases. Besides, some studies have indicated a role for properdin as an initiator of complement activity. Though the AP is a powerful activation route of the complement system, it is also involved in a wide variety of autoimmune and inflammatory diseases, many of which affect the kidneys. The role of properdin in regulating complement in health and disease has not received as much appraisal as the many negative AP regulators, such as factor H. Historically, properdin deficiency has been strongly associated with an increased risk for meningococcal disease. Yet only recently had studies begun to link properdin to other complement-related diseases, including renal diseases. In the light of the upcoming complement-inhibiting therapies, it is interesting whether properdin can be a therapeutic target to attenuate AP-mediated injury. A full understanding of the basic concepts of properdin biology is therefore needed. Here, we first provide an overview of the function of properdin in health and disease. Then, we explore its potential as a therapeutic target for the AP-associated renal diseases C3 glomerulopathy, atypical hemolytic uremic syndrome, and proteinuria-induced tubulointerstitial injury. Considering current knowledge, properdin-inhibiting therapy seems promising in certain cases. However, knowing the complexity of properdin's role in renal pathologies in vivo, further research is required to clarify the exact potential of properdin-targeted therapy in complement-mediated renal diseases.
Subject(s)
Atypical Hemolytic Uremic Syndrome/immunology , Complement Pathway, Alternative , Glomerulonephritis, Membranoproliferative/immunology , Nephritis, Interstitial/immunology , Properdin/metabolism , Atypical Hemolytic Uremic Syndrome/drug therapy , Complement C3/immunology , Complement C3/metabolism , Complement C3 Convertase, Alternative Pathway/metabolism , Glomerulonephritis, Membranoproliferative/drug therapy , Humans , Immunologic Factors/pharmacology , Immunologic Factors/therapeutic use , Nephritis, Interstitial/complications , Nephritis, Interstitial/drug therapy , Nephritis, Interstitial/urine , Properdin/antagonists & inhibitors , Protein Stability/drug effects , Proteinuria/immunologyABSTRACT
Immunoglobulin A nephropathy (IgAN) is characterized by mesangial IgA and IgG co-deposition. As the clinical course of IgAN is highly variable, a lot of patients will eventually develop to end-stage renal disease (ESRD) within years. Hirudin, a potent and specific thrombin inhibitor, has been reported to treat IgAN with hematuria, but the mechanism is unclear. Our study aims to explore the potential of hirudin and the underlying mechanism in the treatment of IgAN. The establishment of IgAN model was set up in rats through oral and intravenous immunization with bovine gamma-globulin (BGG). Results suggested that hirudin could reduce the increased level of proteinuria, serum creatinine and urea nitrogen in IgAN models. Besides that, hirudin ameliorated the elevated number of apoptotic bodies and expressions of apoptosis-related proteins (caspase-3 and caspase-9) in IgAN model. The fibrosis indexes (transforming growth factor ß-1 (TGF-ß1), Collagen-IV (CoI-IV) and Fibronectin-1) of kidney were remarkably suppressed in IgAN rats treated with hirudin compared with IgAN rats with no further treatment. IgAN rats exhibited remarkably increased inflammatory factors (IL-1ß, IL-6, and IL-18), while hirudin treatment significantly alleviated these alterations. Moreover, the reduced levels of CD4+CD25+Foxp3+ Treg and CD4+IFN-γ+ Th1/CD4+IL-4+ Th2 could be reversed by hirudin in IgAN model. Furthermore, in the process of IgAN, hirudin could inactivate various pathways (IκBα, NF-κB, TNF-α, and VCAM-1) compared with IgAN model group. Taken together, our study indicated that hirudin could ameliorate IgAN through suppressing fibrosis and inflammatory response. These findings provide a new therapeutic method to treat IgAN.