Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 30
Filtrar
Más filtros











Intervalo de año de publicación
1.
Cells ; 13(10)2024 May 18.
Artículo en Inglés | MEDLINE | ID: mdl-38786096

RESUMEN

Uncovering the function of understudied G protein-coupled receptors (GPCRs) provides a wealth of untapped therapeutic potential. The poorly understood adhesion GPCR Gpr126 (Adgrg6) is widely expressed in developing kidneys. In adulthood, Gpr126 expression is enriched in parietal epithelial cells (PECs) and epithelial cells of the collecting duct and urothelium. Whether Gpr126 plays a role in kidney disease remains unclear. Here, we characterized Gpr126 expression in diseased kidneys in mice, rats, and humans. RT-PCR data show that Gpr126 expression is altered in kidney disease. A quantitative RNAscope® analysis utilizing cell type-specific markers revealed that Gpr126 expression upon tubular damage is mainly increased in cell types expressing Gpr126 under healthy conditions as well as in cells of the distal and proximal tubules. Upon glomerular damage, an increase was mainly detected in PECs. Notably, Gpr126 expression was upregulated in an ischemia/reperfusion model within hours, while upregulation in a glomerular damage model was only detected after weeks. An analysis of kidney microarray data from patients with lupus nephritis, IgA nephropathy, focal segmental glomerulosclerosis (FSGS), hypertension, and diabetes as well as single-cell RNA-seq data from kidneys of patients with acute kidney injury and chronic kidney disease indicates that GPR126 expression is also altered in human kidney disease. In patients with FSGS, an RNAscope® analysis showed that GPR126 mRNA is upregulated in PECs belonging to FSGS lesions and proximal tubules. Collectively, we provide detailed insights into Gpr126 expression in kidney disease, indicating that GPR126 is a potential therapeutic target.


Asunto(s)
Riñón , Receptores Acoplados a Proteínas G , Receptores Acoplados a Proteínas G/metabolismo , Receptores Acoplados a Proteínas G/genética , Animales , Humanos , Ratas , Ratones , Riñón/metabolismo , Riñón/patología , Enfermedades Renales/metabolismo , Enfermedades Renales/genética , Enfermedades Renales/patología , Masculino , Perfilación de la Expresión Génica , Ratones Endogámicos C57BL , Femenino
2.
Int J Mol Sci ; 25(2)2024 Jan 06.
Artículo en Inglés | MEDLINE | ID: mdl-38255818

RESUMEN

Renal biopsy is the gold standard for making the final diagnosis and for predicting the progression of renal disease, but monitoring disease status by performing biopsies repeatedly is impossible because it is an invasive procedure. Urine tests are non-invasive and may reflect the general condition of the whole kidney better than renal biopsy results. We therefore investigated the diagnostic value of extensive urinary sediment analysis by immunofluorescence staining for markers expressed on kidney-derived cells (cytokeratin: marker for tubular epithelial cells, synaptopodin: marker for podocytes, claudin1: marker for parietal epithelial cells, CD68: marker for macrophages (MΦ), neutrophil elastase: marker for neutrophils). We further examined the expression levels of the mRNAs for these markers by real-time reverse transcription polymerase chain reaction. We also examined the levels of mRNAs associated with the M1 (iNOS, IL-6) and M2 (CD163, CD204, CD206, IL-10) MΦ phenotypes. Evaluated markers were compared with clinical and histological findings for the assessment of renal diseases. Claudin1- and CD68-positive cell counts in urinary sediments were higher in patients with glomerular crescents (especially cellular crescents) than in patients without crescents. The relative levels of mRNA for CD68 and the M2 MΦ markers (CD163, CD204, CD206, and IL-10) in urinary sediments were also higher in patients with glomerular crescents. These data suggest that immunofluorescence staining for claudin1 and CD68 in urinary sediments and the relative levels of mRNA for CD68 and M2 MΦ markers in urinary sediments are useful for evaluating the state of glomerular diseases.


Asunto(s)
Enfermedades Renales , Sistema Urinario , Humanos , Interleucina-10 , Riñón , Técnica del Anticuerpo Fluorescente
3.
Cells ; 12(15)2023 08 02.
Artículo en Inglés | MEDLINE | ID: mdl-37566066

RESUMEN

Adhesion G protein-coupled receptors (aGPCRs) comprise the second-largest class of GPCRs, the most common target for approved pharmacological therapies. aGPCRs play an important role in development and disease and have recently been associated with the kidney. Several aGPCRs are expressed in the kidney and some aGPCRs are either required for kidney development or their expression level is altered in diseased kidneys. Yet, general aGPCR function and their physiological role in the kidney are poorly understood. Here, we characterize in detail Gpr126 (Adgrg6) expression based on RNAscope® technology in zebrafish, mice, and humans during kidney development in adults. Gpr126 expression is enriched in the epithelial linage during nephrogenesis and persists in the adult kidney in parietal epithelial cells, collecting ducts, and urothelium. Single-cell RNAseq analysis shows that gpr126 expression is detected in zebrafish in a distinct ionocyte sub-population. It is co-detected selectively with slc9a3.2, slc4a4a, and trpv6, known to be involved in apical acid secretion, buffering blood or intracellular pH, and to maintain high cytoplasmic Ca2+ concentration, respectively. Furthermore, gpr126-expressing cells were enriched in the expression of potassium transporter kcnj1a.1 and gcm2, which regulate the expression of a calcium sensor receptor. Notably, the expression patterns of Trpv6, Kcnj1a.1, and Gpr126 in mouse kidneys are highly similar. Collectively, our approach permits a detailed insight into the spatio-temporal expression of Gpr126 and provides a basis to elucidate a possible role of Gpr126 in kidney physiology.


Asunto(s)
Receptores Acoplados a Proteínas G , Pez Cebra , Animales , Humanos , Ratones , Proteínas de Unión al ADN , Riñón/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Factores de Transcripción , Pez Cebra/metabolismo , Proteínas de Pez Cebra/genética , Proteínas de Pez Cebra/metabolismo
4.
BMC Nephrol ; 24(1): 139, 2023 05 22.
Artículo en Inglés | MEDLINE | ID: mdl-37217871

RESUMEN

BACKGROUND: Extra-capillary hypercellularity is a common finding in crescentic glomerulonephritis (GN) and focal segmental glomerulosclerosis (FSGS). In diabetic nephropathy (DN), extra-capillary hypercellularity is often observed as a finding of complications such as IgA nephropathy or microscopic polyangiitis superimposed on DN. However, in rare cases, epithelial cell proliferation may accompany DN. We experienced a case of nodular diabetic glomerulosclerosis with marked extra-capillary hypercellularity and revealed the origin of this atypical lesion using immunostainings. CASE PRESENTATION: A man in his 50 s was admitted to the hospital with nephrotic syndrome, and a renal biopsy was performed. Diffuse nodular lesions and extra-capillary hypercellularity were observed, but the results of serological examination or immunofluorescent assays did not implicate any other crescentic GN. Immunostaining for claudin-1 and nephrin was performed to identify the origin of the extra-capillary lesions. Given the clinical course and pathological findings, a diagnosis of DN-associated extra-capillary cell proliferation was made. CONCLUSIONS: Extra-capillary hypercellularity, which resembles FSGS or crescentic GN, is a rare finding in DN and should therefore be treated with caution. In such cases, co-staining for claudin-1 and nephrin may facilitate the diagnosis of DN.


Asunto(s)
Diabetes Mellitus , Nefropatías Diabéticas , Glomerulonefritis por IGA , Glomerulonefritis Membranoproliferativa , Glomeruloesclerosis Focal y Segmentaria , Humanos , Masculino , Proliferación Celular , Claudina-1 , Diabetes Mellitus/diagnóstico , Nefropatías Diabéticas/diagnóstico , Nefropatías Diabéticas/patología , Glomerulonefritis por IGA/patología , Glomeruloesclerosis Focal y Segmentaria/patología , Persona de Mediana Edad , Proteínas de la Membrana , Poliangitis Microscópica
5.
Biomolecules ; 13(2)2023 01 31.
Artículo en Inglés | MEDLINE | ID: mdl-36830635

RESUMEN

Glomerular parietal epithelial cells (PECs) have been increasingly recognized to have crucial functions. Lineage tracking in animal models showed the expression of a podocyte phenotype by PECs during normal glomerular growth and after acute podocyte injury, suggesting a reparative role of PECs. Conversely, activated PECs are speculated to be pathogenic and comprise extracapillary proliferation in focal segmental glomerulosclerosis (FSGS) and crescentic glomerulonephritis (CrescGN). The reparative and pathogenic roles of PECs seem to represent two sides of PEC behavior directed by the local milieu and mediators. Recent studies suggest microRNA-193a (miR193a) is involved in the pathogenesis of FSGS and CrescGN. In a mouse model of primary FSGS, the induction of miR193a caused the downregulation of Wilms' tumor protein, leading to the dedifferentiation of podocytes. On the other hand, the inhibition of miR193a resulted in reduced crescent lesions in a mouse model of CrescGN. Interestingly, in vitro studies report that the downregulation of miR193a induces trans-differentiation of PECs into a podocyte phenotype. This narrative review highlights the critical role of PEC behavior in health and during disease and its modulation by miR193a.


Asunto(s)
Glomeruloesclerosis Focal y Segmentaria , MicroARNs , Podocitos , Ratones , Animales , Glomeruloesclerosis Focal y Segmentaria/metabolismo , Glomeruloesclerosis Focal y Segmentaria/patología , Células Epiteliales/metabolismo , Glomérulos Renales/metabolismo , Glomérulos Renales/patología , Podocitos/metabolismo , MicroARNs/genética
6.
Front Med (Lausanne) ; 9: 697443, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35223886

RESUMEN

Glucocorticoids are commonly used to treat anti-GBM crescentic glomerulonephritis, however, the mechanism underlying its therapeutic effectiveness is not completely understood. Since podocyte EGFR/STAT3 signaling is known to mediate the development of anti-GBM glomerulonephritis, we investigated the effect of glucocorticoids on EGFR/STAT3 signaling in podocytes. We found that the levels of phosphorylated (activated) EGFR and STAT3 in podocytes were markedly elevated in anti-GBM patients without glucocorticoids treatment, but were normalized in patients with glucocorticoids treatment. In a rat model of anti-GBM glomerulonephritis, glucocorticoids treatment significantly attenuated the proteinuria, crescent formation, parietal epithelial cell (PEC) activation and proliferation, accompanied by elimination of podocyte EGFR/STAT3 signaling activation. In cultured podocytes, glucocorticoids were found to inhibit HB-EGF-induced EGFR and STAT3 activation. The conditioned medium from podocytes treated with HB-EGF in the absence but not presence of glucocorticoids was capable of activating Notch signaling (which is known to be involved in PEC proliferation and crescent formation) and enhancing proliferative activity in primary PECs, suggesting that glucocorticoids prevent podocytes from producing secreted factors that cause PEC proliferation and crescent formation. Furthermore, we found that glucocorticoids can downregulate the expression of EGFR ligands, EGF and HB-EGF, while upregulate the expression of EGFR inhibitor, Gene 33, explaining how glucocorticoids suppress EGFR signaling. Taken together, glucocorticoids exert therapeutic effect on anti-GBM crescentic glomerulonephritis through inhibiting podocyte EGFR/STAT3 signaling and the downstream pathway that leads to PEC proliferation and crescent formation.

7.
Front Med (Lausanne) ; 8: 814497, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-35096904

RESUMEN

Organ fibrogenesis is characterized by a common pathophysiological final pathway independent of the underlying progressive disease of the respective organ. This makes it particularly suitable as a therapeutic target. The Transregional Collaborative Research Center "Organ Fibrosis: From Mechanisms of Injury to Modulation of Disease" (referred to as SFB/TRR57) was hosted from 2009 to 2021 by the Medical Faculties of RWTH Aachen University and the University of Bonn. This consortium had the ultimate goal of discovering new common but also different fibrosis pathways in the liver and kidneys. It finally successfully identified new mechanisms and established novel therapeutic approaches to interfere with hepatic and renal fibrosis. This review covers the consortium's key kidney-related findings, where three overarching questions were addressed: (i) What are new relevant mechanisms and signaling pathways triggering renal fibrosis? (ii) What are new immunological mechanisms, cells and molecules that contribute to renal fibrosis?, and finally (iii) How can renal fibrosis be modulated?

8.
Am J Physiol Renal Physiol ; 319(4): F636-F646, 2020 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-32830536

RESUMEN

Mitotic spindle assembly checkpoint protein 2 (MAD2B), a well-known anaphase-promoting complex/cyclosome (APC/C) inhibitor and a small subunit of DNA polymerase-ζ, is critical for mitotic control and DNA repair. Previously, we detected a strong increase of MAD2B in the glomeruli from patients with crescentic glomerulonephritis and anti-glomerular basement membrane (anti-GBM) rats, which predominantly originated from activated parietal epithelial cells (PECs). Consistently, in vitro MAD2B was increased in TNF-α-treated PECs, along with cell activation and proliferation, as well as extracellular matrix accumulation, which could be reversed by MAD2B genetic depletion. Furthermore, we found that expression of S phase kinase-associated protein 2 (Skp2), an APC/CCDH1 substrate, was increased in the glomeruli of anti-GBM rats, and TNF-α-stimulated PECs and could be suppressed by MAD2B depletion. Additionally, genetic deletion of Skp2 inhibited TNF-α-induced PEC activation and dysfunction. Finally, TNF-α blockade or glucocorticoid therapy administered to anti-GBM rats could ameliorate MAD2B and Skp2 accumulation as well as weaken PEC activation. Collectively, our data suggest that MAD2B has a pivotal role in the pathogenesis of glomerular PEC activation and crescent formation through induction of Skp2 expression.


Asunto(s)
Proliferación Celular , Células Epiteliales/metabolismo , Glomerulonefritis/enzimología , Glomérulos Renales/metabolismo , Proteínas Mad2/metabolismo , Proteínas Quinasas Asociadas a Fase-S/metabolismo , Animales , Proliferación Celular/efectos de los fármacos , Células Epiteliales/efectos de los fármacos , Células Epiteliales/patología , Etanercept/farmacología , Matriz Extracelular/metabolismo , Matriz Extracelular/patología , Regulación de la Expresión Génica , Glomerulonefritis/tratamiento farmacológico , Glomerulonefritis/genética , Glomerulonefritis/patología , Glucocorticoides/farmacología , Humanos , Glomérulos Renales/efectos de los fármacos , Glomérulos Renales/patología , Proteínas Mad2/genética , Masculino , Ratones , Prednisolona/análogos & derivados , Prednisolona/farmacología , Células RAW 264.7 , Ratas Endogámicas WKY , Proteínas Quinasas Asociadas a Fase-S/genética , Transducción de Señal
9.
Ann Transl Med ; 8(6): 355, 2020 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-32355799

RESUMEN

BACKGROUND: Mesangial proliferative glomerulonephritis (MsPGN) is an epidemic disease with increasing occurrence. As important as mesangial cells, podocytes are key innate cells for MsPGN prognosis and recovery. Renal progenitor cells, located at the urinary pole (UP) of Bowman's capsule (BC), could alleviate kidney injury through their capacity to differentiate into podocytes. METHODS: Seventy-two male rats were categorized randomly into the sham (n=24), untreated Thy-1 (n=24) and losartan-treated (n=24) groups. We administered vehicle or losartan (50 mg/kg by gavage) daily to treat rats with anti-thy1.1 nephritis, an ideal model to simulate human MsPGN. Two weeks after the intravenous injection of antibody, urinary protein and blood samples were analyzed, pathological changes were examined, the number of podocytes was determined, and renal progenitor cells were studied. RESULTS: Anti-thy1.1 nephritis was significantly alleviated after losartan treatment, as reported previously and as expected. Compared with the untreated Thy-1 group, the number of podocytes in the losartan group increased, and the area of renal progenitor cells significantly increased. The protein expression of components of the p-ERK pathway was determined during the development of renal progenitor cells differentiating into podocytes. CONCLUSIONS: The data in this paper show the direct glomerular cell action of angiotensin II receptor blocker (ARB) treatment in improving outcomes in anti-thy1.1 nephritis. The positive effects of ARB medication on anti-thy1.1 nephritis were due to an increase in the number of renal epithelial progenitor cells (defined as PECs that expressed only stem cell markers without podocyte proteins).

10.
Kidney Dis (Basel) ; 6(2): 119-124, 2020 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-32309294

RESUMEN

INTRODUCTION: Minimal change disease (MCD) and primary focal segmental glomerulosclerosis (FSGS) are representative podocyte diseases. The clinical cause of MCD and FSGS has not been clearly elucidated yet. However, it is important to distinguish MCD and FSGS because their prognoses and responses to treatment are quite different. OBJECTIVE: This study aimed to examine whether parietal epithelial cell (PEC) marker and repeat biopsy are useful for diagnosing primary FSGS. METHODS: Clinicopathological features of 17 patients with the nephrotic syndrome, who underwent kidney biopsy ≥2 times from 1975 to 2017, and had MCD or FSGS were analyzed using PAX8. We defined patients with PAX8+ cells as PAX8+ and the remainder as PAX8- patients. Three cases of sample insufficiency and 1 non-steroid-resistant or frequently relapsing case indicated for repeat biopsy were excluded. RESULTS: Among the 13 patients studied, 4 were PAX8+ and 9 were PAX8- (median age: 41 and 46 years, -respectively, at first biopsy). PAX8+ and PAX8- patients showed no significant differences in clinical data and histological diagnosis except for a significant difference in histological diagnosis at the second biopsy. The number of PAX8+ patients increased to 6. Unlike the first biopsy results, FSGS was present in 5 of 6 (83.3%) PAX8+ patients; MCD occurred in all 7 (100%) PAX8- patients. Three of 6 (50.0%) PAX8+ patients undergoing repeat biopsy were steroid resistant; no (0%) PAX8- patient was steroid resistant. All cases of final FSGS diagnosis were PAX8+ at the first or second biopsy. Only 1 PAX8+ MCD patient was steroid resistant. All PAX8- MCD patients were frequently relapsing. CONCLUSIONS: More PAX8+ patients were diagnosed with FSGS than PAX8- patients. Clinical presentation of MCD in PAX8- patients was frequently relapsing. PEC marker staining in patients with the nephrotic syndrome, e.g., MCD, may help to diagnose FSGS.

11.
Cell Tissue Res ; 380(3): 581-591, 2020 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-31989254

RESUMEN

Phenotypic changes in culture hamper the identification and characterization of cultured podocytes and parietal epithelial cells of the Bowman's capsule (PECs). We have recently established culture conditions that restore podocytes to their differentiated phenotypes. We compared podocytes and PECs cultured under the same conditions to determine the unique characteristics of the two cell types. Performing this comparison under the same conditions accentuated these differences. Podocytes behaved like non-epithelial cells by extending cell processes even at confluence. By contrast, PECs behaved like typical epithelial cells by maintaining a polygonal appearance. Other differences were identified using immunostaining and RT-PCR; podocytes expressed high levels of podocyte-specific markers while PECs expressed high levels of PEC-specific markers. However, while podocytes expressed low levels of PEC markers, PECs expressed low levels of podocyte markers. Therefore, the identification of podocytes and PECs in culture requires the evaluation of respective cell markers and the expression of markers for other cell types.


Asunto(s)
Cápsula Glomerular/citología , Células Epiteliales/citología , Podocitos/citología , Animales , Biomarcadores/metabolismo , Células Cultivadas , Ratas
12.
Am J Physiol Renal Physiol ; 317(6): F1680-F1694, 2019 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-31630546

RESUMEN

In healthy glomeruli, parietal epithelial cell (PEC)-derived extracellular matrix (ECM) proteins include laminin-ß1, perlecan, and collagen type IV-α2 and podocyte-specific ECM proteins include laminin-ß2, agrin, and collagen type IV-α4. This study aimed to define individual ECM protein isoform expression by PECs in both experimental and human focal segmental glomerulosclerosis (FSGS) and diabetic nephropathy (DN) and to determine if changes were CD44 dependent. In experimental FSGS induced with a cytotoxic podocyte antibody and in the BTBR ob/ob mouse model of DN, PEC-derived protein staining was significantly increased in PECs. Dual staining also showed de novo expression of the podocyte-specific ECM proteins laminin-ß2 and agrin in PECs. Similar findings were observed in biopsies from patients with FSGS and DN. Increases in individual ECM proteins colocalized with CD44 in PECs in disease. To determine the role of CD44, FSGS was induced in CD44-/- and CD44+/+ mice. PEC staining for perlecan, collagen type IV-α2, laminin-ß2, and agrin were significantly lower in diseased CD44-/- mice compared with diseased CD44+/+ mice. These results show that in experimental and human FSGS and DN, PECs typically in an activated state, produce both PEC-derived and podocyte-specific ECM protein isoforms, and that the majority of these changes were dependent on CD44.


Asunto(s)
Nefropatías Diabéticas/metabolismo , Células Epiteliales/metabolismo , Proteínas de la Matriz Extracelular/metabolismo , Glomeruloesclerosis Focal y Segmentaria/metabolismo , Podocitos/metabolismo , Agrina/metabolismo , Animales , Colágeno Tipo IV/metabolismo , Nefropatías Diabéticas/patología , Glomeruloesclerosis Focal y Segmentaria/patología , Humanos , Receptores de Hialuranos/genética , Receptores de Hialuranos/metabolismo , Riñón/metabolismo , Riñón/patología , Laminina/metabolismo , Masculino , Ratones , Ratones Noqueados , Ratones Obesos
13.
J Biomed Mater Res A ; 107(12): 2718-2725, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31404486

RESUMEN

Progressive loss of glomerular podocytes during kidney disease leads to irreversible kidney failure, and is exacerbated by the fact that podocytes are terminally differentiated epithelial cells and unable to proliferate. Regeneration of lost podocytes must therefore derive from nonpodocyte sources. Human urine-derived renal progenitor cells (uRPCs) are attractive podocyte progenitors for cell therapy applications due to their availability from patient urine and ability to migrate to injured glomeruli and differentiate into de novo podocytes after intravenous administration. Because gene delivery has emerged as an important strategy to augment the functionality and survival of cell therapies prior to injection, in this work we optimized nonviral gene delivery conditions (cell density, DNA dose, % FBS, and transfection material composition) to primary uRPCs. Using the cationic polymer-peptide conjugate VIPER for gene delivery and the Sleeping Beauty transposon/transposase constructs for gene integration, we optimized transfection parameters to achieve efficient transgene expression (up to 55% transfected cells) and stable transgene expression (>65% integration efficiency) lasting up to 10 days. With these methods, we transfected uRPCs to overexpress CXCR4, an important chemokine receptor that mediates uRPC migration to the kidneys after intravenous injection, and demonstrate that CXCR4-uRPCs exhibit enhanced migration compared to mock-transfected cells.


Asunto(s)
Podocitos/citología , Células Madre/citología , Transfección , Orina/citología , Movimiento Celular , Células Cultivadas , Técnicas de Transferencia de Gen , Humanos , Podocitos/metabolismo , Receptores CXCR4/genética , Células Madre/metabolismo , Transfección/métodos
14.
J Histochem Cytochem ; 67(9): 623-632, 2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-31116068

RESUMEN

Focal segmental glomerulosclerosis (FSGS) presents with scar in parts of some glomeruli and often progresses to global and diffuse glomerulosclerosis. Podocyte injury is the initial target in primary FSGS, induced by a circulating factor. Several gene variants, for example, APOL1, are associated with increased susceptibility to FSGS. Primary FSGS may be due to genetic mutation in key podocyte genes. Increased work stress after loss of nephrons, epigenetic mechanisms, and various profibrotic pathways can contribute to progressive sclerosis, regardless of the initial injury. The progression of FSGS lesions also involves crosstalk between podocytes and other kidney cells, such as parietal epithelial cells, glomerular endothelial cells, and even tubular epithelial cells. New insights related to these mechanisms could potentially lead to new therapeutic strategies to prevent progression of FSGS.


Asunto(s)
Cicatriz/patología , Glomeruloesclerosis Focal y Segmentaria/patología , Glomérulos Renales/patología , Animales , Apolipoproteína L1/genética , Cicatriz/genética , Progresión de la Enfermedad , Células Endoteliales/metabolismo , Células Endoteliales/patología , Epigénesis Genética , Células Epiteliales/metabolismo , Células Epiteliales/patología , Glomeruloesclerosis Focal y Segmentaria/genética , Humanos , Glomérulos Renales/metabolismo , Túbulos Renales/metabolismo , Túbulos Renales/patología , Podocitos/metabolismo , Podocitos/patología
15.
Clin Exp Pharmacol Physiol ; 46(5): 456-464, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-30811624

RESUMEN

Glomerular crescent formation is a hallmark of rapidly progressive forms of glomerulonephritis. Thrombosis and macrophage infiltration are features of crescent formation in human and experimental kidney disease. Protease-activated receptor-2 (PAR-2) is a G-protein coupled receptor that links coagulation and inflammation. This study investigated whether pharmacological inhibition of PAR-2 can suppress glomerular crescent formation in rat nephrotoxic serum nephritis (NTN). Disease was induced in Wistar Kyoto rats by immunisation with sheep IgG followed by administration of sheep nephrotoxic serum. Rats (n = 8/group) received the PAR-2 antagonist (GB88, 10 mg/kg/p.o.), vehicle or no treatment starting 3 days before nephrotoxic serum injection and continuing until day 14. Vehicle and untreated rats developed thrombosis and macrophage infiltration in the glomerular tuft and Bowman's space in conjunction with prominent crescent formation. Activation of JNK signalling and proliferation in parietal epithelial cells was associated with crescent formation. GB88 treatment significantly reduced crescent formation with a substantial reduction in glomerular thrombosis, reduced macrophage infiltration in Bowman's space, and reduced activation of parietal epithelial cells. However, GB88 did not protect against the development of proteinuria, renal function impairment, inflammation or tubular cell damage in the NTN model. In conclusion, PAR-2 plays a specific role in glomerular crescent formation by promoting glomerular thrombosis, macrophage accumulation in Bowman's space and activation of parietal epithelial cells.


Asunto(s)
Glomérulos Renales/efectos de los fármacos , Glomérulos Renales/patología , Nefritis/tratamiento farmacológico , Receptor PAR-2/antagonistas & inhibidores , Animales , Modelos Animales de Enfermedad , Glomérulos Renales/metabolismo , Masculino , Nefritis/metabolismo , Nefritis/patología , Oligopéptidos/farmacología , Oligopéptidos/uso terapéutico , Ratas , Ratas Wistar
16.
Nephron ; 142(1): 71-81, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30799414

RESUMEN

BACKGROUND/AIMS: Calcineurin inhibitors (CNIs) such as cyclosporine A (CsA) and tacrolimus are immunosuppressants that are frequently used as "key drugs" in the fields of transplantation, steroid resistance, refractory nephrotic syndrome, and autoimmune disease. However, long-term CNI use causes nephrotoxicity, the features of which are arteriolar hyalinosis, tubular atrophy, striped interstitial fibrosis, and focal segmental glomerulosclerosis (FSGS). We investigated whether FSGS in CNI-induced nephrotoxicity is associated with CD44-positive glomerular parietal epithelial cells (PECs), which play a significant role in experimental and human FSGS pathogenesis. METHODS: We utilized the mouse model of CsA-induced nephrotoxicity, as well as controls. Histopathological and functional data were sequentially collected from 1 to 25 weeks after CsA injection. Glomerular expression of CD44 was immunohistochemically evaluated, as were markers for glomerular podocytes and PECs. RESULTS: Glomerular CD44 expression occurred from 2 weeks after CsA injection and gradually increased in the CsA group. CD44-positive glomerular cells showed coexpression of claudin-1 (PEC marker) but not of synaptopodin (podocyte marker). From 20 weeks after CsA injection, the cells formed a bridge between Bowman's capsule and the capillary tuft. These features are compatible with those of activated PECs, in which increased foot process effacement leads to bridge formation, and subsequently to an increase in glomerulosclerosis and a decrease in the expression of podocyte markers from 20 weeks. CONCLUSION: CD44-positive (activated) PECs reflect extremely early podocyte injury in the progression of FSGS formation and may be a good marker for early detection of CNI-induced nephrotoxicity.


Asunto(s)
Inhibidores de la Calcineurina/farmacología , Receptores de Hialuranos/metabolismo , Glomérulos Renales/metabolismo , Riñón/efectos de los fármacos , Animales , Ciclosporina/toxicidad , Células Epiteliales/metabolismo , Humanos , Glomérulos Renales/efectos de los fármacos , Masculino , Ratones , Ratones Endogámicos ICR , Modelos Animales
17.
Am J Physiol Renal Physiol ; 315(5): F1449-F1464, 2018 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-30019931

RESUMEN

Under certain circumstances, podocytes can be partially replaced following their loss in disease. The inability of podocytes to proliferate suggests that replacement derives from other cell types. Because neural/glial antigen 2 (NG2)-expressing cells can serve as progenitors in other organs and because herein we showed increased NG2 staining in podocytes following their loss in experimental focal segmental glomerulosclerosis, we used lineage tracing in NG2-CreER tdTomato mice to test the hypothesis that partial podocyte replacement might derive from this cell population. The percentage of glomeruli with red fluorescence protein (RFP)-labeled NG2 cells increased following podocyte depletion, which was augmented by enalapril. However, BrdU was not detected in RFP-labeled cells, consistent with the migration of these cells to the glomerulus. Within glomeruli, RFP-labeled cells did not coexpress podocyte proteins (p57, synaptopodin, nephrin, or podocin) but did coexpress markers for mesangial (α8 integrin, PDGFß receptor) and parietal epithelial cells (PAX8, src-suppressed C-kinase substrate). These results suggest that following podocyte depletion, cells of NG2 lineage do not serve as adult podocyte progenitors but have the ability to transdifferentiate to mesangial and parietal epithelial cell fates.


Asunto(s)
Antígenos/metabolismo , Linaje de la Célula , Proliferación Celular , Transdiferenciación Celular , Glomeruloesclerosis Focal y Segmentaria/patología , Glomérulos Renales/patología , Podocitos/patología , Proteoglicanos/metabolismo , Regeneración , Proteínas de Anclaje a la Quinasa A/metabolismo , Animales , Antígenos/genética , Biomarcadores/metabolismo , Proteínas de Ciclo Celular/metabolismo , Modelos Animales de Enfermedad , Glomeruloesclerosis Focal y Segmentaria/genética , Glomeruloesclerosis Focal y Segmentaria/metabolismo , Cadenas alfa de Integrinas/metabolismo , Glomérulos Renales/metabolismo , Ratones , Ratones Transgénicos , Factor de Transcripción PAX8/metabolismo , Fenotipo , Podocitos/metabolismo , Proteoglicanos/genética , Receptor beta de Factor de Crecimiento Derivado de Plaquetas/metabolismo
18.
Cell Tissue Res ; 369(1): 237-244, 2017 07.
Artículo en Inglés | MEDLINE | ID: mdl-28361304

RESUMEN

In normal glomeruli, parietal epithelial cells (PECs) line the inside of Bowman's capsule and form an inconspicuous sheet of flat epithelial cells in continuity with the proximal tubular epithelial cells (PTECs) at the urinary pole and with the podocytes at the vascular pole. PECs, PTECs and podocytes have a common mesenchymal origin and are the result of divergent differentiation during embryogenesis. Podocytes and PTECs are highly differentiated cells with well-established functions pertaining to the maintenance of the filtration barrier and transport, respectively. For PECs, no specific function other than a structural one has been known until recently. Possible important functions for PECs in the fate of the glomerulus in glomerular disease have now become apparent: (1) PECs may be involved in the replacement of lost podocytes; (2) PECs form the basis of extracapillary proliferative lesions and subsequent sclerosis in glomerular disease. In addition to the acknowledgement that PECs are crucial in glomerular disease, knowledge has been gained regarding the molecular processes driving the phenotypic changes and behavior of PECs. Understanding these molecular processes is important for the development of specific therapeutic approaches aimed at either stimulation of the regenerative function of PECs or inhibition of the pro-sclerotic action of PECs. In this review, we discuss recent advances pertaining to the role of PECs in glomerular regeneration and disease and address the major molecular processes involved.


Asunto(s)
Cápsula Glomerular , Enfermedades Renales , Túbulos Renales Proximales , Podocitos , Animales , Cápsula Glomerular/metabolismo , Cápsula Glomerular/patología , Cápsula Glomerular/fisiopatología , Humanos , Enfermedades Renales/metabolismo , Enfermedades Renales/patología , Enfermedades Renales/fisiopatología , Túbulos Renales Proximales/metabolismo , Túbulos Renales Proximales/patología , Túbulos Renales Proximales/fisiopatología , Podocitos/metabolismo , Podocitos/patología
19.
Aging (Albany NY) ; 9(2): 524-546, 2017 Feb 17.
Artículo en Inglés | MEDLINE | ID: mdl-28222042

RESUMEN

Advanced age portends a poorer prognosis in FSGS. To understand the impact of age on glomerular podocytes and parietal epithelial cells (PECs), experimental FSGS was induced in 3m-old mice (20-year old human age) and 27m-old mice (78-year old human age) by abruptly depleting podocytes with a cytopathic anti-podocyte antibody. Despite similar binding of the disease-inducing antibody, podocyte density was lower in aged FSGS mice compared to young FSGS mice. Activated PEC density was higher in aged versus young FSGS mice, as was the percentage of total activated PECs. Additionally, the percentage of glomeruli containing PECs with evidence of phosphorylated ERK and EMT was higher in aged FSGS mice. Extracellular matrix, measured by collagen IV and silver staining, was higher in aged FSGS mice along Bowman's capsule. However, collagen IV accumulation in the glomerular tufts alone and in glomeruli with both tuft and Bowman's capsule accumulation were similar in young FSGS and aged FSGS mice. Thus, the major difference in collagen IV staining in FSGS was along Bowman's capsule in aged mice. The significant differences in podocytes, PECs and extracellular matrix accumulation between young mice and old mice with FSGS might explain the differences in outcomes in FSGS based on age.


Asunto(s)
Envejecimiento/patología , Células Epiteliales/patología , Glomeruloesclerosis Focal y Segmentaria/patología , Glomérulos Renales/patología , Factores de Edad , Envejecimiento/metabolismo , Animales , Cápsula Glomerular/metabolismo , Cápsula Glomerular/patología , Colágeno/metabolismo , Modelos Animales de Enfermedad , Células Epiteliales/metabolismo , Matriz Extracelular/metabolismo , Matriz Extracelular/patología , Glomeruloesclerosis Focal y Segmentaria/metabolismo , Glomérulos Renales/metabolismo , Ratones , Fosforilación , Podocitos/metabolismo , Podocitos/patología
20.
Turk J Med Sci ; 47(6): 1925-1930, 2017 Dec 19.
Artículo en Inglés | MEDLINE | ID: mdl-29307170

RESUMEN

Background/aim: The purpose of this study was to observe whether islet transplantation could induce glomerular parietal epithelial cells to express podocyte proteins in a rat model of streptozotocin-induced diabetic nephropathy (DN).Materials and methods: A total of 18 rats were given single injections of streptozotocin to induce a DN model. Eight weeks after the modeling, successfully established DN rats were divided into three groups: an untreated group (DN group), an islet-transplanted group (IT group), and an insulin group (IN group). The islets cells were isolated from donor rats and surgically transplanted from under the kidney capsule in the IT group. Four weeks after treatment, pathological changes in the kidney were observed by pathological staining and electron microscopy. Immunohistochemical staining for PAX-2, Ki-67, and synaptopodin was performed to evaluate cell proliferation in the kidney tissues.Results: After 4 weeks of treatment, islet transplantation significantly alleviated damage to the podocytes and increased the number of glomerular transition cells compared to the DN and IN groups, which were defined as cells that double-stained for PAX-2 and synaptopodin in membranous nephropathy. The results of HE staining, PAS staining, and electron microscopy detection also showed that pathological changes were alleviated after islet transplantation.Conclusion: IT restored the glomerular filtration barrier based on the regeneration of podocytes in the DN model rats, and this may provide a promising clinical therapeutic strategy for human diabetes mellitus.


Asunto(s)
Nefropatías Diabéticas/terapia , Trasplante de Islotes Pancreáticos , Podocitos , Regeneración/fisiología , Animales , Diabetes Mellitus Experimental/terapia , Masculino , Podocitos/citología , Podocitos/fisiología , Ratas , Ratas Sprague-Dawley
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA