Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 1.955
Filtrar
1.
Adv Kidney Dis Health ; 31(4): 358-373, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-39084761

RESUMEN

Paraproteinemias are a group of complex diseases associated with an overproduction of a monoclonal immunoglobulin that can cause a diversity of kidney disorders and end-organ damage. In this review, we focus on paraprotein-mediated glomerular diseases. Kidney biopsy plays a crucial role in diagnosing these disorders, enabling the identification of specific histological patterns. These lesions are categorized into organized (such as amyloidosis, immunotactoid glomerulopathy, fibrillary glomerulonephritis, cryoglobulinemic glomerulonephritis, and monoclonal crystalline glomerulopathies) and nonorganized deposits (such as monoclonal Ig deposition disease and proliferative glomerulonephritis with monoclonal Ig deposits) based on the characteristics of immunofluorescence findings and the ultrastructural appearance of deposits on electron microscopy. This review aims to provide an update, highlight, and discuss clinicopathological aspects such as definition, epidemiology, clinical manifestations, mechanisms of kidney injury, histological features, and diagnostic procedures.


Asunto(s)
Glomerulonefritis , Glomérulos Renales , Paraproteinemias , Humanos , Paraproteinemias/patología , Paraproteinemias/diagnóstico , Paraproteinemias/metabolismo , Glomerulonefritis/patología , Glomerulonefritis/metabolismo , Glomerulonefritis/diagnóstico , Glomérulos Renales/patología , Glomérulos Renales/metabolismo , Paraproteínas/metabolismo , Enfermedades Renales/patología , Enfermedades Renales/metabolismo , Biopsia , Amiloidosis/patología , Amiloidosis/metabolismo , Amiloidosis/diagnóstico
2.
Ren Fail ; 46(2): 2371059, 2024 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-38946402

RESUMEN

BACKGROUND: Circular RNAs (circRNAs) have been shown to play critical roles in the initiation and progression of chronic glomerulonephritis (CGN), while their role from mesangial cells in contributing to the pathogenesis of CGN is rarely understood. Our study aims to explore the potential functions of mesangial cell-derived circRNAs using RNA sequencing (RNA-seq) and bioinformatics analysis. METHODS: Mouse mesangial cells (MMCs) were stimulated by lipopolysaccharide (LPS) to establish an in vitro model of CGN. Pro-inflammatory cytokines and cell cycle stages were detected by Enzyme-linked immunosorbent assay (ELISA) and Flow Cytometry experiment, respectively. Subsequently, differentially expressed circRNAs (DE-circRNAs) were identified by RNA-seq. GEO microarrays were used to identify differentially expressed mRNAs (DE-mRNAs) between CGN and healthy populations. Weighted co-expression network analysis (WGCNA) was utilized to explore clinically significant modules of CGN. CircRNA-associated CeRNA networks were constructed by bioinformatics analysis. The hub mRNAs from CeRNA network were identified using LASSO algorithms. Furthermore, utilizing protein-protein interaction (PPI), gene ontology (GO), pathway enrichment (KEGG), and GSEA analyses to explore the potential biological function of target genes from CeRNA network. In addition, we investigated the relationships between immune cells and hub mRNAs from CeRNA network using CIBERSORT. RESULTS: The expression of pro-inflammatory cytokines IL-1ß, IL-6, and TNF-α was drastically increased in LPS-induced MMCs. The number of cells decreased significantly in the G1 phase but increased significantly in the S/G2 phase. A total of 6 DE-mRNAs were determined by RNA-seq, including 4 up-regulated circRNAs and 2 down-regulated circRNAs. WGCNA analysis identified 1747 DE-mRNAs of the turquoise module from CGN people in the GEO database. Then, the CeRNA networks, including 6 circRNAs, 38 miRNAs, and 80 mRNAs, were successfully constructed. The results of GO and KEGG analyses revealed that the target mRNAs were mainly enriched in immune, infection, and inflammation-related pathways. Furthermore, three hub mRNAs (BOC, MLST8, and HMGCS2) from the CeRNA network were screened using LASSO algorithms. GSEA analysis revealed that hub mRNAs were implicated in a great deal of immune system responses and inflammatory pathways, including IL-5 production, MAPK signaling pathway, and JAK-STAT signaling pathway. Moreover, according to an evaluation of immune infiltration, hub mRNAs have statistical correlations with neutrophils, plasma cells, monocytes, and follicular helper T cells. CONCLUSIONS: Our findings provide fundamental and novel insights for further investigations into the role of mesangial cell-derived circRNAs in CGN pathogenesis.


Asunto(s)
Biología Computacional , Glomerulonefritis , Células Mesangiales , ARN Circular , ARN Circular/genética , ARN Circular/metabolismo , Animales , Ratones , Células Mesangiales/metabolismo , Glomerulonefritis/genética , Glomerulonefritis/metabolismo , Análisis de Secuencia de ARN , Redes Reguladoras de Genes , ARN Mensajero/metabolismo , ARN Mensajero/genética , Mapas de Interacción de Proteínas/genética , Enfermedad Crónica , Citocinas/metabolismo , Lipopolisacáridos/farmacología , Perfilación de la Expresión Génica , Modelos Animales de Enfermedad
3.
J Physiol Pharmacol ; 75(3)2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-39042393

RESUMEN

Circular E3 ubiquitin-protein ligase (circ-ITCH), a novel circRNA, is generated from several exons of itchy E3 ubiquitin protein ligase. Reports on circ-ITCH have discussed its pathogenic performance in human diseases. Based on this, this study determines whether and how circ-ITCH is involved in the pathogenesis of chronic glomerulonephritis (CGN). First, a rat model of CGN induced by cationic bovine serum albumin was established. Then, CGN rats were injected with lentiviruses interfering with the expression of circ-ITCH, miR-146a-5p or tyrosine 3-monooxygenase/tryptophan 5-monooxygenase activation protein gamma (YWHAG). Then, blood urea nitrogen and serum creatinine levels were measured to evaluate renal function; inflammatory factor content and fibrosis marker expression in kidney tissue were detected; renal pathological damage was analyzed by hematoxylin-eosin staining and periodic acid-Schiff staining. Finally, the binding relationship between miR-146a-5p and circ-ITCH or YWHAG was verified. Elevating circ-ITCH or depleting miR-146a-5p improved renal function (both P<0.05), reduced inflammatory factor content and fibrosis marker expression (all P<0.05) and alleviated renal pathological damage in CGN rats. Circ-ITCH negatively regulated miR-146a-5p expression by adsorbing miR-146a-5p (P<0.05), and miR-146a-5p inhibited YWHAG expression by binding to the 3'-UTR of YWHAG (P<0.05). Loss of YWHAG reversed the protective effect of upregulated circ-ITCH in CGN rats (all P<0.05). We conclude that circ-ITCH improves renal function and attenuates inflammation and renal injury in rats with CGN via the miR-146a-5p/YWHAG axis.


Asunto(s)
Glomerulonefritis , Inflamación , Riñón , MicroARNs , Ratas Sprague-Dawley , Ubiquitina-Proteína Ligasas , Animales , MicroARNs/genética , MicroARNs/metabolismo , Ratas , Masculino , Riñón/patología , Riñón/metabolismo , Ubiquitina-Proteína Ligasas/genética , Ubiquitina-Proteína Ligasas/metabolismo , Glomerulonefritis/metabolismo , Glomerulonefritis/genética , Inflamación/metabolismo , Inflamación/genética , ARN Circular/genética , ARN Circular/metabolismo , Enfermedad Crónica
4.
Physiol Rep ; 12(13): e16129, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38955668

RESUMEN

Cardiotrophin-1 (CT-1), a member of the interleukin (IL)-6 cytokine family, has renoprotective effects in mouse models of acute kidney disease and tubulointerstitial fibrosis, but its role in glomerular disease is unknown. To address this, we used the mouse model of nephrotoxic nephritis to test the hypothesis that CT-1 also has a protective role in immune-mediated glomerular disease. Using immunohistochemistry and analysis of single-cell RNA-sequencing data of isolated glomeruli, we demonstrate that CT-1 is expressed in the glomerulus in male mice, predominantly in parietal epithelial cells and is downregulated in mice with nephrotoxic nephritis. Furthermore, analysis of data from patients revealed that human glomerular disease is also associated with reduced glomerular CT-1 transcript levels. In male mice with nephrotoxic nephritis and established proteinuria, administration of CT-1 resulted in reduced albuminuria, prevented podocyte loss, and sustained plasma creatinine, compared with mice administered saline. CT-1 treatment also reduced fibrosis in the kidney cortex, peri-glomerular macrophage accumulation and the kidney levels of the pro-inflammatory mediator complement component 5a. In conclusion, CT-1 intervention therapy delays the progression of glomerular disease in mice by preserving kidney function and inhibiting renal inflammation and fibrosis.


Asunto(s)
Citocinas , Glomérulos Renales , Ratones Endogámicos C57BL , Animales , Masculino , Citocinas/metabolismo , Citocinas/genética , Ratones , Glomérulos Renales/metabolismo , Glomérulos Renales/patología , Modelos Animales de Enfermedad , Humanos , Fibrosis , Glomerulonefritis/metabolismo , Glomerulonefritis/patología , Glomerulonefritis/tratamiento farmacológico
5.
Adv Kidney Dis Health ; 31(3): 223-233, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-39004462

RESUMEN

C3 glomerulopathy (C3G) is a rare disorder marked by deposition of C3 in the glomerulus, resulting in damage to the glomerular filtration unit and presenting with features of the nephritic and nephrotic syndromes. Fundamentally, C3G is caused by dysregulation of the alternative pathway of the complement cascade, either due to genetic variants or acquired humoral factors. Despite significant advances in recent years in the understanding of the underlying mechanisms and culprit lesions that result in the development of C3G, treatment options remain severely limited, and the prognosis is often poor. Fortunately, a number of anticomplement therapies are emerging from the drug development pipeline, with several in late-stage testing in patients with C3G, and there is hope that we will soon have more targeted options for managing patients with this devastating disease. In this review, we provide an overview of C3G, as well as summarizing the evidence for current treatments and detailing the clinical trials that are currently underway.


Asunto(s)
Complemento C3 , Humanos , Complemento C3/metabolismo , Complemento C3/genética , Complemento C3/inmunología , Glomérulos Renales/patología , Glomérulos Renales/metabolismo , Vía Alternativa del Complemento/genética , Vía Alternativa del Complemento/efectos de los fármacos , Glomerulonefritis/patología , Glomerulonefritis/metabolismo , Glomerulonefritis/genética , Glomerulonefritis/inmunología , Glomerulonefritis/terapia , Glomerulonefritis/diagnóstico , Síndrome Nefrótico/patología , Síndrome Nefrótico/genética , Síndrome Nefrótico/metabolismo , Síndrome Nefrótico/diagnóstico , Síndrome Nefrótico/terapia
6.
J Exp Med ; 221(9)2024 09 02.
Artículo en Inglés | MEDLINE | ID: mdl-39058386

RESUMEN

Autoantibody-mediated glomerulonephritis (AGN) arises from dysregulated renal inflammation, with urgent need for improved treatments. IL-17 is implicated in AGN and drives pathology in a kidney-intrinsic manner via renal tubular epithelial cells (RTECs). Nonetheless, downstream signaling mechanisms provoking kidney pathology are poorly understood. A noncanonical RNA binding protein (RBP), Arid5a, was upregulated in human and mouse AGN. Arid5a-/- mice were refractory to AGN, with attenuated myeloid infiltration and impaired expression of IL-17-dependent cytokines and transcription factors (C/EBPß, C/EBPδ). Transcriptome-wide RIP-Seq revealed that Arid5a inducibly interacts with conventional IL-17 target mRNAs, including CEBPB and CEBPD. Unexpectedly, many Arid5a RNA targets corresponded to translational regulation and RNA processing pathways, including rRNAs. Indeed, global protein synthesis was repressed in Arid5a-deficient cells, and C/EBPs were controlled at the level of protein rather than RNA accumulation. IL-17 prompted Arid5a nuclear export and association with 18S rRNA, a 40S ribosome constituent. Accordingly, IL-17-dependent renal autoimmunity is driven by Arid5a at the level of ribosome interactions and translation.


Asunto(s)
Autoanticuerpos , Proteínas de Unión al ADN , Glomerulonefritis , Interleucina-17 , Ratones Noqueados , Factores de Transcripción , Animales , Interleucina-17/metabolismo , Glomerulonefritis/inmunología , Glomerulonefritis/genética , Glomerulonefritis/metabolismo , Glomerulonefritis/patología , Humanos , Ratones , Factores de Transcripción/metabolismo , Factores de Transcripción/genética , Autoanticuerpos/inmunología , Proteínas de Unión al ADN/metabolismo , Proteínas de Unión al ADN/genética , Proteína beta Potenciadora de Unión a CCAAT/metabolismo , Proteína beta Potenciadora de Unión a CCAAT/genética , Proteína delta de Unión al Potenciador CCAAT/metabolismo , Proteína delta de Unión al Potenciador CCAAT/genética , Ratones Endogámicos C57BL , Proteínas de Unión al ARN/metabolismo , Proteínas de Unión al ARN/genética , Proteínas de Unión al ARN/inmunología , ARN Mensajero/genética , ARN Mensajero/metabolismo
7.
BMC Nephrol ; 25(1): 211, 2024 Jun 27.
Artículo en Inglés | MEDLINE | ID: mdl-38937701

RESUMEN

OBJECTIVE: To investigate the expression and significance of Fractalkine (CX3CL1, FKN) in serum and renal tissue of myeloperoxidase and anti-neutrophil cytoplasmic antibody associated vasculitis (MPO-AAV) rats. METHODS: Thirty Wistar-Kyoto (WKY) rats were randomly divided into: Control group, MPO-AAV group (400 µg/kg MPO mixed with Freund's complete adjuvant i.p), MPO-AAV + Anti-FKN group (400 µg/kg MPO mixed with Freund's complete adjuvant i.p), anti-FKN group (1 µg/ rat /day, i.p) after 6 weeks. MPO-AAV associated glomerulonephritis model was established by intraperitoneal injection of MPO + Freund's complete adjuvant with 10 mice in each group. The concentration of MPO-ANCA and FKN in serum was detected by Enzyme-linked immunosorbent assay (ELISA). Hematoxylin-eosin (HE) staining was used to detect pathological changes of kidney tissue. Western blot and immunofluorescence staining were used to detect the expression and localization of FKN protein in kidney tissue. Renal function test indicators: 24-hour urinary protein (UAER), blood urea nitrogen (BUN), serum creatinine (Scr). The expression levels of p65NF-κB and IL-6 was detected by Immunohistochemical assays. RESULTS: Compared with the control group, the serum MPO-ANCA antibody expression level in the MPO-AAV group was significantly increased (P < 0.01), and the contents of UAER, BUN and Scr were significantly up-regulated at 24 h (P < 0.01). Compared with the control group, the glomeruli in the MPO-AAV group had different degrees of damage, infiltration of inflammatory cell, and membrane cell hyperplasia and renal tubule edema. Compared with the control group, rats in the MPO-AAV group had significantly higher levels of FKN in serum and renal tissues (P < 0.01), and high expression of p65NF-κB and IL-6 in renal tissues (P < 0.01) (P < 0.05), whereas anti-FKN reversed the expression of the above factors. In MPO-AAV renal tissue, FKN was mainly expressed in the cytoplasm of renal tubular epithelial cells and glomerular podocytes. In addition, the contents of 24 h UAER, BUN and Scr of renal function in MPO-AAV rats were significantly decreased (P < 0.01) and the damage of renal tissue was significantly ameliorated after the administration of antagonistic FKN. CONCLUSION: FKN may play a key role in the pathogenesis of MPO-AAV associated glomerulonephritis.


Asunto(s)
Quimiocina CX3CL1 , Glomerulonefritis , Peroxidasa , Ratas Endogámicas WKY , Animales , Quimiocina CX3CL1/metabolismo , Glomerulonefritis/metabolismo , Glomerulonefritis/patología , Ratas , Peroxidasa/metabolismo , Masculino , Vasculitis Asociada a Anticuerpos Citoplasmáticos Antineutrófilos/metabolismo , Riñón/patología , Riñón/metabolismo , Anticuerpos Anticitoplasma de Neutrófilos , Factor de Transcripción ReIA/metabolismo
8.
Int J Clin Pharmacol Ther ; 62(9): 395-401, 2024 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-38920082

RESUMEN

OBJECTIVES: To investigate the expression of P-glycoprotein in T-cell subpopulations of lymphocytes from adult patients with refractory glomerulonephritis (GN). MATERIALS AND METHODS: Flow cytometry was used to analyze the T-cell subpopulations of lymphocytes from adult patients with refractory GN and healthy individuals. The CD243 antibody marked the membrane P-glycoprotein of immune cells. RESULTS: The mean ± standard deviation (SD) values of percentages of CD3+, CD3+CD4+, CD3+CD8+ cells in lymphocytes from patients with refractory GN were 63.94 ± 26.98, 55.16 ± 4.78, and 37.79 ± 6.01%, respectively. These values in healthy individuals were 74.88 ± 3.75, 56.60 ± 9.22, and 34.20 ± 5.21%, respectively. No significant differences were observed between the patients with refractory GN and healthy individuals. The mean ± SD values of percentages of CD3+CD4+CD243+ and CD3+CD8+CD243+ cells in the lymphocytes of patients with refractory GN were 0.14 ± 0.11 and 0.11 ± 0.07%, respectively. These values in healthy individuals were 0.05 ± 0.02 and 0.04 ± 0.02%, respectively. The difference in CD3+CD8+CD243+ percentage between patients with refractory GN and healthy individuals was significant (p = 0.0216). CONCLUSION: These findings suggest that P-glycoprotein expression on CD3+CD8+ T cells is a promising marker and a suitable target of drug resistance in patients with refractory GN.


Asunto(s)
Glomerulonefritis , Humanos , Masculino , Femenino , Adulto , Persona de Mediana Edad , Glomerulonefritis/inmunología , Glomerulonefritis/metabolismo , Citometría de Flujo , Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/metabolismo , Subfamilia B de Transportador de Casetes de Unión a ATP/metabolismo , Subfamilia B de Transportador de Casetes de Unión a ATP/genética , Estudios de Casos y Controles , Adulto Joven , Subgrupos de Linfocitos T/inmunología , Subgrupos de Linfocitos T/metabolismo
9.
Int J Mol Sci ; 25(10)2024 May 08.
Artículo en Inglés | MEDLINE | ID: mdl-38791159

RESUMEN

Glomerulonephritis (GN) is characterized by podocyte injury or glomerular filtration dysfunction, which results in proteinuria and eventual loss of kidney function. Progress in studying the mechanism of GN, and developing an effective therapy, has been limited by the absence of suitable in vitro models that can closely recapitulate human physiological responses. We developed a microfluidic glomerulus-on-a-chip device that can recapitulate the physiological environment to construct a functional filtration barrier, with which we investigated biological changes in podocytes and dynamic alterations in the permeability of the glomerular filtration barrier (GFB) on a chip. We also evaluated the potential of GN-mimicking devices as a model for predicting responses to human GN. Glomerular endothelial cells and podocytes successfully formed intact monolayers on opposite sides of the membrane in our chip device. Permselectivity analysis confirmed that the chip was constituted by a functional GFB that could accurately perform differential clearance of albumin and dextran. Reduction in cell viability resulting from damage was observed in all serum-induced GN models. The expression of podocyte-specific marker WT1 was also decreased. Albumin permeability was increased in most models of serum-induced IgA nephropathy (IgAN) and membranous nephropathy (MN). However, sera from patients with minimal change disease (MCD) or lupus nephritis (LN) did not induce a loss of permeability. This glomerulus-on-a-chip system may provide a platform of glomerular cell culture for in vitro GFB in formation of a functional three-dimensional glomerular structure. Establishing a disease model of GN on a chip could accelerate our understanding of pathophysiological mechanisms of glomerulopathy.


Asunto(s)
Glomerulonefritis , Glomérulos Renales , Dispositivos Laboratorio en un Chip , Podocitos , Humanos , Podocitos/metabolismo , Podocitos/patología , Glomérulos Renales/metabolismo , Glomérulos Renales/patología , Glomerulonefritis/metabolismo , Glomerulonefritis/fisiopatología , Glomerulonefritis/patología , Barrera de Filtración Glomerular/metabolismo , Glomerulonefritis Membranosa/metabolismo , Glomerulonefritis Membranosa/patología , Glomerulonefritis Membranosa/fisiopatología , Glomerulonefritis por IGA/metabolismo , Glomerulonefritis por IGA/patología , Glomerulonefritis por IGA/fisiopatología , Permeabilidad , Células Endoteliales/metabolismo , Células Endoteliales/patología , Nefritis Lúpica/metabolismo , Nefritis Lúpica/patología , Nefritis Lúpica/fisiopatología , Supervivencia Celular , Nefrosis Lipoidea/metabolismo , Nefrosis Lipoidea/patología , Nefrosis Lipoidea/fisiopatología
10.
Kidney Int ; 106(3): 450-469, 2024 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-38821447

RESUMEN

Unlike classical protein kinase A, with separate catalytic and regulatory subunits, EPACs are single chain multi-domain proteins containing both catalytic and regulatory elements. The importance of cAMP-Epac-signaling as an energy provider has emerged over the last years. However, little is known about Epac1 signaling in chronic kidney disease. Here, we examined the role of Epac1 during the progression of glomerulonephritis (GN). We first observed that total genetic deletion of Epac1 in mice accelerated the progression of nephrotoxic serum (NTS)-induced GN. Next, mice with podocyte-specific conditional deletion of Epac1 were generated and showed that NTS-induced GN was exacerbated in these mice. Gene expression analysis in glomeruli at the early and late phases of GN showed that deletion of Epac1 in podocytes was associated with major alterations in mitochondrial and metabolic processes and significant dysregulation of the glycolysis pathway. In vitro, Epac1 activation in a human podocyte cell line increased mitochondrial function to cope with the extra energy demand under conditions of stress. Furthermore, Epac1-induced glycolysis and lactate production improved podocyte viability. To verify the in vivo therapeutic potential of Epac1 activation, the Epac1 selective cAMP mimetic 8-pCPT was administered in wild type mice after induction of GN. 8-pCPT alleviated the progression of GN by improving kidney function with decreased structural injury with decreased crescent formation and kidney inflammation. Importantly, 8-pCPT had no beneficial effect in mice with Epac1 deletion in podocytes. Thus, our data suggest that Epac1 activation is an essential protective mechanism in GN by reprogramming podocyte metabolism. Hence, targeting Epac1 activation could represent a potential therapeutic approach.


Asunto(s)
AMP Cíclico , Glomerulonefritis , Factores de Intercambio de Guanina Nucleótido , Reprogramación Metabólica , Podocitos , Animales , Humanos , Masculino , Ratones , Línea Celular , AMP Cíclico/metabolismo , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Metabolismo Energético/efectos de los fármacos , Glomerulonefritis/patología , Glomerulonefritis/metabolismo , Glomerulonefritis/genética , Glomerulonefritis/prevención & control , Glucólisis , Factores de Intercambio de Guanina Nucleótido/metabolismo , Factores de Intercambio de Guanina Nucleótido/genética , Ratones Endogámicos C57BL , Ratones Noqueados , Mitocondrias/metabolismo , Mitocondrias/patología , Podocitos/metabolismo , Podocitos/patología , Transducción de Señal
11.
Am J Physiol Renal Physiol ; 326(6): F988-F1003, 2024 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-38634138

RESUMEN

Acid sphingomyelinase (ASM) has been reported to increase tissue ceramide and thereby mediate hyperhomocysteinemia (hHcy)-induced glomerular nucleotide-binding oligomerization domain-like receptor containing pyrin domain 3 (NLRP3) inflammasome activation, inflammation, and sclerosis. In the present study, we tested whether somatic podocyte-specific silencing of Smpd1 gene (mouse ASM gene code) attenuates hHcy-induced NLRP3 inflammasome activation and associated extracellular vesicle (EV) release in podocytes and thereby suppresses glomerular inflammatory response and injury. In vivo, somatic podocyte-specific Smpd1 gene silencing almost blocked hHcy-induced glomerular NLRP3 inflammasome activation in Podocre (podocyte-specific expression of cre recombinase) mice compared with control littermates. By nanoparticle tracking analysis (NTA), floxed Smpd1 shRNA transfection was found to abrogate hHcy-induced elevation of urinary EV excretion in Podocre mice. In addition, Smpd1 gene silencing in podocytes prevented hHcy-induced immune cell infiltration into glomeruli, proteinuria, and glomerular sclerosis in Podocre mice. Such protective effects of podocyte-specific Smpd1 gene silencing were mimicked by global knockout of Smpd1 gene in Smpd1-/- mice. On the contrary, podocyte-specific Smpd1 gene overexpression exaggerated hHcy-induced glomerular pathological changes in Smpd1trg/Podocre (podocyte-specific Smpd1 gene overexpression) mice, which were significantly attenuated by transfection of floxed Smpd1 shRNA. In cell studies, we also confirmed that Smpd1 gene knockout or silencing prevented homocysteine (Hcy)-induced elevation of EV release in the primary cultures of podocyte isolated from Smpd1-/- mice or podocytes of Podocre mice transfected with floxed Smpd1 shRNA compared with WT/WT podocytes. Smpd1 gene overexpression amplified Hcy-induced EV secretion from podocytes of Smpd1trg/Podocre mice, which was remarkably attenuated by transfection of floxed Smpd1 shRNA. Mechanistically, Hcy-induced elevation of EV release from podocytes was blocked by ASM inhibitor (amitriptyline, AMI), but not by NLRP3 inflammasome inhibitors (MCC950 and glycyrrhizin, GLY). Super-resolution microscopy also showed that ASM inhibitor, but not NLRP3 inflammasome inhibitors, prevented the inhibition of lysosome-multivesicular body interaction by Hcy in podocytes. Moreover, we found that podocyte-derived inflammatory EVs (released from podocytes treated with Hcy) induced podocyte injury, which was exaggerated by T cell coculture. Interstitial infusion of inflammatory EVs into renal cortex induced glomerular injury and immune cell infiltration. In conclusion, our findings suggest that ASM in podocytes plays a crucial role in the control of NLRP3 inflammasome activation and inflammatory EV release during hHcy and that the development of podocyte-specific ASM inhibition or Smpd1 gene silencing may be a novel therapeutic strategy for treatment of hHcy-induced glomerular disease with minimized side effect.NEW & NOTEWORTHY In the present study, we tested whether podocyte-specific silencing of Smpd1 gene attenuates hyperhomocysteinemia (hHcy)-induced nucleotide-binding oligomerization domain-like receptor containing pyrin domain 3 (NLRP3) inflammasome activation and associated inflammatory extracellular vesicle (EV) release in podocytes and thereby suppresses glomerular inflammatory response and injury. Our findings suggest that acid sphingomyelinase (ASM) in podocytes plays a crucial role in the control of NLRP3 inflammasome activation and inflammatory EV release during hHcy. Based on our findings, it is anticipated that the development of podocyte-specific ASM inhibition or Smpd1 gene silencing may be a novel therapeutic strategy for treatment of hHcy-induced glomerular disease with minimized side effects.


Asunto(s)
Hiperhomocisteinemia , Inflamasomas , Ratones Noqueados , Proteína con Dominio Pirina 3 de la Familia NLR , Podocitos , Esfingomielina Fosfodiesterasa , Animales , Esfingomielina Fosfodiesterasa/genética , Esfingomielina Fosfodiesterasa/metabolismo , Podocitos/metabolismo , Podocitos/patología , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Proteína con Dominio Pirina 3 de la Familia NLR/genética , Hiperhomocisteinemia/metabolismo , Hiperhomocisteinemia/complicaciones , Hiperhomocisteinemia/genética , Inflamasomas/metabolismo , Inflamasomas/genética , Glomérulos Renales/patología , Glomérulos Renales/metabolismo , Glomerulonefritis/patología , Glomerulonefritis/metabolismo , Glomerulonefritis/genética , Silenciador del Gen , Ratones , Ratones Endogámicos C57BL , Vesículas Extracelulares/metabolismo , Masculino , Modelos Animales de Enfermedad
12.
J Nephrol ; 37(5): 1209-1225, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38689160

RESUMEN

BACKGROUND: Glomerulonephritis inherently leads to the development of chronic kidney disease. It is the second most common diagnosis in patients requiring renal replacement therapy in the United Kingdom. Metabolomics and proteomics can characterise, identify and quantify an individual's protein and metabolite make-up. These techniques have been optimised and can be performed on samples including kidney tissue, blood and urine. Utilising omic techniques in nephrology can uncover disease pathophysiology and transform the diagnostics and treatment options for glomerulonephritis. OBJECTIVES: To evaluate the utility of metabolomics and proteomics using mass spectrometry and nuclear magnetic resonance in glomerulonephritis. METHODS: The systematic review was registered on PROSPERO (CRD42023442092). Standard and extensive Cochrane search methods were used. The latest search date was March 2023. Participants were of any age with a histological diagnosis of glomerulonephritis. Descriptive analysis was performed, and data presented in tabular form. An area under the curve or p-value was presented for potential biomarkers discovered. RESULTS: Twenty-seven studies were included (metabolomics (n = 9)), and (proteomics (n = 18)) with 1818 participants. The samples analysed were urine (n = 19) blood (n = 4) and biopsy (n = 6). The typical outcome themes were potential biomarkers, disease phenotype, risk of progression and treatment response. CONCLUSION: This review shows the potential of metabolomic and proteomic analysis to discover new disease biomarkers that may influence diagnostics and disease management. Further larger-scale research is required to establish the validity of the study outcomes, including the several proposed biomarkers.


Asunto(s)
Biomarcadores , Glomerulonefritis , Metabolómica , Proteómica , Humanos , Metabolómica/métodos , Glomerulonefritis/metabolismo , Glomerulonefritis/terapia , Glomerulonefritis/diagnóstico , Proteómica/métodos , Biomarcadores/orina , Biomarcadores/sangre , Espectrometría de Masas
13.
Naunyn Schmiedebergs Arch Pharmacol ; 397(9): 6779-6789, 2024 09.
Artículo en Inglés | MEDLINE | ID: mdl-38536435

RESUMEN

Chronic glomerulonephritis (CGN) refers to the inflammation of glomeruli in the kidneys. Glomerular mesangial cells (GMCs) play a pivotal role in the development of CGN. In the present study, we investigated the impact of ALKBH5, a m6A demethylase, on inflammation and hyperproliferation in mouse glomerular mesangial cells (MMCs) and elucidated the molecular mechanisms contributing to CGN. Western blotting and reverse transcriptase-polymerase chain reaction (RT-qPCR) were employed to evaluate the expression of ALKBH5 and TRIM13. In addition, enzyme-linked immunosorbent assay (ELISA) was used to measure the levels of inflammatory factors (IL-1ß, TNF-α, and IL-10) in the lipopolysaccharide (LPS)-induced MMCs supernatant. Methylated RNA immunoprecipitation (MeRIP) was performed to investigate the effect of ALKBH5 on the levels of TRIM13-m6A mRNA. The stability of TRIM13 mRNA was evaluated using an actinomycin D assay. Significantly elevated expression of ALKBH5 was found in LPS-induced MMCs. Interference with ALKBH5 expression inhibited inflammation and excessive proliferation in LPS-induced MMCs. Moreover, interfering with ALKBH5 expression significantly reduced the levels of TRIM13-m6A modification. The overexpression of TRIM13 in MMCs reversed the inflammation and proliferation induced by ALKBH5 interference. In addition, interference with TRIM13 expression inhibited the activation of the NF-κB pathway and suppressed inflammation and proliferation in MMCs. Inhibiting ALKBH5 hinders inflammation and hyperproliferation by improving TRIM13-m6A modification in glomerular MCs. We believe these findings will further provide insights into the molecular mechanisms and potential therapeutic targets for CGN.


Asunto(s)
Desmetilasa de ARN, Homólogo 5 de AlkB , Proliferación Celular , Inflamación , Lipopolisacáridos , Células Mesangiales , Animales , Proliferación Celular/efectos de los fármacos , Células Mesangiales/metabolismo , Células Mesangiales/efectos de los fármacos , Desmetilasa de ARN, Homólogo 5 de AlkB/metabolismo , Desmetilasa de ARN, Homólogo 5 de AlkB/genética , Inflamación/metabolismo , Inflamación/patología , Ratones , Lipopolisacáridos/farmacología , Proteínas de Motivos Tripartitos/genética , Proteínas de Motivos Tripartitos/metabolismo , Glomerulonefritis/metabolismo , Glomerulonefritis/patología , Glomerulonefritis/genética , Células Cultivadas , Adenosina/análogos & derivados , Adenosina/metabolismo , Adenosina/farmacología , FN-kappa B/metabolismo
14.
Kidney Int ; 105(6): 1291-1305, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38537677

RESUMEN

Antineutrophil cytoplasmic antibody (ANCA)-associated vasculitis (AAV) is a systemic autoimmune disease pathologically characterized by vascular necrosis with inflammation. During AAV development, activated neutrophils produce reactive oxygen species (ROS), leading to the aberrant formation of neutrophil extracellular traps (NETs) via NETosis and subsequent fibrinoid vascular necrosis. Nuclear factor-erythroid 2-related factor 2 (Nrf2) functions as an intracellular defense system to counteract oxidative stress by providing antioxidant properties. Herein, we explored the role of Nrf2 in the pathogenesis of AAV. The role and mechanism of Nrf2 in ANCA-stimulated neutrophils and subsequent endothelial injury were evaluated in vitro using Nrf2 genetic deletion and Nrf2 activator treatment. In corresponding in vivo studies, the role of Nrf2 in ANCA-transfer AAV and spontaneous AAV murine models was examined. Pharmacological activation of Nrf2 in vitro suppressed ANCA-induced NET formation via the inhibition of ROS. In contrast, NET formation was enhanced in Nrf2-deficient neutrophils. Furthermore, Nrf2 activation protected endothelial cells from ANC-induced NETs-mediated injury. In vivo, Nrf2 activation ameliorated glomerulonephritis in two AAV models by upregulating antioxidants and inhibiting ROS-mediated NETs. Furthermore, Nrf2 activation restrained the expansion of splenic immune cells, including T lymphocytes and limited the infiltration of Th17 cells into the kidney. In contrast, Nrf2 genetic deficiency exacerbated vasculitis in a spontaneous AAV model. Thus, the pathophysiological process in AAV may be downregulated by Nrf2 activation, potentially leading to a new therapeutic strategy by regulating NETosis.


Asunto(s)
Vasculitis Asociada a Anticuerpos Citoplasmáticos Antineutrófilos , Modelos Animales de Enfermedad , Trampas Extracelulares , Ratones Noqueados , Factor 2 Relacionado con NF-E2 , Neutrófilos , Peroxidasa , Especies Reactivas de Oxígeno , Animales , Factor 2 Relacionado con NF-E2/metabolismo , Factor 2 Relacionado con NF-E2/genética , Trampas Extracelulares/inmunología , Trampas Extracelulares/metabolismo , Vasculitis Asociada a Anticuerpos Citoplasmáticos Antineutrófilos/inmunología , Vasculitis Asociada a Anticuerpos Citoplasmáticos Antineutrófilos/patología , Vasculitis Asociada a Anticuerpos Citoplasmáticos Antineutrófilos/genética , Neutrófilos/inmunología , Neutrófilos/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Peroxidasa/metabolismo , Peroxidasa/genética , Ratones , Humanos , Estrés Oxidativo/inmunología , Ratones Endogámicos C57BL , Células Endoteliales/inmunología , Células Endoteliales/metabolismo , Células Endoteliales/patología , Glomerulonefritis/inmunología , Glomerulonefritis/patología , Glomerulonefritis/genética , Glomerulonefritis/metabolismo , Glomerulonefritis/etiología , Anticuerpos Anticitoplasma de Neutrófilos/inmunología , Masculino , Riñón/patología , Riñón/inmunología , Transducción de Señal/inmunología
15.
Nat Commun ; 15(1): 2598, 2024 Mar 22.
Artículo en Inglés | MEDLINE | ID: mdl-38519468

RESUMEN

Activation of the mechanistic target of rapamycin (mTOR) is a key metabolic checkpoint of pro-inflammatory T-cell development that contributes to the pathogenesis of autoimmune diseases, such as systemic lupus erythematosus (SLE), however, the underlying mechanisms remain poorly understood. Here, we identify a functional role for Rab4A-directed endosome traffic in CD98 receptor recycling, mTOR activation, and accumulation of mitochondria that connect metabolic pathways with immune cell lineage development and lupus pathogenesis. Based on integrated analyses of gene expression, receptor traffic, and stable isotope tracing of metabolic pathways, constitutively active Rab4AQ72L exerts cell type-specific control over metabolic networks, dominantly impacting CD98-dependent kynurenine production, mTOR activation, mitochondrial electron transport and flux through the tricarboxylic acid cycle and thus expands CD4+ and CD3+CD4-CD8- double-negative T cells over CD8+ T cells, enhancing B cell activation, plasma cell development, antinuclear and antiphospholipid autoantibody production, and glomerulonephritis in lupus-prone mice. Rab4A deletion in T cells and pharmacological mTOR blockade restrain CD98 expression, mitochondrial metabolism and lineage skewing and attenuate glomerulonephritis. This study identifies Rab4A-directed endosome traffic as a multilevel regulator of T cell lineage specification during lupus pathogenesis.


Asunto(s)
Glomerulonefritis , Lupus Eritematoso Sistémico , Animales , Ratones , Linfocitos T CD8-positivos/metabolismo , Endosomas/metabolismo , Glomerulonefritis/metabolismo , Quinurenina/metabolismo , Mitocondrias/metabolismo , Mitofagia , Serina-Treonina Quinasas TOR/metabolismo , Proteínas de Unión al GTP rab4/metabolismo
16.
Inhal Toxicol ; 36(2): 106-123, 2024 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-38477125

RESUMEN

OBJECTIVE: Occupational exposure to respirable crystalline silica (cSiO2) has been linked to lupus development. Previous studies in young lupus-prone mice revealed that intranasal cSiO2 exposure triggered autoimmunity, preventable with docosahexaenoic acid (DHA). This study explores cSiO2 and DHA effects in mature lupus-prone adult mice, more representative of cSiO2-exposed worker age. METHODS: Female NZBWF1 mice (14-week old) were fed control (CON) or DHA-supplemented diets. After two weeks, mice were intranasally instilled saline (VEH) or 1 mg cSiO2 weekly for four weeks. Cohorts were then analyzed 1- and 5-weeks postinstillation for lung inflammation, cell counts, chemokines, histopathology, B- and T-cell infiltration, autoantibodies, and gene signatures, with results correlated to autoimmune glomerulonephritis onset. RESULTS: VEH/CON mice showed no pathology. cSiO2/CON mice displayed significant ectopic lymphoid tissue formation in lungs at 1 week, increasing by 5 weeks. cSiO2/CON lungs exhibited elevated cellularity, chemokines, CD3+ T-cells, CD45R + B-cells, IgG + plasma cells, gene expression, IgG autoantibodies, and glomerular hypertrophy. DHA supplementation mitigated all these effects. DISCUSSION: The mature adult NZBWF1 mouse used here represents a life-stage coincident with immunological tolerance breach and one that more appropriately represents the age (20-30 yr) of cSiO2-exposed workers. cSiO2-induced robust pulmonary inflammation, autoantibody responses, and glomerulonephritis in mature adult mice, surpassing effects observed previously in young adults. DHA at a human-equivalent dosage effectively countered cSiO2-induced inflammation/autoimmunity in mature mice, mirroring protective effects in young mice. CONCLUSION: These results highlight life-stage significance in this preclinical lupus model and underscore omega-3 fatty acids' therapeutic potential against toxicant-triggered autoimmune responses.


Asunto(s)
Ácidos Grasos Omega-3 , Glomerulonefritis , Neumonía , Femenino , Ratones , Humanos , Animales , Ácidos Grasos Omega-3/toxicidad , Autoinmunidad , Dióxido de Silicio/toxicidad , Neumonía/inducido químicamente , Glomerulonefritis/inducido químicamente , Glomerulonefritis/metabolismo , Glomerulonefritis/patología , Ácidos Docosahexaenoicos/toxicidad , Quimiocinas/toxicidad , Autoanticuerpos , Inmunoglobulina G
17.
J Nanobiotechnology ; 22(1): 9, 2024 Jan 03.
Artículo en Inglés | MEDLINE | ID: mdl-38169389

RESUMEN

Glomerulonephritis (GN) is the most common cause of end-stage renal failure worldwide; in most cases, it cannot be cured and can only delay the progression of the disease. At present, the main treatment methods include symptomatic therapy, immunosuppressive therapy, and renal replacement therapy. However, effective treatment of GN is hindered by issues such as steroid resistance, serious side effects, low bioavailability, and lack of precise targeting. With the widespread application of nanoparticles in medical treatment, novel methods have emerged for the treatment of kidney diseases. Targeted transportation of drugs, nucleic acids, and other substances to kidney tissues and even kidney cells through nanodrug delivery systems can reduce the systemic effects and adverse reactions of drugs and improve treatment effectiveness. The high specificity of nanoparticles enables them to bind to ion channels and block or enhance channel gating, thus improving inflammation. This review briefly introduces the characteristics of GN, describes the treatment status of GN, systematically summarizes the research achievements of nanoparticles in the treatment of primary GN, diabetic nephropathy and lupus nephritis, analyzes recent therapeutic developments, and outlines promising research directions, such as gas signaling molecule nanodrug delivery systems and ultrasmall nanoparticles. The current application of nanoparticles in GN is summarized to provide a reference for better treatment of GN in the future.


Asunto(s)
Nefropatías Diabéticas , Glomerulonefritis , Nefritis Lúpica , Humanos , Glomerulonefritis/tratamiento farmacológico , Glomerulonefritis/metabolismo , Riñón/metabolismo , Nanotecnología
18.
Biomed Pharmacother ; 165: 115233, 2023 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-37536037

RESUMEN

Shenhua tablet (SH), a formulation of traditional Chinese medicine, exerts renoprotective effect on chronic kidney diseases, and it has been found to restrain inflammation, but the mechanism is still unclear. Here, we explored the potential renoprotection of SH in mesangial proliferative glomerulonephritis (MsPGN) rat model induced by anti-Thy1 antibody. Administration of SH reduced urinary albumin/creatinine ratio (UACR) and significantly attenuated mesangial cell proliferation and renal inflammation. Notably, SH protected rats against renal inflammation, which was associated with decreasing macrophage infiltration and promoting macrophage anti-inflammatory activity. Network analysis combined with arrays identified the Janus kinase signal transducer and activator of transcription (JAK-STAT) signaling pathway as the main pathways of SH could target inflammation. Furthermore, it was confirmed that mesangial cell proliferation, which response to inflammation, were alleviated by ASS1 expression enhanced after SH administration both in vivo and in vitro. Collectively, SH has the beneficial on relieving the progression of MsPGN to alleviate inflammation and mesangial proliferation by inhibiting STAT3 phosphorylation and maintains the expression level of ASS1, might be an effective strategy for treating MsPGN.


Asunto(s)
Glomerulonefritis , Nefritis , Ratas , Animales , Ratas Wistar , Glomerulonefritis/tratamiento farmacológico , Glomerulonefritis/metabolismo , Inflamación/tratamiento farmacológico , Proliferación Celular , Comprimidos/efectos adversos
19.
Acupunct Med ; 41(5): 307-316, 2023 10.
Artículo en Inglés | MEDLINE | ID: mdl-37166069

RESUMEN

OBJECTIVE: The hypothalamic paraventricular nucleus (PVN) acts as a critical integrating center of endocrine/autonomic responses and regulates visceral functional activities. However, its involvement in electroacupuncture (EA) treatment of chronic glomerulonephritis (CGN) remains unclear. METHODS: Over four experiments, we randomized 111 rats into: control, untreated model (CGN) or EA-treated model (CGN + EA) groups, a model group receiving EA after PVN damage (CGN + EA + Lesion) or untreated model groups injected with adeno-associated viral vectors encoding human M4 muscarinic receptor (CGN + hM4D) or enhanced green fluorescent protein (CGN + EGFP). CGN was modeled by intraperitoneal injection of bovine serum albumin for 2 weeks. Rats in the CGN + EA and CGN + EA + Lesion groups received EA at bilateral ST36 and KI3 for 14 days. Urine/serum samples were collected to evaluate inflammatory factors and changes in renal function. RESULTS: EA inhibited the release of interleukin (IL)-6, tumor necrosis factor (TNF)-α and IL-1ß, and decreased urine protein (PRO), creatinine (Cre) and blood urea nitrogen (BUN) levels. PVN damage influenced the effect of EA on the levels of these parameters. EA appeared to inhibit the firing frequency and spectral energy of PVN neurons. In the viral vector experiment, levels of PRO, Cre, IL-6, IL-1ß and TNF-α in the CGN group were increased in CGN versus control groups (p < 0.0001), decreased in CGN + hM4D versus CGN groups (p < 0.05) and did not differ between CGN + EGFP and control groups (p > 0.05). CONCLUSION: Our findings indicate that EA at ST36 and KI3 improves CGN in this rat model by weakening the activity of PVN neurons, alleviating impairment of renal function impairment and restricting the release of inflammatory factors.


Asunto(s)
Electroacupuntura , Glomerulonefritis , Humanos , Ratas , Animales , Núcleo Hipotalámico Paraventricular/metabolismo , Enfermedad Crónica , Factor de Necrosis Tumoral alfa/metabolismo , Glomerulonefritis/metabolismo , Interleucina-6/metabolismo
20.
Pediatr Nephrol ; 38(11): 3711-3719, 2023 11.
Artículo en Inglés | MEDLINE | ID: mdl-37231123

RESUMEN

BACKGROUND: The current study tested the hypothesis that urinary angiotensinogen (UAGT) and urinary monocyte chemoattractant protein-1 (UMCP-1) levels provide a specific index of intrarenal renin-angiotensin system (RAS) status and the degree of infiltration of macrophages associated with RAS blockade and immunosuppressant treatment in pediatric patients with chronic glomerulonephritis. METHODS: We measured baseline UAGT and UMCP-1 levels to examine the correlation between glomerular injury in 48 pediatric chronic glomerulonephritis patients before treatment. Furthermore, we performed immunohistochemical analysis of angiotensinogen (AGT) and CD68 in 27 pediatric chronic glomerulonephritis patients treated with RAS blockades and immunosuppressants for 2 years. Finally, we examined the effects of angiotensin II (Ang II) on monocyte chemoattractant protein-1 (MCP-1) expression in cultured human mesangial cells (MCs). RESULTS: Baseline UAGT and UMCP-1 levels positively correlated with urinary protein levels, scores for mesangial hypercellularity, rate of crescentic formation, and expression levels of AGT and CD68 in renal tissues (p < 0.05). UAGT and UMCP-1 levels were significantly decreased after RAS blockade and immunosuppressant treatment (p < 0.01), which was accompanied by AGT and CD68 (p < 0.01), as well as the magnitude of glomerular injury. Cultured human MCs showed increased MCP-1 messenger ribonucleic acid and protein levels after Ang II treatment (p < 0.01). CONCLUSIONS: The data indicates that UAGT and UMCP-1 are useful biomarkers of the degree of glomerular injury during RAS blockade and immunosuppressant treatment in pediatric patients with chronic glomerulonephritis.


Asunto(s)
Glomerulonefritis , Sistema Renina-Angiotensina , Humanos , Niño , Angiotensinógeno/orina , Riñón/metabolismo , Quimiocina CCL2 , Glomerulonefritis/metabolismo , Angiotensina II/metabolismo , Enfermedad Crónica , Inmunosupresores/farmacología , Inmunosupresores/uso terapéutico , Macrófagos/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA