Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 212
Filter
2.
Front Immunol ; 15: 1441017, 2024.
Article in English | MEDLINE | ID: mdl-39185424

ABSTRACT

The most characteristic feature of membranous nephropathy (MN) is the presence of subepithelial electron dense deposits and the consequential thickening of the glomerular basement membrane. There have been great advances in the understanding of the destiny of immune complexes in MN by the benefit of experimental models represented by Heymann nephritis. Subepithelial immune complexes are formed in situ by autoantibodies targeting native autoantigens or exogenous planted antigens such as the phospholipase A2 receptor (PLA2R) and cationic BSA respectively. The nascent immune complexes would not be pathogenic until they develop into immune deposits. Podocytes are the major source of autoantigens in idiopathic membranous nephropathy. They also participate in the modulation and removal of the immune complexes to a large extent. The balance between deposition and clearance is regulated by a wide range of factors such as the composition and physicochemical properties of the immune complexes and the complement system. Complement components such as C3 and C1q have been reported to be precipitated with the deposits whereas a complement regulatory protein CR1 expressed by podocytes is involved in the phagocytosis of immune complexes by podocytes. Podocytes regulate the dynamic change of immune complexes which is disturbed in membranous nephropathy. To elucidate the precise fate of the immune complexes is essential for developing more rational and novel therapies for membranous nephropathy.


Subject(s)
Antigen-Antibody Complex , Glomerulonephritis, Membranous , Podocytes , Glomerulonephritis, Membranous/immunology , Humans , Antigen-Antibody Complex/immunology , Antigen-Antibody Complex/metabolism , Podocytes/immunology , Podocytes/metabolism , Animals , Autoantibodies/immunology , Autoantigens/immunology , Receptors, Phospholipase A2/immunology
4.
Ren Fail ; 46(2): 2373276, 2024 Dec.
Article in English | MEDLINE | ID: mdl-38967134

ABSTRACT

BACKGROUND: Podocytes, as intrinsic renal cells, can also express MHC-II and costimulatory molecules under inflammatory conditions, suggesting that they may act as antigen-presenting cells (APCs) to activate immune cell responses and then lead to immune-mediated renal injury. They are already recognized as main targets in the pathogenic mechanism of hepatitis B virus (HBV)-associated glomerulonephritis (HBV-GN). Previous studies also have indicated that inflammatory cells infiltration and immune-mediated tissue injury are evident in the kidney samples of patients with HBV-GN. However, the role of podocytes immune disorder in the pathogenic mechanism of HBV-GN remains unclear. METHODS: Renal function and inflammatory cells infiltration were measured in HBV transgenic (HBV-Tg) mice. In vitro, podocytes/CD4+ T cells or macrophages co-culture system was established. Then, the expression of HBx, CD4, and CD68 was determined by immunohistochemistry, while the expression of MHC-II, CD40, and CD40L was determined by immunofluorescence. Co-stimulatory molecules expression was examined by flow cytometry. The levels of inflammatory factors were detected by ELISA. RESULTS: In vivo, renal function was obviously impaired in HBV-Tg mice. HBx was significantly upregulated and immune cells infiltrated in the glomerulus of HBV-Tg mice. Expression of MHC-II and costimulatory molecule CD40 increased in the podocytes of HBV-Tg mice; CD4+ T cells exhibited increased CD40L expression in glomerulus. In vitro, CD40 expression was markedly elevated in HBx-podocytes. In co-culture systems, HBx-podocytes stimulated CD4+ T cells activation and caused the imbalance between IFN-γ and IL-4. HBx-podocytes also enhanced the adhesion ability of macrophages and induced the release of proinflammatory mediators. CONCLUSION: Taken together, these podocyte-related immune disorder may be involved in the pathogenic mechanism of HBV-GN.


Subject(s)
Glomerulonephritis , Hepatitis B virus , Mice, Transgenic , Podocytes , Trans-Activators , Viral Regulatory and Accessory Proteins , Animals , Podocytes/immunology , Podocytes/pathology , Podocytes/metabolism , Mice , Trans-Activators/metabolism , Trans-Activators/genetics , Glomerulonephritis/immunology , Glomerulonephritis/pathology , Glomerulonephritis/virology , Hepatitis B virus/immunology , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , Macrophages/immunology , Macrophages/metabolism , Hepatitis B/immunology , Hepatitis B/complications , Humans , Coculture Techniques , Male , Disease Models, Animal , Mice, Inbred C57BL
6.
Cells ; 13(13)2024 Jul 06.
Article in English | MEDLINE | ID: mdl-38995008

ABSTRACT

Accumulating evidence indicates that inflammatory and immunologic processes play a significant role in the development and progression of glomerular diseases. Podocytes, the terminally differentiated epithelial cells, are crucial for maintaining the integrity of the glomerular filtration barrier. Once injured, podocytes cannot regenerate, leading to progressive proteinuric glomerular diseases. However, emerging evidence suggests that podocytes not only maintain the glomerular filtration barrier and are important targets of immune responses but also exhibit many features of immune-like cells, where they are involved in the modulation of the activity of innate and adaptive immunity. This dual role of podocytes may lead to the discovery and development of new therapeutic targets for treating glomerular diseases. This review aims to provide an overview of the innate immunity mechanisms involved in podocyte injury and the progression of proteinuric glomerular diseases.


Subject(s)
Immunity, Innate , Podocytes , Podocytes/immunology , Podocytes/pathology , Humans , Animals , Kidney Diseases/immunology , Kidney Diseases/pathology , Kidney Glomerulus/pathology , Kidney Glomerulus/immunology
7.
Int J Mol Sci ; 25(11)2024 May 29.
Article in English | MEDLINE | ID: mdl-38892120

ABSTRACT

Following the discovery of podocyte phospholipase A2 receptor and thrombospondin type-1 domain-containing 7A, various potential target antigens for membranous nephropathy (MN) have been reported one after another. MN target antigens have now been identified in a significant proportion of patients, and a new classification framework classifies patients with MN based on the detected antigen and associated disease phenotype. A serology-based approach that does not require a histological diagnosis for patients suspected of having MN has also been proposed. However, there have been cases in which dual positivity for MN antigens and/or corresponding antibodies has been shown. Importantly, some of them showed a transition of the affected patient's immune responses to MN antigens, suggesting that serological diagnosis changes depending on the timing of the analysis. In this review, we provide detailed information on these cases and present an overview of our recent understanding of their putative mechanisms involved in these cases. Greater awareness is required to adequately recognize and develop appropriate therapeutic strategies for this condition.


Subject(s)
Glomerulonephritis, Membranous , Glomerulonephritis, Membranous/immunology , Glomerulonephritis, Membranous/diagnosis , Glomerulonephritis, Membranous/blood , Humans , Receptors, Phospholipase A2/immunology , Receptors, Phospholipase A2/metabolism , Autoantigens/immunology , Prevalence , Podocytes/metabolism , Podocytes/immunology , Podocytes/pathology , Autoantibodies/immunology , Autoantibodies/blood , Thrombospondins/immunology , Thrombospondins/metabolism
8.
J Clin Invest ; 134(15)2024 Jun 06.
Article in English | MEDLINE | ID: mdl-38842935

ABSTRACT

Proliferative glomerulonephritis is a severe condition that often leads to kidney failure. There is a significant lack of effective treatment for these disorders. Here, following the identification of a somatic PIK3CA gain-of-function mutation in podocytes of a patient, we demonstrate using multiple genetically engineered mouse models, single-cell RNA sequencing, and spatial transcriptomics the crucial role played by this pathway for proliferative glomerulonephritis development by promoting podocyte proliferation, dedifferentiation, and inflammation. Additionally, we show that alpelisib, a PI3Kα inhibitor, improves glomerular lesions and kidney function in different mouse models of proliferative glomerulonephritis and lupus nephritis by targeting podocytes. Surprisingly, we determined that pharmacological inhibition of PI3Kα affects B and T lymphocyte populations in lupus nephritis mouse models, with a decrease in the production of proinflammatory cytokines, autoantibodies, and glomerular complement deposition, which are all characteristic features of PI3Kδ inhibition, the primary PI3K isoform expressed in lymphocytes. Importantly, PI3Kα inhibition does not impact lymphocyte function under normal conditions. These findings were then confirmed in human lymphocytes isolated from patients with active lupus nephritis. In conclusion, we demonstrate the major role played by PI3Kα in proliferative glomerulonephritis and show that in this condition, alpelisib acts on both podocytes and the immune system.


Subject(s)
Class I Phosphatidylinositol 3-Kinases , Disease Models, Animal , Lupus Nephritis , Podocytes , Animals , Female , Humans , Mice , B-Lymphocytes/immunology , B-Lymphocytes/pathology , Class I Phosphatidylinositol 3-Kinases/genetics , Class I Phosphatidylinositol 3-Kinases/antagonists & inhibitors , Class I Phosphatidylinositol 3-Kinases/metabolism , Glomerulonephritis/pathology , Glomerulonephritis/immunology , Glomerulonephritis/genetics , Glomerulonephritis/enzymology , Glomerulonephritis/drug therapy , Lupus Nephritis/pathology , Lupus Nephritis/immunology , Lupus Nephritis/genetics , Lupus Nephritis/enzymology , Phosphoinositide-3 Kinase Inhibitors/pharmacology , Podocytes/pathology , Podocytes/immunology , Podocytes/metabolism , T-Lymphocytes/immunology , T-Lymphocytes/pathology , Thiazoles
9.
Cell Rep ; 43(5): 114249, 2024 May 28.
Article in English | MEDLINE | ID: mdl-38758648

ABSTRACT

Signal-regulatory protein alpha (SIRPα) has recently been found to be highly expressed in podocytes and is essential for maintaining podocyte function. However, its immunoregulatory function in podocytes remains elusive. Here, we report that SIRPα controls podocyte antigen presentation in specific T cell activation via inhibiting spleen tyrosine kinase (Syk) phosphorylation. First, podocyte SIRPα under lupus nephritis (LN) conditions is strongly downregulated. Second, podocyte-specific deletion of SIRPα exacerbates renal disease progression in lupus-prone mice, as evidenced by an increase in T cell infiltration. Third, SIRPα deletion or knockdown enhances podocyte antigen presentation, which activates specific T cells, via enhancing Syk phosphorylation. Supporting this, Syk inhibitor GS-9973 prevents podocyte antigen presentation, resulting in a decrease of T cell activation and mitigation of renal disease caused by SIRPα knockdown or deletion. Our findings reveal an immunoregulatory role of SIRPα loss in promoting podocyte antigen presentation to activate specific T cell immune responses in LN.


Subject(s)
Lupus Nephritis , Podocytes , Receptors, Immunologic , Syk Kinase , T-Lymphocytes , Animals , Female , Humans , Mice , Antigen Presentation/immunology , Inflammation/pathology , Inflammation/metabolism , Lupus Nephritis/pathology , Lupus Nephritis/immunology , Lupus Nephritis/metabolism , Lymphocyte Activation/immunology , Mice, Inbred C57BL , Phosphorylation , Podocytes/metabolism , Podocytes/pathology , Podocytes/immunology , Receptors, Immunologic/metabolism , Receptors, Immunologic/genetics , Syk Kinase/metabolism , T-Lymphocytes/immunology , T-Lymphocytes/metabolism
10.
Adv Sci (Weinh) ; 11(29): e2404151, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38785168

ABSTRACT

Primary membranous nephropathy (PMN) is one of the leading causes of end-stage renal disease, and the most frequent cause of massive proteinuria in nondiabetic adults, resulting in fatal complications. However, the underlying pathomechanisms of PMN remain largely unclear. Here, single-cell RNA sequencing is employed to analyze kidney biopsies from eleven PMN patients and seven healthy subjects. Profiling 44 060 cells from patients allowed us to characterize the cellular composition and cell-type-specific gene expression in the PMN kidney. The complement-induced BMP2/pSMAD1/COL4 pathway is identified as the pathogenic pathway in podocytes, bridging two key events, i.e., complement system activation and glomerular basement membrane thickening in PMN. Augmented infiltration and activation of myeloid leukocytes and B lymphocytes are found, profiling delicate crosstalk of immune cells in PMN kidneys. Overall, these results provide valuable insights into the roles of podocytes and immune cells in PMN, and comprehensive resources toward the complete understanding of PMN pathophysiology.


Subject(s)
Bone Morphogenetic Protein 2 , Glomerulonephritis, Membranous , Podocytes , Glomerulonephritis, Membranous/immunology , Glomerulonephritis, Membranous/pathology , Glomerulonephritis, Membranous/genetics , Glomerulonephritis, Membranous/metabolism , Humans , Podocytes/immunology , Podocytes/metabolism , Podocytes/pathology , Bone Morphogenetic Protein 2/metabolism , Bone Morphogenetic Protein 2/genetics , Male , Female , Adult , Middle Aged , Biopsy
11.
N Engl J Med ; 391(5): 422-433, 2024 Aug 01.
Article in English | MEDLINE | ID: mdl-38804512

ABSTRACT

BACKGROUND: Minimal change disease and primary focal segmental glomerulosclerosis in adults, along with idiopathic nephrotic syndrome in children, are immune-mediated podocytopathies that lead to nephrotic syndrome. Autoantibodies targeting nephrin have been found in patients with minimal change disease, but their clinical and pathophysiological roles are unclear. METHODS: We conducted a multicenter study to analyze antinephrin autoantibodies in adults with glomerular diseases, including minimal change disease, focal segmental glomerulosclerosis, membranous nephropathy, IgA nephropathy, antineutrophil cytoplasmic antibody-associated glomerulonephritis, and lupus nephritis, as well as in children with idiopathic nephrotic syndrome and in controls. We also created an experimental mouse model through active immunization with recombinant murine nephrin. RESULTS: The study included 539 patients (357 adults and 182 children) and 117 controls. Among the adults, antinephrin autoantibodies were found in 46 of the 105 patients (44%) with minimal change disease, 7 of 74 (9%) with primary focal segmental glomerulosclerosis, and only in rare cases among the patients with other conditions. Of the 182 children with idiopathic nephrotic syndrome, 94 (52%) had detectable antinephrin autoantibodies. In the subgroup of patients with active minimal change disease or idiopathic nephrotic syndrome who were not receiving immunosuppressive treatment, the prevalence of antinephrin autoantibodies was as high as 69% and 90%, respectively. At study inclusion and during follow-up, antinephrin autoantibody levels were correlated with disease activity. Experimental immunization induced a nephrotic syndrome, a minimal change disease-like phenotype, IgG localization to the podocyte slit diaphragm, nephrin phosphorylation, and severe cytoskeletal changes in mice. CONCLUSIONS: In this study, circulating antinephrin autoantibodies were common in patients with minimal change disease or idiopathic nephrotic syndrome and appeared to be markers of disease activity. Their binding at the slit diaphragm induced podocyte dysfunction and nephrotic syndrome, which highlights their pathophysiological significance. (Funded by Deutsche Forschungsgemeinschaft and others.).


Subject(s)
Autoantibodies , Membrane Proteins , Nephrotic Syndrome , Podocytes , Membrane Proteins/immunology , Autoantibodies/blood , Autoantibodies/immunology , Humans , Animals , Mice , Child , Podocytes/immunology , Adult , Nephrotic Syndrome/immunology , Male , Female , Middle Aged , Disease Models, Animal , Adolescent , Nephrosis, Lipoid/immunology , Child, Preschool , Glomerulosclerosis, Focal Segmental/immunology , Young Adult , Aged
12.
Kidney Int ; 105(1): 54-64, 2024 01.
Article in English | MEDLINE | ID: mdl-38707675

ABSTRACT

The neonatal Fc receptor (FcRn) was initially discovered as the receptor that allowed passive immunity in newborns by transporting maternal IgG through the placenta and enterocytes. Since its initial discovery, FcRn has been found to exist throughout all stages of life and in many different cell types. Beyond passive immunity, FcRn is necessary for intrinsic albumin and IgG recycling and is important for antigen processing and presentation. Given its multiple important roles, FcRn has been utilized in many disease treatments including a new class of agents that were developed to inhibit FcRn for treatment of a variety of autoimmune diseases. Certain cell populations within the kidney also express high levels of this receptor. Specifically, podocytes, proximal tubule epithelial cells, and vascular endothelial cells have been found to utilize FcRn. In this review, we summarize what is known about FcRn and its function within the kidney. We also discuss how FcRn has been used for therapeutic benefit, including how newer FcRn inhibiting agents are being used to treat autoimmune diseases. Lastly, we will discuss what renal diseases may respond to FcRn inhibitors and how further work studying FcRn within the kidney may lead to therapies for kidney diseases.


Subject(s)
Histocompatibility Antigens Class I , Kidney Diseases , Receptors, Fc , Humans , Histocompatibility Antigens Class I/metabolism , Histocompatibility Antigens Class I/immunology , Histocompatibility Antigens Class I/genetics , Receptors, Fc/metabolism , Receptors, Fc/immunology , Receptors, Fc/genetics , Kidney Diseases/metabolism , Kidney Diseases/drug therapy , Kidney Diseases/therapy , Kidney Diseases/immunology , Animals , Kidney/metabolism , Kidney/immunology , Kidney/pathology , Podocytes/metabolism , Podocytes/immunology , Immunoglobulin G/metabolism , Immunoglobulin G/immunology , Autoimmune Diseases/drug therapy , Autoimmune Diseases/immunology , Autoimmune Diseases/metabolism
13.
Kidney Int ; 105(6): 1165-1167, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38777401

ABSTRACT

The Oxford histopathologic classification (MEST-C: scores for lesions indicating active glomerular inflammation, mesangial [M] and endocapillary [E] hypercellularity as well as cellular or fibrocellular crescents [C], and for segmental glomerulosclerosis [S] and interstitial fibrosis and/or tubular atrophy [T]) is useful in helping assess prognosis in patients with IgA nephropathy. Elements of this classification indicative of active glomerular inflammation, endocapillary hypercellularity and crescents, also have been found to be responsive to immunosuppressive therapy, potentially including newer agents specifically targeting mediators of such inflammation. In this issue of Kidney International, Bellur and coworkers identify histopathologic subtypes of segmental glomerulosclerosis in IgA nephropathy showing podocyte injury that also behave like active lesions, including showing improved outcomes with immunosuppression. This podocyte injury, identifiable only by kidney biopsy, may represent a potential therapeutic target in some patients with IgA nephropathy.


Subject(s)
Glomerulonephritis, IGA , Podocytes , Glomerulonephritis, IGA/pathology , Glomerulonephritis, IGA/drug therapy , Glomerulonephritis, IGA/immunology , Humans , Podocytes/pathology , Podocytes/immunology , Podocytes/drug effects , Biopsy , Immunosuppressive Agents/therapeutic use , Glomerulosclerosis, Focal Segmental/pathology , Glomerulosclerosis, Focal Segmental/immunology , Glomerulosclerosis, Focal Segmental/drug therapy , Kidney Glomerulus/pathology , Kidney Glomerulus/immunology , Kidney Glomerulus/drug effects , Prognosis
14.
Kidney Int ; 105(6): 1279-1290, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38554992

ABSTRACT

Evidence from the Oxford IgA nephropathy (IgAN) cohort supports the clinical value of subclassifying focal segmental glomerulosclerosis lesions (S1). Using the larger Validation in IgA (VALIGA) study cohort, we investigated the association between podocytopathic changes and higher proteinuria, kidney outcome and response to immunosuppressive therapy. All biopsies were evaluated for glomeruli with segmental capillary occlusion by matrix ("not otherwise specified", NOS lesion), simple capsular adhesion without capillary occlusion (Adh), tip lesions, and podocyte hypertrophy (PH). S1 required a NOS lesion and/or Adh. A Chi-Squared Automatic Interaction Detection method was used to identify subgroups of FSGS lesions associated with distinctive proteinuria at biopsy. We assessed survival from a combined event (kidney failure or 50% decline in estimated glomerular filtration rate). Finally, we evaluated within each subgroup if immunosuppression was associated with a favorable outcome using propensity analysis. In 1147 patients, S1 was found in 70% of biopsies. Subclassification found NOS lesions in 44%, Adh in 59%, PH in 13%, and tip lesions in 3%, with much overlap. Four subgroups were identified with progressively higher proteinuria: from lowest, S1 without NOS, S1 with NOS but without Adh/PH, to highest, S1 with NOS and Adh but without PH, and S1 with NOS and PH. These four subgroups showed progressively worse kidney survival. Immunosuppression was associated with a better outcome only in the two highest proteinuria subgroups. Propensity analysis in these two groups, adjusted for clinical and pathological findings, found a significantly reduced time-dependent hazard of combined outcome with corticosteroids. Podocyte hypertrophy and glomeruli with simple adhesions appeared to reflect active lesions associated with a response to corticosteroids, while other S1 lesions defined chronicity. Thus, our findings support subclassifying S1 lesions in IgAN.


Subject(s)
Glomerular Filtration Rate , Glomerulonephritis, IGA , Glomerulosclerosis, Focal Segmental , Immunosuppressive Agents , Proteinuria , Humans , Glomerulonephritis, IGA/pathology , Glomerulonephritis, IGA/classification , Glomerulonephritis, IGA/diagnosis , Glomerulonephritis, IGA/immunology , Glomerulosclerosis, Focal Segmental/pathology , Glomerulosclerosis, Focal Segmental/classification , Glomerulosclerosis, Focal Segmental/immunology , Male , Female , Adult , Proteinuria/etiology , Proteinuria/pathology , Biopsy , Middle Aged , Immunosuppressive Agents/therapeutic use , Podocytes/pathology , Podocytes/immunology , Kidney Glomerulus/pathology , Kidney Glomerulus/immunology , Hypertrophy , Disease Progression , Treatment Outcome
15.
J Biochem Mol Toxicol ; 36(1): e22935, 2022 Jan.
Article in English | MEDLINE | ID: mdl-34726812

ABSTRACT

The primary consequences of membranous nephropathy (MN) are the development of nephrotic syndrome including hypogammaglobulinemia, the increased infectious risk, the loss of protein-bound vitamin D, and, above all, an elevated thromboembolic incidence of up to 50% in severe proteinuria patients. Membrane nephropathy may be either idiopathic or primary, not recognized (70%-80%) or secondary (20%-30%) to pathological sicknesses such as hepatitis B, systemic lupus erythematosus, malignancies, and side-effects of medicines. The immunological responses in MN involve multiple components: immunoglobulin G (IgG), long-escaped antigens, and the membrane attachment complex, formed by the supplement to form C5b-9. In general, IgG4 is the most significant IgG subclass deposited in idiopathic membranous nephropathic disease but fluctuating IgG1 levels also are linked with immunological deposits. In contrast, IgG1, IgG2, and IgG3 deposition are greater than IgG4 deposition in secondary nephropathy. Fluconazole is a synthetic antifungal triazole that is often used. It is well tolerated in general and has never been identified as a cause of nephropathies. We report on the development of MN caused by fluconazole therapy that could potentiate podocyte autophagy.


Subject(s)
Autophagy , Fluconazole/adverse effects , Glomerulonephritis, Membranous/chemically induced , Glomerulonephritis, Membranous/immunology , Podocytes/immunology , Autophagy/drug effects , Autophagy/immunology , Fluconazole/therapeutic use , Glomerulonephritis, Membranous/therapy , Humans , Immunoglobulin G/immunology
16.
Clin Immunol ; 232: 108869, 2021 11.
Article in English | MEDLINE | ID: mdl-34600127

ABSTRACT

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/immunology
17.
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
18.
Front Immunol ; 12: 704526, 2021.
Article in English | MEDLINE | ID: mdl-34497607

ABSTRACT

Objective: Baricitinib, a selective inhibitor for janus kinase (JAK) 1 and JAK2, is approved for use in rheumatoid arthritis. Systemic lupus erythematosus (SLE) is recently regarded as a potential candidate targeted by JAK inhibitors because of the relationship between its pathogenesis and JAK/signal transducer and activator of transcription (STAT) pathway-mediated cytokines such as type I interferons. The objective of this study was to determine whether baricitinib could effectively ameliorate SLE using a murine model. Methods: To investigate effects of baricitinib on various autoimmune features, especially renal involvements in SLE, eight-week-old MRL/Mp-Faslpr (MRL/lpr) mice were used as a lupus-prone animal model and treated with baricitinib for eight weeks. Immortalized podocytes and primary podocytes and B cells isolated from C57BL/6 mice were used to determine the in vitro efficacy of baricitinib. Results: Baricitinib remarkably suppressed lupus-like phenotypes of MRL/lpr mice, such as splenomegaly, lymphadenopathy, proteinuria, and systemic autoimmunity including circulating autoantibodies and pro-inflammatory cytokines. It also modulated immune cell populations and effectively ameliorated renal inflammation, leading to the recovery of the expression of structural proteins in podocytes. According to in vitro experiments, baricitinib treatment could mitigate B cell differentiation and restore disrupted cytoskeletal structures of podocytes under inflammatory stimulation by blocking the JAK/STAT pathway. Conclusions: The present study demonstrated that baricitinib could effectively attenuate autoimmune features including renal inflammation of lupus-prone mice by suppressing aberrant B cell activation and podocyte abnormalities. Thus, baricitinib as a selective JAK inhibitor could be a promising therapeutic candidate in the treatment of SLE.


Subject(s)
Azetidines/pharmacology , Lupus Erythematosus, Systemic , Podocytes , Purines/pharmacology , Pyrazoles/pharmacology , Sulfonamides/pharmacology , Animals , Disease Models, Animal , Female , Janus Kinase 1/antagonists & inhibitors , Janus Kinase 1/immunology , Janus Kinase 2/antagonists & inhibitors , Janus Kinase 2/immunology , Lupus Erythematosus, Systemic/drug therapy , Lupus Erythematosus, Systemic/immunology , Lupus Erythematosus, Systemic/pathology , Mice , Mice, Inbred MRL lpr , Podocytes/immunology , Podocytes/pathology , Signal Transduction/drug effects
19.
J Immunol Res ; 2021: 8163298, 2021.
Article in English | MEDLINE | ID: mdl-34337081

ABSTRACT

Membranous nephropathy (MN) is an organ-restricted autoimmune disease mainly caused by circulating autoantibodies against podocyte antigens, including the M-type phospholipase A2 receptor (PLA2R) and thrombospondin domain-containing 7A (THSD7A). Antibodies against PLA2R are present in 70%-80% and against THSD7A in 2% of adult patients, which provides a paradigm shift in molecular diagnosis and management monitoring. Both antigens share some similar characteristics: they are expressed by podocytes and have wide tissue distributions; they are bound by autoantibodies only under nonreducing conditions, and the subtype of most autoantibodies is IgG4. However, the factors triggering autoantibody production as well as the association among air pollution, malignancy, and the pathogenesis of MN remain unclear. In this review, we discuss the similarity between the pathological mechanisms triggered by disparate antigens and their associated diseases. Furthermore, we demonstrated the possibility that PM2.5, malignancy, and gene expression specifically induce exposure of these antigens through conformational changes, molecular mimicry, or increased expression eliciting autoimmune responses. Thus, this review provides novel insights into the pathological mechanism of MN.


Subject(s)
Autoantibodies/immunology , Autoantigens/immunology , Glomerulonephritis, Membranous/immunology , Receptors, Phospholipase A2/immunology , Thrombospondins/immunology , Animals , Antigens, Surface/immunology , Antigens, Surface/metabolism , Autoantibodies/blood , Autoantigens/metabolism , Disease Models, Animal , Glomerulonephritis, Membranous/blood , Glomerulonephritis, Membranous/pathology , Humans , Membrane Proteins/immunology , Membrane Proteins/metabolism , Mice , Podocytes/immunology , Podocytes/metabolism , Podocytes/pathology , Receptors, Phospholipase A2/metabolism , Thrombospondins/metabolism
20.
Monoclon Antib Immunodiagn Immunother ; 40(3): 134-140, 2021 Jun.
Article in English | MEDLINE | ID: mdl-34161160

ABSTRACT

Podoplanin (PDPN) plays an important role in the development of many normal tissues and is expressed in various cancers. We have previously developed multiple monoclonal antibodies (mAbs) against PDPNs from a variety of animal species and characterized each of these PDPNs using the anti-PDPN mAbs. In this study, we evaluated whether these anti-PDPN mAbs possess cross-reactivity with ferret PDPN (ferPDPN) using flow cytometry. Comprehensive analysis using 17 differing anti-PDPN mAbs available for immunohistochemistry use, demonstrated that the anti-bear PDPN mAb (clone PMab-241) strongly cross-reacts with ferPDPN-overexpressed Chinese hamster ovary-K1 (CHO/ferPDPN) cells. Immunohistochemistry analysis demonstrated intense PMab-241 staining within Bowman's capsules and glomeruli of the ferret kidney, and lymphatic endothelial cells of the ferret lung. These results demonstrate that PMab-241 is suitable for the detection of PDPN in ferret tissues.


Subject(s)
Antibodies, Monoclonal/immunology , Antibody Specificity/immunology , Ferrets/immunology , Membrane Glycoproteins/immunology , Animals , Antibodies, Monoclonal/pharmacology , CHO Cells , Cricetulus , Epitope Mapping , Flow Cytometry , Humans , Membrane Glycoproteins/antagonists & inhibitors , Mice , Podocytes/immunology , Rats
SELECTION OF CITATIONS
SEARCH DETAIL