Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 163
Filter
1.
Adv Kidney Dis Health ; 31(3): 206-215, 2024 May.
Article in English | MEDLINE | ID: mdl-39004460

ABSTRACT

Anti-glomerular basement membrane disease is a small-vessel vasculitis involving the kidneys (∼90%) and the lungs (∼60%). Antibodies against the glomerular basement membrane are directly pathogenic in anti-glomerular basement membrane disease; however, recent research has highlighted the critical role of T cells. Novel autoantigens within the glomerular basement membrane are also now recognized. Atypical forms of the disease are reported along with preceding triggers, such as immune checkpoint inhibitors, immunomodulatory drugs, and vaccines. Kidney outcomes in anti-glomerular basement membrane disease remain poor despite significant improvement in patient survival in the last 2 to 3 decades. Treatment typically relies on combined plasmapheresis with intensive immunosuppression. Dialysis dependency at presentation is a dominant predictor of kidney outcome. Histologically, a low (<10%) percentage of normal glomeruli, 100% crescents, together with dialysis dependency at presentation, is associated with poor kidney outcomes. In such cases, an individualized approach weighing the risks and benefits of treatment is recommended. There is a need for better ways to stop the toxic inflammatory activity associated with this disease. In this narrative review, we discuss recent updates on the pathogenesis and management of anti-glomerular basement membrane disease relevant to patients of all ages.


Subject(s)
Anti-Glomerular Basement Membrane Disease , Humans , Anti-Glomerular Basement Membrane Disease/therapy , Anti-Glomerular Basement Membrane Disease/immunology , Anti-Glomerular Basement Membrane Disease/diagnosis , Anti-Glomerular Basement Membrane Disease/pathology , Plasmapheresis , Autoantibodies/immunology , Renal Dialysis , Immunosuppressive Agents/therapeutic use , Autoantigens/immunology , Glomerular Basement Membrane/immunology , Glomerular Basement Membrane/pathology
2.
Adv Kidney Dis Health ; 31(3): 216-222, 2024 May.
Article in English | MEDLINE | ID: mdl-39004461

ABSTRACT

Membranoproliferative glomerulonephritis (MPGN) is no longer a disease but a pattern of injury in various diseases. Characterized by electron-dense deposits, mesangial proliferation, and duplication of the glomerular basement membrane, MPGN was previously classified by findings seen by electron microscopy. However, recognizing complement dysfunction in relation to cases with the MPGN pattern of injury substantially changed our view of its pathogenesis. A new classification, including immune complex-mediated and complement-mediated MPGN, has become preferable and has been adopted by international guidelines. Despite these advancements, accurate diagnosis of MPGN remains a clinical challenge, given the pathological and clinical similarities between immune complex-mediated and complement-mediated MPGN. Additional testing, such as molecular and genetic testing, is often necessary. Here, we will summarize our current understanding of the MPGN pattern of injury from a pathology perspective as an introductory article in the following chapters.


Subject(s)
Glomerulonephritis, Membranoproliferative , Humans , Glomerulonephritis, Membranoproliferative/pathology , Glomerulonephritis, Membranoproliferative/immunology , Glomerulonephritis, Membranoproliferative/diagnosis , Microscopy, Electron , Complement System Proteins/genetics , Complement System Proteins/immunology , Glomerular Basement Membrane/pathology , Glomerular Basement Membrane/immunology , Kidney Glomerulus/pathology , Kidney Glomerulus/immunology
3.
Am J Kidney Dis ; 83(6): 713-728.e1, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38171412

ABSTRACT

RATIONALE & OBJECTIVE: Atypical anti-glomerular basement membrane (GBM) nephritis is characterized by a bright linear immunoglobulin staining along the GBM by immunofluorescence without a diffuse crescentic glomerulonephritis nor serum anti-GBM antibodies by conventional enzyme-linked immunosorbent assay (ELISA). We characterized a series of patients with atypical anti-GBM disease. STUDY DESIGN: Case series. SETTING & PARTICIPANTS: Patients identified by the French Nephropathology Group as having atypical anti-GBM nephritis between 2003 and 2022. FINDINGS: Among 38 potential cases, 25 were included, of whom 14 (56%) were female and 23 (92%) had hematuria. The median serum creatinine at diagnosis was 150 (IQR, 102-203) µmol/L and median urine protein-creatinine ratio (UPCR) was 2.4 (IQR, 1.3-5.2) g/g. Nine patients (36%) had endocapillary proliferative glomerulonephritis (GN), 4 (16%) had mesangial proliferative GN, 4 (16%) had membranoproliferative GN, 2 (8%) had pure and focal crescentic GN, 1 (4%) had focal segmental glomerulosclerosis, and 5 had glomeruli that were unremarkable on histopathology. Nine patients (36%) had crescents, involving a median of 9% of glomeruli. Bright linear staining for IgG was seen in 22 cases (88%) and for IgA in 3 cases (12%). The 9 patients (38%) who had a monotypic staining pattern tended to be older with less proteinuria and rarely had crescents. Kidney survival rate at 1 year was 83% and did not appear to be associated with the light chain restriction. LIMITATIONS: Retrospective case series with a limited number of biopsies including electron microscopy. CONCLUSIONS: Compared with typical anti-GBM disease, atypical anti-GBM nephritis frequently presents with an endocapillary or mesangial proliferative glomerulonephritis pattern and appears to have a slower disease progression. Further studies are needed to fully characterize its pathophysiology and associated clinical outcomes. PLAIN-LANGUAGE SUMMARY: Atypical anti-glomerular basement membrane (GBM) nephritis is characterized histologically by bright linear immunoglobulin staining along the GBM without diffuse crescentic glomerulonephritis or circulating anti-GBM antibodies. We report a case series of 25 atypical cases of anti-GBM nephritis in collaboration with the French Nephropathology Group. Compared with typical anti-GBM disease, we observed a slower disease progression. Patients frequently presented with heavy proteinuria and commonly had evidence of endocapillary or mesangial proliferative glomerulonephritis. About half of the patients displayed a monotypic immune staining pattern; they tended to be older, with less proteinuria, and commonly without glomerular crescents in biopsy specimens. No concomitant circulating monoclonal gammopathy was detected. Further studies are needed to fully characterize its pathophysiology and associated clinical outcomes.


Subject(s)
Anti-Glomerular Basement Membrane Disease , Humans , Female , Male , Anti-Glomerular Basement Membrane Disease/diagnosis , Anti-Glomerular Basement Membrane Disease/pathology , Anti-Glomerular Basement Membrane Disease/immunology , Adult , Middle Aged , France/epidemiology , Retrospective Studies , Aged , Glomerular Basement Membrane/pathology , Glomerular Basement Membrane/immunology , Glomerular Basement Membrane/ultrastructure , Autoantibodies
4.
J Am Soc Nephrol ; 32(11): 2777-2794, 2021 11.
Article in English | MEDLINE | ID: mdl-34716242

ABSTRACT

BACKGROUND: Autoantibodies binding to podocyte antigens cause idiopathic membranous glomerulonephritis (iMGN). However, it remains elusive how autoantibodies reach the subepithelial space because the glomerular filtration barrier (GFB) is size selective and almost impermeable for antibodies. METHODS: Kidney biopsies from patients with iMGN, cell culture, zebrafish, and mouse models were used to investigate the role of nephronectin (NPNT) regulating microRNAs (miRs) for the GFB. RESULTS: Glomerular endothelial cell (GEC)-derived miR-192-5p and podocyte-derived miR-378a-3p are upregulated in urine and glomeruli of patients with iMGN, whereas glomerular NPNT is reduced. Overexpression of miR-192-5p and morpholino-mediated npnt knockdown induced edema, proteinuria, and podocyte effacement similar to podocyte-derived miR-378a-3p in zebrafish. Structural changes of the glomerular basement membrane (GBM) with increased lucidity, splitting, and lamellation, especially of the lamina rara interna, similar to ultrastructural findings seen in advanced stages of iMGN, were found. IgG-size nanoparticles accumulated in lucidity areas of the lamina rara interna and lamina densa of the GBM in npnt-knockdown zebrafish models. Loss of slit diaphragm proteins and severe structural impairment of the GBM were further confirmed in podocyte-specific Npnt knockout mice. GECs downregulate podocyte NPNT by transfer of miR-192-5p-containing exosomes in a paracrine manner. CONCLUSIONS: Podocyte NPNT is important for proper glomerular filter function and GBM structure and is regulated by GEC-derived miR-192-5p and podocyte-derived miR-378a-3p. We hypothesize that loss of NPNT in the GBM is an important part of the initial pathophysiology of iMGN and enables autoantigenicity of podocyte antigens and subepithelial immune complex deposition in iMGN.


Subject(s)
Endothelial Cells/metabolism , Extracellular Matrix Proteins/biosynthesis , Glomerular Basement Membrane/metabolism , Glomerular Basement Membrane/physiopathology , Glomerulonephritis, Membranous/genetics , Kidney Glomerulus/metabolism , MicroRNAs/physiology , Animals , Antigen-Antibody Complex/analysis , Autoantigens/genetics , Autoantigens/immunology , Cells, Cultured , Coculture Techniques , Exosomes/metabolism , Extracellular Matrix Proteins/deficiency , Extracellular Matrix Proteins/physiology , Gene Expression Regulation , Gene Targeting , Glomerular Basement Membrane/immunology , Glomerular Basement Membrane/ultrastructure , Glomerulonephritis, Membranous/immunology , Glomerulonephritis, Membranous/metabolism , Glomerulonephritis, Membranous/physiopathology , Gold Sodium Thiosulfate , Humans , Metal Nanoparticles , Mice , MicroRNAs/biosynthesis , MicroRNAs/genetics , MicroRNAs/urine , Paracrine Communication , Permeability , Podocytes/immunology , Podocytes/metabolism , Proteinuria/etiology , Transfection , Zebrafish , Zebrafish Proteins/deficiency , Zebrafish Proteins/genetics
5.
J Pathol ; 255(2): 107-119, 2021 10.
Article in English | MEDLINE | ID: mdl-34124781

ABSTRACT

Experimental autoimmune vasculitis (EAV) is a model of antineutrophil cytoplasm antibody (ANCA)-associated vasculitis (AAV) induced by immunisation of susceptible rat strains with myeloperoxidase (MPO). Animals develop circulating MPO-ANCA, pulmonary haemorrhage, and glomerulonephritis, although renal injury is mild and recovers spontaneously without treatment. In this study we aimed to augment the severity of glomerulonephritis. Following induction of EAV on day 0, a sub-nephritogenic dose of nephrotoxic serum (NTS) containing heterologous antibodies to glomerular basement membrane was administered on day 14. This resulted in a significant increase in disease severity at day 28 compared to MPO immunisation alone - with more urinary abnormalities, infiltrating glomerular leucocytes, and crescent formation that progressed to glomerular and tubulointerstitial scarring by day 56, recapitulating important features of human disease. Importantly, the glomerulonephritis remained pauci-immune, and was strictly dependent on the presence of autoimmunity to MPO, as there was no evidence of renal disease following administration of sub-nephritogenic NTS alone or after immunisation with a control protein in place of MPO. Detailed phenotyping of glomerular leucocytes identified an early infiltrate of non-classical monocytes following NTS administration that, in the presence of autoimmunity to MPO, may initiate the subsequent influx of classical monocytes which augment glomerular injury. We also showed that this model can be used to test novel therapeutics by using a small molecule kinase inhibitor (fostamatinib) that rapidly attenuated both glomerular and pulmonary injury over a 4-day treatment period. We believe that this enhanced model of MPO-AAV will prove useful for the study of glomerular leucocyte behaviour and novel therapeutics in AAV in the future. © 2021 The Authors. The Journal of Pathology published by John Wiley & Sons, Ltd. on behalf of The Pathological Society of Great Britain and Ireland.


Subject(s)
Anti-Neutrophil Cytoplasmic Antibody-Associated Vasculitis/immunology , Autoantigens/immunology , Disease Models, Animal , Glomerulonephritis/immunology , Peroxidase/immunology , Animals , Autoantibodies/immunology , Glomerular Basement Membrane/immunology , Male , Rats
6.
Nephron ; 145(4): 428-444, 2021.
Article in English | MEDLINE | ID: mdl-33910203

ABSTRACT

BACKGROUND/AIM: Necrotizing crescentic glomerulonephritis (GN) associated with anti-neutrophil cytoplasmic antibodies (ANCA) against myeloperoxidase (MPO) is a devastating disease that quickly progresses to kidney failure. Current therapies are broadly immunosuppressive and associated with adverse effects. We wanted to set up a model that could be suitable for testing narrowly targeted therapies. METHODS: The model was constructed in male Wistar Kyoto rats through injections of human MPO (hMPO) and pertussis toxin, followed by a sub-nephritogenic dose of sheep anti-rat glomerular basement membrane (GBM) serum to boost the disease. Rats were monitored for 35 days. Rats given hMPO alone, saline, or human serum albumin with or without anti-GBM serum were also studied. RESULTS: Rats receiving hMPO developed circulating anti-hMPO and anti-rat MPO antibodies. Challenging hMPO-immunized rats with the anti-GBM serum led to more glomerular neutrophil infiltration and MPO release, and severe haematuria, heavy proteinuria, and higher blood urea nitrogen than hMPO alone. Pauci-immune GN developed with crescents, affecting 25% of glomeruli. The majority of crescents were fibrocellular. Necrotizing lesions and Bowman capsule ruptures were detected. Cells double positive for claudin-1 (a marker of parietal epithelial cells [PECs]) and neural cell adhesion molecule (NCAM; progenitor PECs) were present in crescents. Double staining for NCAM and Ki-67 established proliferative status of progenitor PECs. Podocyte damage was associated with endothelial and GBM changes by electron microscopy. Monocyte/macrophages and CD4+ and CD8+ T cells accumulated in glomeruli and the surrounding area and in the tubulointerstitium. Lung haemorrhage also manifested. CONCLUSION: This model reflects histological lesions of human ANCA-associated rapidly progressive GN and may be useful for investigating new therapies.


Subject(s)
Antibodies, Antineutrophil Cytoplasmic/immunology , Glomerulonephritis/immunology , Peroxidase/immunology , Animals , Blood Urea Nitrogen , Bowman Capsule/pathology , Epithelial Cells/pathology , Glomerular Basement Membrane/immunology , Hematuria/etiology , Humans , Kidney Glomerulus/pathology , Male , Neutrophil Infiltration , Pertussis Toxin/pharmacology , Proteinuria/etiology , Rats , Rats, Inbred WKY
7.
Mol Immunol ; 132: 41-52, 2021 04.
Article in English | MEDLINE | ID: mdl-33545624

ABSTRACT

Pathogens such as the Epstein Barr virus (EBV) have long been implicated in the etiology of systemic lupus erythematosus (SLE). The Epstein Barr virus nuclear antigen I (EBNA-1) has been shown to play a role in the development of anti-nuclear antibodies characteristic of SLE. One mechanism by which EBV may play a role in SLE is molecular mimicry. We previously generated two monoclonal antibodies (mAbs) to EBNA-1 and demonstrated that they cross-react with double-stranded DNA (dsDNA). In the present study, we demonstrate that these mAbs have pathogenic potential. We show that they can bind to isolated rat glomeruli and that binding can be greatly diminished by pretreatment of glomeruli with DNase I, suggesting that these mAbs bind dsDNA in the kidney. We also demonstrate that these antibodies can deposit in the kidney when injected into mice and can induce proteinuria and elicit histopathological alterations consistent with glomerulonephritis. Finally, we show that these antibodies can cross-react with laminin and collagen IV in the extracellular matrix suggesting that direct binding to the glomerular basement membrane or mesangial matrix may also contribute to the antibody deposition in the kidney. In summary, our results indicate that EBNA-1 can elicit antibodies that cross-react with dsDNA, that can deposit in the kidney, and induce kidney damage. These results are significant because they support the role of a viral protein in SLE and lupus nephritis.


Subject(s)
Antibodies, Antinuclear/toxicity , Antibodies, Monoclonal/toxicity , Antibodies, Viral/immunology , DNA/immunology , Epstein-Barr Virus Infections/immunology , Epstein-Barr Virus Nuclear Antigens/immunology , Kidney Glomerulus/immunology , Animals , Antibodies, Antinuclear/immunology , Antibodies, Monoclonal/genetics , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/isolation & purification , Collagen/immunology , Cross Reactions/immunology , Deoxyribonuclease I , Epstein-Barr Virus Infections/metabolism , Extracellular Matrix/genetics , Extracellular Matrix/immunology , Female , Glomerular Basement Membrane/immunology , Glomerular Basement Membrane/metabolism , Glomerulonephritis/immunology , Glomerulonephritis/pathology , Glomerulonephritis/virology , HEK293 Cells , Humans , Immunoglobulin G/immunology , Kidney Glomerulus/pathology , Laminin/immunology , Mice , Mice, Inbred BALB C , Molecular Mimicry , Proteinuria/immunology , Rats , Rats, Sprague-Dawley
8.
Am J Kidney Dis ; 78(2): 219-225.e1, 2021 08.
Article in English | MEDLINE | ID: mdl-33421452

ABSTRACT

RATIONALE & OBJECTIVE: Anti-glomerular basement membrane (GBM) disease is a rapidly progressive glomerulonephritis which, in some instances, occurs concurrently with other diseases such as antineutrophil cytoplasmic antibody (ANCA)-associated vasculitis. Rarely, it also occurs with membranous nephropathy (MN). We report a series of such patients, characterizing their long-term follow up. STUDY DESIGN: Case series. SETTING & PARTICIPANTS: Twelve patients referred to 1 medical center between 2001 and 2019 with anti-GBM disease and MN whose clinical characteristics and course were described. RESULTS: Patients' ages ranged from 20 to 81 years old, and all presented with severe acute kidney injury requiring dialysis on presentation or shortly thereafter. Only 1 patient had pulmonary findings on presentation. The predominant pathology was crescentic and necrotizing glomerulonephritis with linear staining for immunoglobulin G along the GBM associated with epimembranous electron-dense immune-type deposits. All 11 patients who were tested had significant titers of anti-GBM antibodies, but only 1 of the 5 tested for phospholipase A2 receptor (PLA2R) on biopsy was positive. Eight patients received therapy with cyclophosphamide, prednisone, and plasmapheresis; 2 patients with prednisone and plasmapheresis; and 2 with rituximab-based regimens. Progression to a requirement for kidney replacement therapy occurred in all 12 patients, but 2 patients later recovered kidney function. Recurrence of anti-GBM disease did not occur for any of the patients studied. LIMITATIONS: Incomplete testing for PLA2R in biopsy and serum, limited sample size, and lack of uniform treatment regimen. CONCLUSIONS: In this case series, the presentation of concurrent anti-GBM disease and MN was characterized by rapidly progressive glomerulonephritis and poor kidney outcomes. These findings suggest possible value from earlier diagnosis and the need for identification of more effective treatment regimens.


Subject(s)
Anti-Glomerular Basement Membrane Disease/complications , Glomerulonephritis, Membranous/complications , Acute Kidney Injury/etiology , Acute Kidney Injury/therapy , Adult , Aged , Aged, 80 and over , Anti-Glomerular Basement Membrane Disease/immunology , Anti-Glomerular Basement Membrane Disease/therapy , Autoantibodies/immunology , Cyclophosphamide/therapeutic use , Female , Glomerular Basement Membrane/immunology , Glomerulonephritis, Membranous/therapy , Glucocorticoids/therapeutic use , Humans , Immunoglobulin G/immunology , Immunosuppressive Agents/therapeutic use , Male , Middle Aged , Plasmapheresis , Prednisone/therapeutic use , Renal Dialysis , Young Adult
9.
Front Immunol ; 11: 2035, 2020.
Article in English | MEDLINE | ID: mdl-33013861

ABSTRACT

Background: Atypical cases of anti-glomerular basement membrane (GBM) disease had absent circulating antibodies but linear IgG deposits along GBM in the kidneys. Herein, we reported the clinical-pathological features and outcome of these rare cases. Methods: Linear IgG deposit along GBM were examined by immunofluorescence on renal specimens, with exclusion of diabetic kidney disease. Circulating anti-GBM antibodies were tested by commercial ELISA assay. Clinical, pathological and follow-up data were retrospectively analyzed. Results: From 2013 to 2018, a total of 60 patients were diagnosed as atypical anti-GBM disease. They had a male predominance, with an average age of 51.7 ± 15.6 years. Three (5.0%) patients had alveolar hemorrhage. Forty five percent of them presented with acute kidney disease. All patients had linear IgG deposit along GBM, some in addition on tubular basement membrane and/or Bowmans' capsules. C3 deposition was found in 65.0% of the patients. 41.7% (25/60) of the patients showed crescent formation and the percentage of crescent was (34.7 ± 23.5)% in those patients. They had higher prevalence of hematuria and C3 deposit, higher levels of serum creatinine, worse renal and patient survival than those without crescent (P < 0.05). During the follow-up of 35.7 ± 21.4 months, 14 (23.3%) patients progressed to ESRD. The serum creatinine on diagnosis [per 200 µmol/L increase, HR (95% CI): 2.663 (1.372, 5.172), P = 0.004], serum C3 [per 0.1 g/L increase, HR (95% CI): 0.689(0.483, 0.984), P = 0.040] and the intensity of kidney C3 staining [per 1+ increase, HR (95% CI): 2.770 (1.115, 6.877), P = 0.028] were independent predictive factors for kidney outcome. Nine (15.0%) patients died of all causes. Conclusions: Atypical anti-GBM disease manifested milder kidney injury and scarce pulmonary hemorrhage compared to the classical cases. Though heterogeneous, a substantial number of the patients had complement activation and crescent formation. Patients having crescents presented with more severe clinical course and worse outcomes. The poor kidney and patient prognosis emphasize prompt interventions from physicians. The immunosuppressive intervention was not associated with kidney or patient outcome. Further studies are needed to address the optimal therapeutic regimen.


Subject(s)
Anti-Glomerular Basement Membrane Disease/diagnosis , Anti-Glomerular Basement Membrane Disease/etiology , Autoantibodies/immunology , Glomerular Basement Membrane/immunology , Adult , Aged , Anti-Glomerular Basement Membrane Disease/metabolism , Anti-Glomerular Basement Membrane Disease/therapy , Biomarkers , Biopsy , Cohort Studies , Complement C3/immunology , Complement C3/metabolism , Disease Management , Disease Progression , Disease Susceptibility , Female , Fluorescent Antibody Technique , Glomerular Basement Membrane/metabolism , Glomerular Basement Membrane/pathology , Glomerular Basement Membrane/ultrastructure , Humans , Male , Middle Aged , Prognosis , Proportional Hazards Models
10.
Int J Mol Sci ; 21(19)2020 Sep 23.
Article in English | MEDLINE | ID: mdl-32977372

ABSTRACT

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/pathology
12.
Am J Nephrol ; 51(4): 304-317, 2020.
Article in English | MEDLINE | ID: mdl-32097941

ABSTRACT

BACKGROUND: Primary membranous nephropathy (pMN) is less common in women of child-bearing age. The kidney risk factors to adverse maternal-fetal outcomes and the effects of pregnancy on pMN process need to be investigated. METHODS: We retrospectively screened all the patients with biopsy-proven pMN from 2008 to 2018. Any cases of pregnancy that occurred at the time of pMN diagnosis or during follow-up were included in the study. Clinical and pathological data were collected from all patients at the time of kidney biopsy and their gestational results were recorded. RESULTS: Of the 27 pregnancies with gestational time of 35.9 ± 4.5 weeks, 10 adverse maternal-fetal events occurred, including fetal loss (11%), preterm delivery (26%), and severe preeclampsia (15%). The kidney parameters were relatively stable with all preserved kidney function. Time-averaged urinary protein (p < 0.001) and serum albumin (p < 0.001), maximum urinary protein (p = 0.001) and minimum serum albumin (p = 0.01) before week 20, anti-phospholipase A2 receptor (PLA2R) positivity (p = 0.03), and no remission during pregnancy (p = 0.004) were risk factors to adverse maternal-fetal outcomes. Time-averaged urinary protein and serum albumin correlated with the birth weight percentile of neonates. CONCLUSIONS: Pregnancy in pMN patients showed risks to adverse maternal-fetal events. Heavy proteinuria, especially before week 20 of gestation, severe hypoalbuminemia, positive anti-PLA2R, and no remission were risk factors to worse outcomes.


Subject(s)
Autoantibodies/blood , Fetal Death , Glomerulonephritis, Membranous/complications , Pre-Eclampsia/epidemiology , Premature Birth/epidemiology , Adult , Autoantibodies/immunology , Biopsy , Birth Weight/immunology , Female , Glomerular Basement Membrane/immunology , Glomerular Basement Membrane/pathology , Glomerular Basement Membrane/ultrastructure , Glomerulonephritis, Membranous/blood , Glomerulonephritis, Membranous/diagnosis , Glomerulonephritis, Membranous/immunology , Humans , Microscopy, Electron , Pre-Eclampsia/blood , Pre-Eclampsia/immunology , Pre-Eclampsia/urine , Pregnancy , Premature Birth/blood , Premature Birth/immunology , Premature Birth/urine , Receptors, Phospholipase A2/immunology , Retrospective Studies , Risk Factors , Serum Albumin, Human/analysis
13.
Pediatr Nephrol ; 35(5): 829-842, 2020 05.
Article in English | MEDLINE | ID: mdl-32052153

ABSTRACT

BACKGROUND: To date, there is insufficient knowledge about crescentic glomerulonephritis (cGN), the most frequent immunologic cause of acute kidney injury in children. METHODS: Over a period of 16 years, we retrospectively analyzed kidney biopsy results, the clinical course, and laboratory data in 60 pediatric patients diagnosed with cGN. RESULTS: The underlying diseases were immune complex GN (n = 45/60, 75%), including IgA nephropathy (n = 19/45, 42%), lupus nephritis (n = 10/45, 22%), Henoch-Schoenlein purpura nephritis (n = 7/45, 16%) and post-infectious GN (n = 7/45, 16%), ANCA-associated pauci-immune GN (n = 10/60, 17%), and anti-glomerular basement-membrane GN (n = 1/60, 2%). Patient CKD stages at time of diagnosis and at a median of 362 days (range 237-425) were CKD I: n = 13/n = 29, CKD II: n = 15/n = 9, CKD III: n = 16/n = 7, CKD IV: n = 3/n = 3, CKD V: n = 13/n = 5. Course of cGN was different according to class of cGN, duration of disease from first clinical signs to diagnosis of cGN by biopsy, percentage of crescentic glomeruli, amount of tubular atrophy/interstitial fibrosis and necrosis on renal biopsy, gender, age, nephrotic syndrome, arterial hypertension, dialysis at presentation, and relapse. Forty-eight/60 children were treated with ≥ 5 (methyl-) prednisolone pulses and 53 patients received oral prednis(ol)one in combination with mycophenolate mofetil (n = 20), cyclosporine A (n = 20), and/or cyclophosphamide (n = 6), rituximab (n = 5), azathioprine (n = 2), tacrolimus (n = 1), and plasmapheresis/immunoadsorption (n = 5). CONCLUSIONS: The treatment success of cGN is dependent on early diagnosis and aggressive therapy, as well as on the percentage of crescentic glomeruli on renal biopsy and on the underlying type of cGN. CsA and MMF seem to be effective alternatives to cyclophosphamide.


Subject(s)
Acute Kidney Injury/diagnosis , Glomerular Basement Membrane/pathology , Glomerulonephritis/diagnosis , Immunosuppressive Agents/administration & dosage , Plasmapheresis , Acute Kidney Injury/immunology , Acute Kidney Injury/mortality , Acute Kidney Injury/therapy , Adolescent , Antibodies, Antineutrophil Cytoplasmic/blood , Antibodies, Antineutrophil Cytoplasmic/immunology , Biopsy , Child , Child, Preschool , Combined Modality Therapy , Drug Therapy, Combination , Female , Fibrosis , Glomerular Basement Membrane/immunology , Glomerular Filtration Rate , Glomerulonephritis/complications , Glomerulonephritis/immunology , Glomerulonephritis/therapy , Humans , Male , Necrosis/complications , Necrosis/diagnosis , Necrosis/immunology , Necrosis/therapy , Prednisolone/administration & dosage , Prognosis , Pulse Therapy, Drug , Retrospective Studies , Severity of Illness Index , Treatment Outcome
14.
Kidney Int ; 97(1): 163-174, 2020 01.
Article in English | MEDLINE | ID: mdl-31901340

ABSTRACT

Membranous nephropathy is characterized by deposition of immune complexes along the glomerular basement membrane. PLA2R and THSD7A are target antigens in 70% and 1-5% of primary membranous nephropathy cases, respectively. In the remaining cases, the target antigen is unknown. Here, laser microdissection of glomeruli followed by mass spectrometry was used to identify novel antigen(s) in PLA2R-negative membranous nephropathy. An initial pilot mass spectrometry study in 35 cases of PLA2R-negative membranous nephropathy showed high spectral counts for neural tissue encoding protein with EGF-like repeats, NELL-1, in six cases. Mass spectrometry failed to detect NELL-1 in 23 PLA2R-associated membranous nephropathy and 88 controls. NELL-1 was localized by immunohistochemistry, which showed bright granular glomerular basement membrane staining for NELL-1 in all six cases. Next, an additional 23 NELL-1 positive cases of membranous nephropathy were identified by immunohistochemistry in a discovery cohort of 91 PLA2R-negative membranous nephropathy cases, 14 were confirmed by mass spectrometry. Thus, 29 of 126 PLA2R-negative cases were positive for NELL-1. PLA2R-associated membranous nephropathy and controls stained negative for NELL-1. We then identified five NELL-1 positive cases of membranous nephropathy out of 84 PLA2R and THSD7A-negative cases in two validation cohorts from France and Belgium. By confocal microscopy, both IgG and NELL-1 co-localized to the glomerular basement membrane. Western blot analysis showed reactivity to NELL-1 in five available sera, but no reactivity in control sera. Clinical and biopsy findings of NELL-1 positive membranous nephropathy showed features of primary membranous nephropathy. Thus, a subset of membranous nephropathy is associated with accumulation and co-localization of NELL-1 and IgG along the glomerular basement membrane, and with anti-NELL-1 antibodies in the serum. Hence, NELL-1 defines a distinct type of primary membranous nephropathy.


Subject(s)
Autoantigens/immunology , Calcium-Binding Proteins/immunology , Glomerular Basement Membrane/pathology , Glomerulonephritis, Membranous/immunology , Aged , Autoantibodies/analysis , Autoantibodies/blood , Autoantibodies/immunology , Autoantigens/analysis , Biopsy , Calcium-Binding Proteins/analysis , Case-Control Studies , Cohort Studies , Female , Glomerular Basement Membrane/immunology , Glomerular Basement Membrane/ultrastructure , Glomerulonephritis, Membranous/blood , Glomerulonephritis, Membranous/diagnosis , Glomerulonephritis, Membranous/pathology , Humans , Laser Capture Microdissection , Male , Mass Spectrometry , Microscopy, Confocal , Microscopy, Electron , Microscopy, Fluorescence , Middle Aged , Pilot Projects , Receptors, Phospholipase A2/analysis , Receptors, Phospholipase A2/immunology , Thrombospondins/analysis , Thrombospondins/immunology
15.
Ren Fail ; 42(1): 48-53, 2020 Nov.
Article in English | MEDLINE | ID: mdl-31885301

ABSTRACT

Objective: Autoantibodies to the α3 chain noncollagen 1 domain of type IV collagen (α3(IV)NC1) are a serological hallmark in the diagnosis of anti-glomerular basement membrane (GBM) disease. The objective of our study was to compare the performance of anti-glomerular basement membrane (GBM) antibody detection by chemiluminescence immunoassay (CIA) and by enzyme-linked immunosorbent assays (ELISAs).Methods: Sera from outpatients who were suspected to have anti-GBM disease and 31 patients with biopsy-proven anti-GBM disease were collected. Thirty normal controls were also included. All samples were tested for anti-GBM antibodies by CIA and commercial ELISA. The anti-GBM antibody-positive samples were confirmed by a homemade ELISA coated with recombinant human α3(IV)NC1.Results: Compared with detection of anti-GBM antibodies with ELISA, detection of anti-GBM antibodies with CIA showed a positivity agreement of 70% and a negativity agreement of 98.6%. Among the 4 patients with different results, the anti-GBM antibody detection by CIA was in agreement with the homemade ELISA coated with recombinant human α3(IV)NC1 and the clinical diagnosis. In 31 patients with anti-GBM disease, good agreement was achieved in the detection of anti-GBM antibodies with CIA, commercial ELISA and the homemade ELISA (100%, 100%). The AUC for CIA and commercial ELISA was 0.987 and 0.966, respectively.Conclusions: The detection of anti-GBM antibodies with CIA demonstrated good sensitivity and specificity and was in good agreement with our homemade ELISA, which seems better than the commercial ELISA in suspected anti-GBM disease patients. The three assays performed in parallel in the diagnosis of anti-GBM disease patients.


Subject(s)
Anti-Glomerular Basement Membrane Disease/diagnosis , Autoantibodies/isolation & purification , Enzyme-Linked Immunosorbent Assay/methods , Luminescent Measurements/methods , Adult , Aged , Anti-Glomerular Basement Membrane Disease/blood , Anti-Glomerular Basement Membrane Disease/immunology , Anti-Glomerular Basement Membrane Disease/pathology , Autoantibodies/blood , Autoantibodies/immunology , Biopsy , Female , Glomerular Basement Membrane/immunology , Glomerular Basement Membrane/pathology , Humans , Male , Middle Aged , Sensitivity and Specificity
16.
Kidney Int ; 96(6): 1320-1331, 2019 12.
Article in English | MEDLINE | ID: mdl-31530475

ABSTRACT

Mammalian immune systems are not mature until well after birth. However, transfer of maternal IgG to the fetus and newborn usually provides immunoprotection from infectious diseases. IgG transfer occurs before birth in humans across the placenta and continues after birth across the intestine in many mammalian species, including rodents. Transfer, which is mediated by the neonatal IgG Fc receptor, occurs by transcytosis across placental syncytiotrophoblasts and intestinal epithelium. Although maternal IgG is generally beneficial, harmful maternal allo- and autoantibodies can also be transferred to the fetus/infant, resulting in serious disease. To test this we generated transgenic mice that widely express human laminin α5 in their basement membranes. When huLAMA5 transgenic males were crossed with wild-type females, there was a maternal anti-human laminin α5 immune response. Maternal IgG alloantibody crossed the yolk sac and post-natal intestine invivo and bound in bright, linear patterns to kidney glomerular basement membranes of transgenic fetuses/neonates but not those of wild-type siblings. By postnatal day 18, most transgenic mice were proteinuric, had glomerular C3 deposits and inflammatory cell infiltrates, thickened and split glomerular basement membranes, and podocyte foot process effacement. Thus, our novel model of perinatal anti-glomerular basement membrane disease may prove useful for studying pediatric glomerulopathies, formation of the fetomaternal interface, and maternal alloimmunization.


Subject(s)
Anti-Glomerular Basement Membrane Disease/immunology , Glomerular Basement Membrane/immunology , Immunoglobulin G/immunology , Laminin/immunology , Animals , Animals, Newborn , Female , Glomerular Basement Membrane/ultrastructure , Humans , Immunity, Humoral , Male , Mice, Transgenic , Pregnancy
18.
Kidney Int ; 96(4): 942-956, 2019 10.
Article in English | MEDLINE | ID: mdl-31402171

ABSTRACT

Innate lymphoid cells play an important role in the early effector cytokine-mediated response. In Wistar Kyoto rats, CD8+ non-T lymphocytes (CD8+Lym) infiltrate into glomeruli during the development of anti-glomerular basement membrane (anti-GBM) glomerulonephritis. Here, we examined the profiles and roles of CD8+Lym in anti-GBM glomerulonephritis. The regulation of CD8+Lym by peroxisome proliferator-activated receptor (PPAR)-α in anti-GBM glomerulonephritis was also evaluated. Glomerular infiltrating CD8+Lym were lineage-negative cells that showed markedly high expression of IFN-γ and T-bet mRNAs but not Eomes, indicating these cells are group 1 innate lymphoid cells. In anti-GBM glomerulonephritis, the glomerular mRNAs of innate lymphoid cell-related cytokines (IFN-γ and TNF-α) and chemokines (CXCL9, CXCL10, and CXCL11) are significantly increased. Treatment with a PPARα agonist ameliorated renal injury, with reduced expression of these mRNAs. In vitro, enhanced IFN-γ production from innate lymphoid cells upon IL-12 and IL-18 stimulation was reduced by the PPARα agonist. Moreover, CXCL9 mRNA in glomerular endothelial cells and CXCL9, CXCL10, and CXCL11 mRNAs in podocytes and macrophages were upregulated by IFN-γ, whereas the PPARα agonist downregulated their expression. We also detected the infiltration of innate lymphoid cells into glomeruli in human anti-GBM glomerulonephritis. Thus, innate lymphoid cells are involved in the progression of anti-GBM glomerulonephritis and regulated directly or indirectly by PPARα. Our findings suggest that innate lymphoid cells could serve as novel therapeutic targets for anti-GBM glomerulonephritis.


Subject(s)
Anti-Glomerular Basement Membrane Disease/immunology , Glomerular Basement Membrane/pathology , Immunity, Innate , Lymphocyte Subsets/immunology , PPAR alpha/metabolism , Animals , Anti-Glomerular Basement Membrane Disease/drug therapy , Anti-Glomerular Basement Membrane Disease/pathology , Biopsy , CD8 Antigens/metabolism , Cells, Cultured , Chemokines, CXC/immunology , Chemokines, CXC/metabolism , Disease Models, Animal , Disease Progression , Down-Regulation/drug effects , Down-Regulation/immunology , Fenofibrate/pharmacology , Fenofibrate/therapeutic use , Glomerular Basement Membrane/cytology , Glomerular Basement Membrane/immunology , Humans , Lymphocyte Subsets/metabolism , Male , PPAR alpha/agonists , Primary Cell Culture , Rats
19.
Exp Mol Pathol ; 107: 165-170, 2019 04.
Article in English | MEDLINE | ID: mdl-30817909

ABSTRACT

The major epitopes recognized by autoantibodies in anti-glomerular basement membrane (GBM) disease are found in the α3-subunit non-collagenous domain of type IV collagen [α3(IV)NC1], which is present in the glomerular and alveolar basement membranes. These epitopes are structurally cryptic, owing to the hexamer formation of the non-collagenous domain of α3, α4, and α5 subunits and are expressed by the dissociation of the hexamer. Anti-GBM disease usually manifests as a single attack (SA), and we rarely see patients who repeatedly relapse. We recently treated a patient with anti-GBM disease who exhibited repeated relapse (RR). Here, we conducted immunohistochemistry of formalin-fixed paraffin-embedded normal kidney sections and immunoblotting using recombinant human α3(IV)NC1 to compare the epitopes recognized by anti-GBM antibodies in the RR patient and SA patients. Although a clear staining of GBM especially in the connecting basement membrane of Bowman's capsule was observed when IgGs of SA patients were used as primary antibodies, such staining was not obtained when IgG of the RR patient was employed. In immunoblotting of α3(IV)NC1 using the IgG of the RR patient as a primary antibody, an 18-kDa band was detected besides the 56.8-kDa band corresponding to the whole-size α3(IV)NC1. Whereas the 56.8-kDa band disappeared after digestion of the recombinant α3(IV)NC1 by protease, the 18-kDa band remained. Furthermore, the 18-kDa band was not detected by a commercially available anti-α3(IV)NC1 monoclonal antibody. These findings suggest that the IgG of the RR patient recognizes the epitope distinct from that recognized by the anti-α3(IV)NC1 monoclonal antibody.


Subject(s)
Anti-Glomerular Basement Membrane Disease/immunology , Autoantibodies/immunology , Autoantigens/immunology , Epitopes, B-Lymphocyte/immunology , Glomerular Basement Membrane/immunology , Female , Humans , Recurrence , Young Adult
20.
BMC Nephrol ; 20(1): 6, 2019 01 08.
Article in English | MEDLINE | ID: mdl-30621605

ABSTRACT

BACKGROUND: Goodpasture Syndrome (GS) is an autoimmune disease caused by the development of auto-antibodies against the Glomerular Basement Membrane (GBM). Linear deposit of immunoglobulins G on the GBM detected by immunofluorescence analysis of renal biopsies is a GS pathognomonic finding. GS is commonly monophasic and its incidence is 1.6 case per million per year. CASE PRESENTATION: This report describes and discusses the case of a 40-year-old woman who one year after allograft kidney transplant, presented with acute pulmonary and renal symptoms of GS, leading to acute graft dysfunction, without circulating anti-GBM antibody detection in laboratory assays. She received a living donor kidney transplant 4 years after the first diagnosis of GS without circulating anti-GBM antibodies, when considered in remission. CONCLUSIONS: In both episodes, the diagnosis of GS was based exclusively on the kidney biopsy that showed rapidly progressing glomerulonephritis with deposition of immunoglobulins G on the GBM. Although rare, the management of patients with GS without circulating anti-GBM antibodies is difficult due to the lack of standardized follow-up guidelines to reduce the risk of GS recurrence after kidney transplantation.


Subject(s)
Anti-Glomerular Basement Membrane Disease/etiology , Autoantibodies/blood , Kidney Transplantation , Postoperative Complications/etiology , Adult , Anti-Glomerular Basement Membrane Disease/immunology , Anti-Glomerular Basement Membrane Disease/pathology , Anti-Glomerular Basement Membrane Disease/therapy , Female , Glomerular Basement Membrane/immunology , Humans , Immunosuppressive Agents/therapeutic use , Kidney Glomerulus/pathology , Living Donors , Plasma Exchange , Postoperative Complications/immunology , Recurrence , Renal Dialysis , Reoperation
SELECTION OF CITATIONS
SEARCH DETAIL
...