Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
1.
Kidney Int ; 102(5): 1000-1012, 2022 11.
Artículo en Inglés | MEDLINE | ID: mdl-35870643

RESUMEN

Dysregulated extracellular matrix is the hallmark of fibrosis, and it has a profound impact on kidney function in disease. Furthermore, perturbation of matrix homeostasis is a feature of aging and is associated with declining kidney function. Understanding these dynamic processes, in the hope of developing therapies to combat matrix dysregulation, requires the integration of data acquired by both well-established and novel technologies. Owing to its complexity, the extracellular proteome, or matrisome, still holds many secrets and has great potential for the identification of clinical biomarkers and drug targets. The molecular resolution of matrix composition during aging and disease has been illuminated by cutting-edge mass spectrometry-based proteomics in recent years, but there remain key questions about the mechanisms that drive altered matrix composition. Basement membrane components are particularly important in the context of kidney function; and data from proteomic studies suggest that switches between basement membrane and interstitial matrix proteins are likely to contribute to organ dysfunction during aging and disease. Understanding the impact of such changes on physical properties of the matrix, and the subsequent cellular response to altered stiffness and viscoelasticity, is of critical importance. Likewise, the comparison of proteomic data sets from multiple organs is required to identify common matrix biomarkers and shared pathways for therapeutic intervention. Coupled with single-cell transcriptomics, there is the potential to identify the cellular origin of matrix changes, which could enable cell-targeted therapy. This review provides a contemporary perspective of the complex kidney matrisome and draws comparison to altered matrix in heart and liver disease.


Asunto(s)
Proteoma , Proteómica , Proteoma/metabolismo , Proteínas de la Matriz Extracelular/metabolismo , Matriz Extracelular/metabolismo , Riñón/metabolismo , Biomarcadores/metabolismo
2.
Kidney Int ; 101(3): 527-540, 2022 03.
Artículo en Inglés | MEDLINE | ID: mdl-34774562

RESUMEN

Nephrotic syndrome is characterized by severe proteinuria, hypoalbuminaemia, edema and hyperlipidaemia. Genetic studies of nephrotic syndrome have led to the identification of proteins playing a crucial role in slit diaphragm signaling, regulation of actin cytoskeleton dynamics and cell-matrix interactions. The laminin α5 chain is essential for embryonic development and, in association with laminin ß2 and laminin γ1, is a major component of the glomerular basement membrane, a critical component of the glomerular filtration barrier. Mutations in LAMA5 were recently identified in children with nephrotic syndrome. Here, we have identified a novel missense mutation (E884G) in the uncharacterized L4a domain of LAMA5 where homozygous mice develop nephrotic syndrome with severe proteinuria with histological and ultrastructural changes in the glomerulus mimicking the progression seen in most patients. The levels of LAMA5 are reduced in vivo and the assembly of the laminin 521 heterotrimer significantly reduced in vitro. Proteomic analysis of the glomerular extracellular fraction revealed changes in the matrix composition. Importantly, the genetic background of the mice had a significant effect on aspects of disease progression from proteinuria to changes in podocyte morphology. Thus, our novel model will provide insights into pathologic mechanisms of nephrotic syndrome and pathways that influence the response to a dysfunctional glomerular basement membrane that may be important in a range of kidney diseases.


Asunto(s)
Síndrome Nefrótico , Animales , Antecedentes Genéticos , Membrana Basal Glomerular/patología , Humanos , Ratones , Mutación , Síndrome Nefrótico/patología , Mutación Puntual , Proteinuria/genética , Proteinuria/metabolismo , Proteómica
3.
J Am Soc Nephrol ; 32(7): 1713-1732, 2021 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-34049963

RESUMEN

BACKGROUND: Accumulation of extracellular matrix in organs and tissues is a feature of both aging and disease. In the kidney, glomerulosclerosis and tubulointerstitial fibrosis accompany the decline in function, which current therapies cannot address, leading to organ failure. Although histologic and ultrastructural patterns of excess matrix form the basis of human disease classifications, a comprehensive molecular resolution of abnormal matrix is lacking. METHODS: Using mass spectrometry-based proteomics, we resolved matrix composition over age in mouse models of kidney disease. We compared the changes in mice with a global characterization of human kidneymatrix during aging and to existing kidney disease datasets to identify common molecular features. RESULTS: Ultrastructural changes in basement membranes are associated with altered cell adhesion and metabolic processes and with distinct matrix proteomes during aging and kidney disease progression in mice. Within the altered matrix, basement membrane components (laminins, type IV collagen, type XVIII collagen) were reduced and interstitial matrix proteins (collagens I, III, VI, and XV; fibrinogens; and nephronectin) were increased, a pattern also seen in human kidney aging. Indeed, this signature of matrix proteins was consistently modulated across all age and disease comparisons, and the increase in interstitial matrix was also observed in human kidney disease datasets. CONCLUSIONS: This study provides deep molecular resolution of matrix accumulation in kidney aging and disease, and identifies a common signature of proteins that provides insight into mechanisms of response to kidney injury and repair.

4.
Pediatr Nephrol ; 35(5): 733-742, 2020 05.
Artículo en Inglés | MEDLINE | ID: mdl-31044288

RESUMEN

Alport syndrome is caused by mutations in the genes COL4A3, COL4A4 or COL4A5 and is characterised by progressive glomerular disease, sensorineural hearing loss and ocular defects. Occurring in less than 1:5000, Alport syndrome is a rare genetic disorder but still accounts for > 1% of the prevalent population receiving renal replacement therapy. There is also increasing awareness about the risk of chronic kidney disease in individuals with heterozygous mutations in Alport syndrome genes. The mainstay of current therapy is the use of angiotensin-converting enzyme inhibitors and angiotensin receptor blockers, yet potential new therapies are now entering clinical trials. The 2017 International Workshop on Alport Syndrome in Glasgow was a pre-conference workshop ahead of the 50th anniversary meeting of the European Society for Pediatric Nephrology. It focussed on updates in clinical practice, genetics and basic science and also incorporated patient perspectives. More than 80 international experts including clinicians, geneticists, researchers from academia and industry, and patient representatives took part in panel discussions and breakout groups. This report summarises the workshop proceedings and the relevant contemporary literature. It highlights the unique clinician, patient and researcher collaborations achieved by regular engagement between the groups.


Asunto(s)
Investigación Biomédica/organización & administración , Colaboración Intersectorial , Nefritis Hereditaria/terapia , Participación del Paciente , Enfermedades Raras/terapia , Inhibidores de la Enzima Convertidora de Angiotensina/uso terapéutico , Autoantígenos/genética , Investigación Biomédica/normas , Niño , Ensayos Clínicos como Asunto , Colágeno Tipo IV/genética , Congresos como Asunto , Humanos , Mutación , Nefritis Hereditaria/complicaciones , Nefritis Hereditaria/genética , Nefrología/métodos , Nefrología/organización & administración , Nefrología/normas , Pediatría/métodos , Pediatría/organización & administración , Pediatría/normas , Guías de Práctica Clínica como Asunto , Enfermedades Raras/complicaciones , Enfermedades Raras/genética , Insuficiencia Renal Crónica/genética , Insuficiencia Renal Crónica/prevención & control , Terapia de Reemplazo Renal , Sociedades Médicas , Terapias en Investigación
5.
Kidney Int ; 95(5): 1138-1152, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-30885509

RESUMEN

Mutations in leucine-rich-repeats and immunoglobulin-like-domains 2 (LRIG2) or in heparanase 2 (HPSE2) cause urofacial syndrome, a devastating autosomal recessive disease of functional bladder outlet obstruction. It has been speculated that urofacial syndrome has a neural basis, but it is unknown whether defects in urinary bladder innervation are present. We hypothesized that urofacial syndrome features a peripheral neuropathy of the bladder. Mice with homozygous targeted Lrig2 mutations had urinary defects resembling those found in urofacial syndrome. There was no anatomical blockage of the outflow tract, consistent with a functional bladder outlet obstruction. Transcriptome analysis revealed differential expression of 12 known transcripts in addition to Lrig2, including 8 with established roles in neurobiology. Mice with homozygous mutations in either Lrig2 or Hpse2 had increased nerve density within the body of the urinary bladder and decreased nerve density around the urinary outflow tract. In a sample of 155 children with chronic kidney disease and urinary symptoms, we discovered novel homozygous missense LRIG2 variants that were predicted to be pathogenic in 2 individuals with non-syndromic bladder outlet obstruction. These observations provide evidence that a peripheral neuropathy is central to the pathobiology of functional bladder outlet obstruction in urofacial syndrome, and emphasize the importance of LRIG2 and heparanase 2 for nerve patterning in the urinary tract.


Asunto(s)
Glucuronidasa/genética , Glicoproteínas de Membrana/genética , Enfermedades del Sistema Nervioso Periférico/genética , Obstrucción del Cuello de la Vejiga Urinaria/genética , Vejiga Urinaria/inervación , Enfermedades Urológicas/genética , Animales , Niño , Análisis Mutacional de ADN , Facies , Femenino , Perfilación de la Expresión Génica , Homocigoto , Humanos , Masculino , Ratones , Ratones Noqueados , Mutación Missense , Enfermedades del Sistema Nervioso Periférico/patología , Vejiga Urinaria/patología , Obstrucción del Cuello de la Vejiga Urinaria/patología , Enfermedades Urológicas/patología
6.
J Am Soc Nephrol ; 26(12): 3021-34, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25896609

RESUMEN

Glomerular disease often features altered histologic patterns of extracellular matrix (ECM). Despite this, the potential complexities of the glomerular ECM in both health and disease are poorly understood. To explore whether genetic background and sex determine glomerular ECM composition, we investigated two mouse strains, FVB and B6, using RNA microarrays of isolated glomeruli combined with proteomic glomerular ECM analyses. These studies, undertaken in healthy young adult animals, revealed unique strain- and sex-dependent glomerular ECM signatures, which correlated with variations in levels of albuminuria and known predisposition to progressive nephropathy. Among the variation, we observed changes in netrin 4, fibroblast growth factor 2, tenascin C, collagen 1, meprin 1-α, and meprin 1-ß. Differences in protein abundance were validated by quantitative immunohistochemistry and Western blot analysis, and the collective differences were not explained by mutations in known ECM or glomerular disease genes. Within the distinct signatures, we discovered a core set of structural ECM proteins that form multiple protein-protein interactions and are conserved from mouse to man. Furthermore, we found striking ultrastructural changes in glomerular basement membranes in FVB mice. Pathway analysis of merged transcriptomic and proteomic datasets identified potential ECM regulatory pathways involving inhibition of matrix metalloproteases, liver X receptor/retinoid X receptor, nuclear factor erythroid 2-related factor 2, notch, and cyclin-dependent kinase 5. These pathways may therefore alter ECM and confer susceptibility to disease.


Asunto(s)
Matriz Extracelular/genética , Matriz Extracelular/metabolismo , Enfermedades Renales/genética , Glomérulos Renales/metabolismo , Albuminuria/genética , Albuminuria/metabolismo , Animales , Colágeno Tipo I/genética , Colágeno Tipo I/metabolismo , Quinasa 5 Dependiente de la Ciclina/metabolismo , Matriz Extracelular/ultraestructura , Femenino , Factor 2 de Crecimiento de Fibroblastos/genética , Factor 2 de Crecimiento de Fibroblastos/metabolismo , Predisposición Genética a la Enfermedad , Membrana Basal Glomerular/ultraestructura , Enfermedades Renales/metabolismo , Receptores X del Hígado , Masculino , Metaloproteinasas de la Matriz/metabolismo , Metaloendopeptidasas/genética , Metaloendopeptidasas/metabolismo , Ratones , Ratones Endogámicos , Factor 2 Relacionado con NF-E2/metabolismo , Factores de Crecimiento Nervioso/genética , Factores de Crecimiento Nervioso/metabolismo , Netrinas , Análisis de Secuencia por Matrices de Oligonucleótidos , Receptores Nucleares Huérfanos/metabolismo , ARN/análisis , Factores Sexuales , Transducción de Señal , Tenascina/genética , Tenascina/metabolismo
7.
Pediatr Nephrol ; 30(9): 1459-65, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-25739341

RESUMEN

BACKGROUND: Mutations in podocyte and basement membrane genes are associated with a growing spectrum of glomerular disease affecting adults and children. Investigation of familial cases has helped to build understanding of both normal physiology and disease. METHODS: We investigated a consanguineous family with a wide clinical phenotype of glomerular disease using clinical, histological, and new genetic studies. RESULTS: We report striking variability in severity of nephropathy within an X-linked Alport syndrome (XLAS) family. Four siblings each carried a mutant COL4A5 allele, p.(Gly953Val) and p.(Gly1033Arg). Two boys had signs limited to hematuria and mild/moderate proteinuria. In striking contrast, a sister presented with end-stage renal disease (ESRD) at 8 years of age and an infant brother presented with nephrotic syndrome, progressing to ESRD by 3 years of age. Both were subsequently found to have homozygous variants in MYO1E, p.(Lys118Glu) and p.(Thr876Arg). MYO1E is a gene implicated in focal segmental glomerulosclerosis and it encodes a podocyte-expressed non-muscle myosin. Bioinformatic modeling demonstrated that the collagen IV-alpha3,4,5 extracellular network connected via known protein-protein interactions to intracellular myosin 1E. CONCLUSIONS: COL4A5 and MYO1E mutations may summate to perturb common signaling pathways, resulting in more severe disease than anticipated independently. We suggest screening for MYO1E and other non-COL4 'podocyte gene' mutations in XLAS when clinical nephropathy is more severe than expected for an individual's age and sex.


Asunto(s)
Colágeno Tipo IV/genética , Glomérulos Renales/patología , Miosina Tipo I/genética , Nefritis Hereditaria , Adulto , Niño , Preescolar , Femenino , Humanos , Lactante , Patrón de Herencia/genética , Masculino , Mutación , Nefritis Hereditaria/diagnóstico , Nefritis Hereditaria/genética , Nefritis Hereditaria/fisiopatología , Linaje , Índice de Severidad de la Enfermedad , Hermanos
8.
J Am Soc Nephrol ; 25(5): 939-51, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24436468

RESUMEN

The glomerulus contains unique cellular and extracellular matrix (ECM) components, which are required for intact barrier function. Studies of the cellular components have helped to build understanding of glomerular disease; however, the full composition and regulation of glomerular ECM remains poorly understood. We used mass spectrometry-based proteomics of enriched ECM extracts for a global analysis of human glomerular ECM in vivo and identified a tissue-specific proteome of 144 structural and regulatory ECM proteins. This catalog includes all previously identified glomerular components plus many new and abundant components. Relative protein quantification showed a dominance of collagen IV, collagen I, and laminin isoforms in the glomerular ECM together with abundant collagen VI and TINAGL1. Protein network analysis enabled the creation of a glomerular ECM interactome, which revealed a core of highly connected structural components. More than one half of the glomerular ECM proteome was validated using colocalization studies and data from the Human Protein Atlas. This study yields the greatest number of ECM proteins relative to previous investigations of whole glomerular extracts, highlighting the importance of sample enrichment. It also shows that the composition of glomerular ECM is far more complex than previously appreciated and suggests that many more ECM components may contribute to glomerular development and disease processes. The mass spectrometry proteomics data have been deposited to the ProteomeXchange Consortium with the dataset identifier PXD000456.


Asunto(s)
Proteínas de la Matriz Extracelular/metabolismo , Glomérulos Renales/metabolismo , Proteoma/química , Adulto , Colágeno Tipo VI/química , Proteínas de la Matriz Extracelular/química , Proteínas de la Matriz Extracelular/genética , Proteínas de la Matriz Extracelular/aislamiento & purificación , Ontología de Genes , Humanos , Glomérulos Renales/química , Glomérulos Renales/citología , Lipocalinas/química , Masculino , Espectrometría de Masas , Persona de Mediana Edad , Mapas de Interacción de Proteínas , Proteoma/genética
9.
J Am Soc Nephrol ; 25(5): 953-66, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24436469

RESUMEN

The glomerular basement membrane (GBM) is a specialized extracellular matrix (ECM) compartment within the glomerulus that contains tissue-restricted isoforms of collagen IV and laminin. It is integral to the capillary wall and therefore, functionally linked to glomerular filtration. Although the composition of the GBM has been investigated with global and candidate-based approaches, the relative contributions of glomerular cell types to the production of ECM are not well understood. To characterize specific cellular contributions to the GBM, we used mass spectrometry-based proteomics to analyze ECM isolated from podocytes and glomerular endothelial cells in vitro. These analyses identified cell type-specific differences in ECM composition, indicating distinct contributions to glomerular ECM assembly. Coculture of podocytes and endothelial cells resulted in an altered composition and organization of ECM compared with monoculture ECMs, and electron microscopy revealed basement membrane-like ECM deposition between cocultured cells, suggesting the involvement of cell-cell cross-talk in the production of glomerular ECM. Notably, compared with monoculture ECM proteomes, the coculture ECM proteome better resembled a tissue-derived glomerular ECM dataset, indicating its relevance to GBM in vivo. Protein network analyses revealed a common core of 35 highly connected structural ECM proteins that may be important for glomerular ECM assembly. Overall, these findings show the complexity of the glomerular ECM and suggest that both ECM composition and organization are context-dependent.


Asunto(s)
Proteínas de la Matriz Extracelular/fisiología , Matriz Extracelular/fisiología , Glomérulos Renales/fisiología , Receptor Cross-Talk/fisiología , Células Cultivadas , Técnicas de Cocultivo , Medios de Cultivo Condicionados , Matriz Extracelular/metabolismo , Matriz Extracelular/ultraestructura , Proteínas de la Matriz Extracelular/biosíntesis , Humanos , Glomérulos Renales/metabolismo , Glomérulos Renales/ultraestructura , Fenotipo , Podocitos/fisiología , Mapas de Interacción de Proteínas
10.
Matrix Biol ; 90: 61-78, 2020 08.
Artículo en Inglés | MEDLINE | ID: mdl-32147508

RESUMEN

Cells have evolved mechanisms to sense the composition of their adhesive microenvironment. Although much is known about general mechanisms employed by adhesion receptors to relay signals between the extracellular environment and the cytoskeleton, the nuances of ligand-specific signalling remain undefined. Here, we investigated how glomerular podocytes, and four other basement membrane-associated cell types, respond morphologically to different basement membrane ligands. We defined the composition of the respective adhesion complexes using mass spectrometry-based proteomics. On type IV collagen, all epithelial cell types adopted a round morphology, with a single lamellipodium and large adhesion complexes rich in actin-binding proteins. On laminin (511 or 521), all cell types attached to a similar degree but were polygonal in shape with small adhesion complexes enriched in endocytic and microtubule-binding proteins. Consistent with their distinctive morphologies, cells on type IV collagen exhibited high Rac1 activity, while those on laminin had elevated PKCα. Perturbation of PKCα was able to interchange morphology consistent with a key role for this pathway in matrix ligand-specific signalling. Therefore, this study defines the switchable basement membrane adhesome and highlights two key signalling pathways within the systems that determine distinct cell morphologies. Proteomic data are availableviaProteomeXchange with identifier PXD017913.


Asunto(s)
Membrana Basal/metabolismo , Ganglios Espinales/citología , Laminina/farmacología , Proteómica/métodos , Animales , Línea Celular , Forma de la Célula/efectos de los fármacos , Colágeno Tipo IV/metabolismo , Ganglios Espinales/efectos de los fármacos , Ganglios Espinales/metabolismo , Células HEK293 , Humanos , Integrina alfa3/metabolismo , Ligandos , Espectrometría de Masas , Ratones , Neuronas/citología , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Neuropéptidos/metabolismo , Proteína Quinasa C-alfa/metabolismo , Transducción de Señal , Proteína de Unión al GTP rac1/metabolismo
11.
J Tissue Eng Regen Med ; 12(1): 252-264, 2018 01.
Artículo en Inglés | MEDLINE | ID: mdl-28084682

RESUMEN

An ex vivo experimental strategy that replicates in vivo intestinal development would in theory provide an accessible setting with which to study normal and dysmorphic gut biology. The current authors recently described a system in which mouse embryonic jejunal segments were explanted onto semipermeable platforms and fed with chemically defined serum-free media. Over 3 days in organ culture, explants formed villi and they began to undergo spontaneous peristalsis. As defined in the current study, the wall of the explanted gut failed to form a robust longitudinal smooth muscle (SM) layer as it would do in vivo over the same time period. Given the role of transforming growth factor ß1 (TGFß1) in SM differentiation in other organs, it was hypothesized that exogenous TGFß1 would enhance SM differentiation in these explants. In vivo, TGFß receptors I and II were both detected in embryonic longitudinal jejunal SM cells and, in organ culture, exogenous TGFß1 induced robust differentiation of longitudinal SM. Microarray profiling showed that TGFß1 increased SM specific transcripts in a dose dependent manner. TGFß1 proteins were detected in amniotic fluid at a time when the intestine was physiologically herniated. By analogy with the requirement for exogenous TGFß1 for SM differentiation in organ culture, the TGFß1 protein that was demonstrated to be present in the amniotic fluid may enhance intestinal development when it is physiologically herniated in early gestation. Future studies of embryonic intestinal cultures should include TGFß1 in the defined media to produce a more faithful model of in vivo muscle differentiation. Copyright © 2017 The Authors Journal of Tissue Engineering and Regenerative Medicine Published by John Wiley & Sons, Ltd.


Asunto(s)
Diferenciación Celular/efectos de los fármacos , Yeyuno/embriología , Miocitos del Músculo Liso/citología , Factor de Crecimiento Transformador beta1/farmacología , Animales , Proliferación Celular/efectos de los fármacos , Embrión de Mamíferos/efectos de los fármacos , Humanos , Ratones , Miocitos del Músculo Liso/efectos de los fármacos , ARN Mensajero/genética , ARN Mensajero/metabolismo , Receptores de Factores de Crecimiento Transformadores beta/metabolismo , Transcriptoma/genética
12.
Matrix Biol ; 57-58: 12-28, 2017 01.
Artículo en Inglés | MEDLINE | ID: mdl-27553508

RESUMEN

Basement membranes are formed from condensed networks of extracellular matrix (ECM) proteins. These structures underlie all epithelial, mesothelial and endothelial sheets and provide an essential structural scaffold. Candidate-based investigations have established that predominant components of basement membranes are laminins, collagen type IV, nidogens and heparan sulphate proteoglycans. More recently, global proteomic approaches have been applied to investigate ECM and these analyses confirm tissue-specific ECM proteomes with a high degree of complexity. The proteomes consist of structural as well as regulatory ECM proteins such as proteases and growth factors. This review is focused on the proteomic analysis of basement membranes and illustrates how this approach can be used to build our understanding of ECM regulation in health and disease.


Asunto(s)
Aneurisma de la Aorta Abdominal/genética , Membrana Basal/metabolismo , Proteoglicanos de Heparán Sulfato/metabolismo , Nefritis Lúpica/genética , Mutación , Neoplasias/genética , Agrina/genética , Agrina/metabolismo , Animales , Aneurisma de la Aorta Abdominal/metabolismo , Aneurisma de la Aorta Abdominal/patología , Membrana Basal/química , Membrana Basal/patología , Colágeno Tipo IV/genética , Colágeno Tipo IV/metabolismo , Matriz Extracelular/química , Matriz Extracelular/metabolismo , Matriz Extracelular/patología , Regulación de la Expresión Génica , Proteoglicanos de Heparán Sulfato/química , Humanos , Laminina/genética , Laminina/metabolismo , Nefritis Lúpica/metabolismo , Nefritis Lúpica/patología , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/metabolismo , Neoplasias/metabolismo , Neoplasias/patología , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Proteómica , Transducción de Señal
13.
Sci Rep ; 7(1): 6876, 2017 07 31.
Artículo en Inglés | MEDLINE | ID: mdl-28761153

RESUMEN

Phospholipase A2 receptor (PLA2R) is a member of the mannose receptor family found in podocytes in human kidney. PLA2R is the target of the autoimmune disease, membranous nephropathy, characterised by production of anti-PLA2R autoantibodies which bind to the podocyte. However the function of PLA2R in health and in disease remains unclear. To gain insight into the molecular mechanisms of PLA2R function, we searched for its endogenous binding partners. Proteomic analysis identified annexinA2 as a potential interactor with the extracellular domains of PLA2R. We confirmed that PLA2R binds to annexinA2-S100A10 (A2t) complex with specific high affinity to the S100A10 component. The binding occured within the PLA2R NC3 fragment and was increased in acidic pH. Furthermore Ca2+ promoted the association of the PLA2R-A2t complex with phospholipid membranes in vitro. Within the podocyte, all three proteins were enriched in the plasma membrane and organelle membrane compartments. PLA2R co-localised with S100A10 at the cell surface and in extracellular vesicles. This novel interaction between PLA2R and the A2t complex offers insights into the role of PLA2R in podocytes and how autoantibodies might disrupt PLA2R function. The ability of podocytes to secrete vesicles containing PLA2R provides a route for engagement of PLA2R with the immune system.


Asunto(s)
Anexina A2/metabolismo , Podocitos/metabolismo , Receptores de Fosfolipasa A2/metabolismo , Proteínas S100/metabolismo , Calcio/metabolismo , Línea Celular , Membrana Celular/metabolismo , Vesículas Extracelulares/metabolismo , Humanos , Unión Proteica
14.
Sci Rep ; 6: 35068, 2016 10 11.
Artículo en Inglés | MEDLINE | ID: mdl-27725732

RESUMEN

Glomeruli are highly sophisticated filters and glomerular disease is the leading cause of kidney failure. Morphological change in glomerular podocytes and the underlying basement membrane are frequently observed in disease, irrespective of the underlying molecular etiology. Standard electron microscopy techniques have enabled the identification and classification of glomerular diseases based on two-dimensional information, however complex three-dimensional ultrastructural relationships between cells and their extracellular matrix cannot be easily resolved with this approach. We employed serial block face-scanning electron microscopy to investigate Alport syndrome, the commonest monogenic glomerular disease, and compared findings to other genetic mouse models of glomerular disease (Myo1e-/-, Ptpro-/-). These analyses revealed the evolution of basement membrane and cellular defects through the progression of glomerular injury. Specifically we identified sub-podocyte expansions of the basement membrane with both cellular and matrix gene defects and found a corresponding reduction in podocyte foot process number. Furthermore, we discovered novel podocyte protrusions invading into the glomerular basement membrane in disease and these occurred frequently in expanded regions of basement membrane. These findings provide new insights into mechanisms of glomerular barrier dysfunction and suggest that common cell-matrix-adhesion pathways are involved in the progression of disease regardless of the primary insult.


Asunto(s)
Enfermedades Renales/diagnóstico por imagen , Microscopía Electrónica de Rastreo/métodos , Nefritis Hereditaria/diagnóstico por imagen , Animales , Modelos Animales de Enfermedad , Técnicas de Inactivación de Genes , Membrana Basal Glomerular/diagnóstico por imagen , Humanos , Imagenología Tridimensional/métodos , Enfermedades Renales/genética , Ratones , Miosina Tipo I , Miosinas/genética , Nefritis Hereditaria/etiología , Proteínas Tirosina Fosfatasas Clase 3 Similares a Receptores/genética
15.
Artículo en Inglés | MEDLINE | ID: mdl-25352829

RESUMEN

Podocytes are specialized epithelial cells that cover the outer surfaces of glomerular capillaries. Unique cell junctions, known as slit diaphragms, which feature nephrin and Neph family proteins in addition to components of adherens, tight, and gap junctions, connect adjacent podocyte foot processes. Single gene disorders affecting the slit diaphragm result in nephrotic syndrome in humans, characterized by massive loss of protein across the capillary wall. In addition to specialized cell junctions, interconnecting podocytes also adhere to the glomerular basement membrane (GBM) of the capillary wall. The GBM is a dense network of secreted, extracellular matrix (ECM) components and contains tissue-restricted isoforms of collagen IV and laminin in addition to other structural proteins and ECM regulators such as proteases and growth factors. The specialized niche of the GBM provides a scaffold for endothelial cells and podocytes to support their unique functions and human genetic mutations in GBM components lead to renal failure, thus highlighting the importance of cell-matrix interactions in the glomerulus. Cells adhere to ECM via adhesion receptors, including integrins, syndecans, and dystroglycan and in particular the integrin heterodimer α3ß1 is required to maintain barrier integrity. Therefore, the sophisticated function of glomerular filtration relies on podocyte adhesion both at cell junctions and at the interface with the ECM. In health, the podocyte coordinates signals from cell junctions and cell-matrix interactions, in response to environmental cues in order to regulate filtration and as our understanding of mechanisms that control cell adhesion in the glomerulus develops, then insight into the effects of disease will improve. The ultimate goal will be to develop targeted therapies to prevent or repair defects in the filtration barrier and to restore glomerular function.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA