Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 40
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
J Pathol ; 264(2): 212-227, 2024 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-39177649

RESUMO

WT1 encodes a podocyte transcription factor whose variants can cause an untreatable glomerular disease in early childhood. Although WT1 regulates many podocyte genes, it is poorly understood which of them are initiators in disease and how they subsequently influence other cell-types in the glomerulus. We hypothesised that this could be resolved using single-cell RNA sequencing (scRNA-seq) and ligand-receptor analysis to profile glomerular cell-cell communication during the early stages of disease in mice harbouring an orthologous human mutation in WT1 (Wt1R394W/+). Podocytes were the most dysregulated cell-type in the early stages of Wt1R394W/+ disease, with disrupted angiogenic signalling between podocytes and the endothelium, including the significant downregulation of transcripts for the vascular factors Vegfa and Nrp1. These signalling changes preceded glomerular endothelial cell loss in advancing disease, a feature also observed in biopsy samples from human WT1 glomerulopathies. Addition of conditioned medium from murine Wt1R394W/+ primary podocytes to wild-type glomerular endothelial cells resulted in impaired endothelial looping and reduced vascular complexity. Despite the loss of key angiogenic molecules in Wt1R394W/+ podocytes, the pro-vascular molecule adrenomedullin was upregulated in Wt1R394W/+ podocytes and plasma and its further administration was able to rescue the impaired looping observed when glomerular endothelium was exposed to Wt1R394W/+ podocyte medium. In comparative analyses, adrenomedullin upregulation was part of a common injury signature across multiple murine and human glomerular disease datasets, whilst other gene changes were unique to WT1 disease. Collectively, our study describes a novel role for altered angiogenic signalling in the initiation of WT1 glomerulopathy. We also identify adrenomedullin as a proangiogenic factor, which despite being upregulated in early injury, offers an insufficient protective response due to the wider milieu of dampened vascular signalling that results in endothelial cell loss in later disease. © 2024 The Author(s). The Journal of Pathology published by John Wiley & Sons Ltd on behalf of The Pathological Society of Great Britain and Ireland.


Assuntos
Glomérulos Renais , Podócitos , Transdução de Sinais , Análise de Célula Única , Transcriptoma , Proteínas WT1 , Animais , Podócitos/metabolismo , Podócitos/patologia , Proteínas WT1/metabolismo , Proteínas WT1/genética , Humanos , Glomérulos Renais/metabolismo , Glomérulos Renais/patologia , Glomérulos Renais/irrigação sanguínea , Células Endoteliais/metabolismo , Células Endoteliais/patologia , Camundongos , Neovascularização Patológica/genética , Neovascularização Patológica/metabolismo , Modelos Animais de Doenças , Mutação , Nefropatias/genética , Nefropatias/metabolismo , Nefropatias/patologia , Adrenomedulina/genética , Adrenomedulina/metabolismo , Fator A de Crescimento do Endotélio Vascular/metabolismo , Fator A de Crescimento do Endotélio Vascular/genética , Comunicação Celular , Células Cultivadas
2.
Nat Commun ; 15(1): 1452, 2024 Feb 16.
Artigo em Inglês | MEDLINE | ID: mdl-38365780

RESUMO

The development of vascular networks in microfluidic chips is crucial for the long-term culture of three-dimensional cell aggregates such as spheroids, organoids, tumoroids, or tissue explants. Despite rapid advancement in microvascular network systems and organoid technologies, vascularizing organoids-on-chips remains a challenge in tissue engineering. Most existing microfluidic devices poorly reflect the complexity of in vivo flows and require complex technical set-ups. Considering these constraints, we develop a platform to establish and monitor the formation of endothelial networks around mesenchymal and pancreatic islet spheroids, as well as blood vessel organoids generated from pluripotent stem cells, cultured for up to 30 days on-chip. We show that these networks establish functional connections with the endothelium-rich spheroids and vascular organoids, as they successfully provide intravascular perfusion to these structures. We find that organoid growth, maturation, and function are enhanced when cultured on-chip using our vascularization method. This microphysiological system represents a viable organ-on-chip model to vascularize diverse biological 3D tissues and sets the stage to establish organoid perfusions using advanced microfluidics.


Assuntos
Ilhotas Pancreáticas , Microfluídica , Organoides , Engenharia Tecidual/métodos , Endotélio , Ilhotas Pancreáticas/irrigação sanguínea
3.
bioRxiv ; 2023 Mar 29.
Artigo em Inglês | MEDLINE | ID: mdl-37034801

RESUMO

Background: The kidney vasculature is exquisitely structured to orchestrate renal function. Structural profiling of the vasculature in intact rodent kidneys, has provided insights into renal haemodynamics and oxygenation, but has never been extended to the human kidney beyond a few vascular generations. We hypothesised that synchrotron-based imaging of a human kidney would enable assessment of vasculature across the whole organ. Methods: An intact kidney from a 63-year-old male was scanned using hierarchical phase-contrast tomography (HiP-CT), followed by semi-automated vessel segmentation and quantitative analysis. These data were compared to published micro-CT data of whole rat kidney. Results: The intact human kidney vascular network was imaged with HiP-CT at 25 µm voxels, representing a 20-fold increase in resolution compared to clinical CT scanners. Our comparative quantitative analysis revealed the number of vessel generations, vascular asymmetry and a structural organisation optimised for minimal resistance to flow, are conserved between species, whereas the normalised radii are not. We further demonstrate regional heterogeneity in vessel geometry between renal cortex, medulla, and hilum, showing how the distance between vessels provides a structural basis for renal oxygenation and hypoxia. Conclusions: Through the application of HiP-CT, we have provided the first quantification of the human renal arterial network, with a resolution comparable to that of light microscopy yet at a scale several orders of magnitude larger than that of a renal punch biopsy. Our findings bridge anatomical scales, profiling blood vessels across the intact human kidney, with implications for renal physiology, biophysical modelling, and tissue engineering.

4.
Sci Rep ; 12(1): 12172, 2022 07 16.
Artigo em Inglês | MEDLINE | ID: mdl-35842494

RESUMO

Plasma ultrafiltration in the kidney occurs across glomerular capillaries, which are surrounded by epithelial cells called podocytes. Podocytes have a unique shape maintained by a complex cytoskeleton, which becomes disrupted in glomerular disease resulting in defective filtration and albuminuria. Lack of endogenous thymosin ß4 (TB4), an actin sequestering peptide, exacerbates glomerular injury and disrupts the organisation of the podocyte actin cytoskeleton, however, the potential of exogenous TB4 therapy to improve podocyte injury is unknown. Here, we have used Adriamycin (ADR), a toxin which injures podocytes and damages the glomerular filtration barrier leading to albuminuria in mice. Through interrogating single-cell RNA-sequencing data of isolated glomeruli we demonstrate that ADR injury results in reduced levels of podocyte TB4. Administration of an adeno-associated viral vector encoding TB4 increased the circulating level of TB4 and prevented ADR-induced podocyte loss and albuminuria. ADR injury was associated with disorganisation of the podocyte actin cytoskeleton in vitro, which was ameliorated by treatment with exogenous TB4. Collectively, we propose that systemic gene therapy with TB4 prevents podocyte injury and maintains glomerular filtration via protection of the podocyte cytoskeleton thus presenting a novel treatment strategy for glomerular disease.


Assuntos
Nefropatias , Podócitos , Albuminúria , Animais , Células Cultivadas , Doxorrubicina , Terapia Genética , Glomérulos Renais , Camundongos , Timosina
5.
Elife ; 112022 01 25.
Artigo em Inglês | MEDLINE | ID: mdl-35076391

RESUMO

Basement membranes (BMs) are complex macromolecular networks underlying all continuous layers of cells. Essential components include collagen IV and laminins, which are affected by human genetic variants leading to a range of debilitating conditions including kidney, muscle, and cerebrovascular phenotypes. We investigated the dynamics of BM assembly in human pluripotent stem cell-derived kidney organoids. We resolved their global BM composition and discovered a conserved temporal sequence in BM assembly that paralleled mammalian fetal kidneys. We identified the emergence of key BM isoforms, which were altered by a pathogenic variant in COL4A5. Integrating organoid, fetal, and adult kidney proteomes, we found dynamic regulation of BM composition through development to adulthood, and with single-cell transcriptomic analysis we mapped the cellular origins of BM components. Overall, we define the complex and dynamic nature of kidney organoid BM assembly and provide a platform for understanding its wider relevance in human development and disease.


Assuntos
Membrana Basal/patologia , Membrana Basal/fisiologia , Nefropatias/patologia , Rim/fisiologia , Organoides/fisiologia , Animais , Biópsia , Técnicas de Cultura de Células em Três Dimensões/métodos , Linhagem Celular , Pré-Escolar , Colágeno Tipo IV/genética , Proteínas da Matriz Extracelular/genética , Feminino , Humanos , Rim/patologia , Nefropatias/genética , Masculino , Camundongos , Células-Tronco Pluripotentes/fisiologia , Proteômica/métodos
6.
Sci Rep ; 11(1): 15529, 2021 07 30.
Artigo em Inglês | MEDLINE | ID: mdl-34330963

RESUMO

Diabetes mellitus (DM) is the leading cause of chronic kidney disease and diabetic nephropathy is widely studied. In contrast, the pathobiology of diabetic urinary bladder disease is less understood despite dysfunctional voiding being common in DM. We hypothesised that diabetic cystopathy has a characteristic molecular signature. We therefore studied bladders of hyperglycaemic and polyuric rats with streptozotocin (STZ)-induced DM. Sixteen weeks after induction of DM, as assessed by RNA arrays, wide-ranging changes of gene expression occurred in DM bladders over and above those induced in bladders of non-hyperglycaemic rats with sucrose-induced polyuria. The altered transcripts included those coding for extracellular matrix regulators and neural molecules. Changes in key genes deregulated in DM rat bladders were also detected in db/db mouse bladders. In DM rat bladders there was reduced birefringent collagen between detrusor muscle bundles, and atomic force microscopy showed a significant reduction in tissue stiffness; neither change was found in bladders of sucrose-treated rats. Thus, altered extracellular matrix with reduced tissue rigidity may contribute to voiding dysfunction in people with long-term DM. These results serve as an informative stepping stone towards understanding the complex pathobiology of diabetic cystopathy.


Assuntos
Diabetes Mellitus Experimental/metabolismo , Bexiga Urinária/metabolismo , Animais , Ensaio de Imunoadsorção Enzimática , Masculino , Microscopia de Força Atômica , Análise de Sequência com Séries de Oligonucleotídeos , Ratos , Ratos Wistar , Transcriptoma/genética , Transcriptoma/fisiologia
7.
J Am Soc Nephrol ; 31(6): 1178-1190, 2020 06.
Artigo em Inglês | MEDLINE | ID: mdl-32295825

RESUMO

The kidney contains a network of lymphatic vessels that clear fluid, small molecules, and cells from the renal interstitium. Through modulating immune responses and via crosstalk with surrounding renal cells, lymphatic vessels have been implicated in the progression and maintenance of kidney disease. In this Review, we provide an overview of the development, structure, and function of lymphatic vessels in the healthy adult kidney. We then highlight the contributions of lymphatic vessels to multiple forms of renal pathology, emphasizing CKD, transplant rejection, and polycystic kidney disease and discuss strategies to target renal lymphatics using genetic and pharmacologic approaches. Overall, we argue the case for lymphatics playing a fundamental role in renal physiology and pathology and treatments modulating these vessels having therapeutic potential across the spectrum of kidney disease.


Assuntos
Nefropatias/etiologia , Vasos Linfáticos/fisiologia , Imunidade Adaptativa , Rejeição de Enxerto , Humanos , Nefropatias/fisiopatologia , Transplante de Rim/efeitos adversos , Linfa/fisiologia , Linfangiogênese , Vasos Linfáticos/anatomia & histologia , Vasos Linfáticos/citologia , Doenças Renais Policísticas/fisiopatologia , Fator A de Crescimento do Endotélio Vascular/fisiologia
8.
Cell Signal ; 72: 109624, 2020 08.
Artigo em Inglês | MEDLINE | ID: mdl-32243961

RESUMO

Whereas targeting the cyst epithelium and its molecular machinery has been the prevailing clinical strategy for polycystic kidney disease, the endothelium, including blood vasculature and lymphatics, is emerging as an important player in this disorder. In this Review, we provide an overview of the structural and functional alterations to blood vasculature and lymphatic vessels in the polycystic kidney. We also discuss evidence for vascular endothelial growth factor signalling, otherwise critical for endothelial cell development and maintenance, as being a fundamental molecular pathway in polycystic kidney disease and a potential therapeutic target for modulating cyst expansion.


Assuntos
Comunicação Celular , Células Endoteliais/patologia , Células Epiteliais/patologia , Doenças Renais Policísticas/patologia , Transdução de Sinais , Fator A de Crescimento do Endotélio Vascular/metabolismo , Animais , Humanos
9.
Elife ; 82019 12 06.
Artigo em Inglês | MEDLINE | ID: mdl-31808745

RESUMO

Heterogeneity of lymphatic vessels during embryogenesis is critical for organ-specific lymphatic function. Little is known about lymphatics in the developing kidney, despite their established roles in pathology of the mature organ. We performed three-dimensional imaging to characterize lymphatic vessel formation in the mammalian embryonic kidney at single-cell resolution. In mouse, we visually and quantitatively assessed the development of kidney lymphatic vessels, remodeling from a ring-like anastomosis under the nascent renal pelvis; a site of VEGF-C expression, to form a patent vascular plexus. We identified a heterogenous population of lymphatic endothelial cell clusters in mouse and human embryonic kidneys. Exogenous VEGF-C expanded the lymphatic population in explanted mouse embryonic kidneys. Finally, we characterized complex kidney lymphatic abnormalities in a genetic mouse model of polycystic kidney disease. Our study provides novel insights into the development of kidney lymphatic vasculature; a system which likely has fundamental roles in renal development, physiology and disease.


Assuntos
Rim/metabolismo , Linfangiogênese/genética , Vasos Linfáticos/metabolismo , Doenças Renais Policísticas/genética , Animais , Regulação da Expressão Gênica no Desenvolvimento , Heterogeneidade Genética , Humanos , Rim/embriologia , Cinética , Vasos Linfáticos/embriologia , Mamíferos/embriologia , Mamíferos/genética , Mamíferos/metabolismo , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Doenças Renais Policísticas/embriologia , Doenças Renais Policísticas/metabolismo , Análise Espaço-Temporal , Fator C de Crescimento do Endotélio Vascular/genética , Fator C de Crescimento do Endotélio Vascular/metabolismo
10.
Diabetes ; 68(9): 1841-1852, 2019 09.
Artigo em Inglês | MEDLINE | ID: mdl-31217174

RESUMO

Damage to the vasculature is the primary mechanism driving chronic diabetic microvascular complications such as diabetic nephropathy, which manifests as albuminuria. Therefore, treatments that protect the diabetic vasculature have significant therapeutic potential. Soluble neurite outgrowth inhibitor-B (sNogo-B) is a circulating N-terminus isoform of full-length Nogo-B, which plays a key role in vascular remodeling following injury. However, there is currently no information on the role of sNogo-B in the context of diabetic nephropathy. We demonstrate that overexpression of sNogo-B in the circulation ameliorates diabetic kidney disease by reducing albuminuria, hyperfiltration, and abnormal angiogenesis and protecting glomerular capillary structure. Systemic sNogo-B overexpression in diabetic mice also associates with dampening vascular endothelial growth factor-A signaling and reducing endothelial nitric oxide synthase, AKT, and GSK3ß phosphorylation. Furthermore, sNogo-B prevented the impairment of tube formation, which occurred when human endothelial cells were exposed to sera from patients with diabetic kidney disease. Collectively, these studies provide the first evidence that sNogo-B protects the vasculature in diabetes and may represent a novel therapeutic target for diabetic vascular complications.


Assuntos
Capilares/metabolismo , Diabetes Mellitus Experimental/metabolismo , Nefropatias Diabéticas/metabolismo , Células Endoteliais da Veia Umbilical Humana/metabolismo , Glomérulos Renais/irrigação sanguínea , Proteínas Nogo/metabolismo , Angiopoietina-1/metabolismo , Angiopoietina-2/metabolismo , Animais , Diabetes Mellitus Experimental/sangue , Diabetes Mellitus Experimental/genética , Nefropatias Diabéticas/sangue , Nefropatias Diabéticas/genética , Humanos , Glomérulos Renais/metabolismo , Camundongos , Neovascularização Patológica/genética , Neovascularização Patológica/metabolismo , Óxido Nítrico Sintase Tipo III/metabolismo , Proteínas Nogo/sangue , Proteínas Nogo/genética , Fosforilação , Proteínas Proto-Oncogênicas c-akt/metabolismo , Fator A de Crescimento do Endotélio Vascular/metabolismo , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/metabolismo
11.
Dis Model Mech ; 10(4): 409-423, 2017 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-28237967

RESUMO

Lung diseases impose a huge economic and health burden worldwide. A key aspect of several adult lung diseases, such as idiopathic pulmonary fibrosis (IPF) and chronic obstructive pulmonary disease (COPD), including emphysema, is aberrant tissue repair, which leads to an accumulation of damage and impaired respiratory function. Currently, there are few effective treatments available for these diseases and their incidence is rising. The planar cell polarity (PCP) pathway is critical for the embryonic development of many organs, including kidney and lung. We have previously shown that perturbation of the PCP pathway impairs tissue morphogenesis, which disrupts the number and shape of epithelial tubes formed within these organs during embryogenesis. However, very little is known about the role of the PCP pathway beyond birth, partly because of the perinatal lethality of many PCP mouse mutant lines. Here, we investigate heterozygous Looptail (Lp) mice, in which a single copy of the core PCP gene, Vangl2, is disrupted. We show that these mice are viable but display severe airspace enlargement and impaired adult lung function. Underlying these defects, we find that Vangl2Lp/+ lungs exhibit altered distribution of actin microfilaments and abnormal regulation of the actin-modifying protein cofilin. In addition, we show that Vangl2Lp/+ lungs exhibit many of the hallmarks of tissue damage, including an altered macrophage population, abnormal elastin deposition and elevated levels of the elastin-modifying enzyme, Mmp12, all of which are observed in emphysema. In vitro, disruption of VANGL2 impairs directed cell migration and reduces the rate of repair following scratch wounding of human alveolar epithelial cells. Moreover, using population data from a birth cohort of young adults, all aged 31, we found evidence of an interactive effect between VANGL2 and smoking on lung function. Finally, we show that PCP genes VANGL2 and SCRIB are significantly downregulated in lung tissue from patients with emphysema. Our data reveal an important novel role for the PCP pathway in adult lung homeostasis and repair and shed new light on the genetic factors which may modify destructive lung diseases such as emphysema.


Assuntos
Envelhecimento/patologia , Polaridade Celular , Homeostase , Pulmão/patologia , Proteínas do Tecido Nervoso/genética , Cicatrização , Células A549 , Citoesqueleto de Actina/metabolismo , Animais , Movimento Celular , Regulação para Baixo/genética , Elastina/metabolismo , Embrião de Mamíferos/patologia , Técnicas de Silenciamento de Genes , Heterozigoto , Humanos , Pulmão/embriologia , Pulmão/fisiopatologia , Macrófagos/patologia , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Camundongos , Modelos Biológicos , Mutação/genética , Fenótipo , Polimorfismo Genético , Doença Pulmonar Obstrutiva Crônica/genética , Fumar/efeitos adversos , Proteínas Supressoras de Tumor/genética , Proteínas Supressoras de Tumor/metabolismo
12.
Kidney Int ; 90(5): 1056-1070, 2016 11.
Artigo em Inglês | MEDLINE | ID: mdl-27575556

RESUMO

Glomerular disease is characterized by morphologic changes in podocyte cells accompanied by inflammation and fibrosis. Thymosin ß4 regulates cell morphology, inflammation, and fibrosis in several organs and administration of exogenous thymosin ß4 improves animal models of unilateral ureteral obstruction and diabetic nephropathy. However, the role of endogenous thymosin ß4 in the kidney is unknown. We demonstrate that thymosin ß4 is expressed prominently in podocytes of developing and adult mouse glomeruli. Global loss of thymosin ß4 did not affect healthy glomeruli, but accelerated the severity of immune-mediated nephrotoxic nephritis with worse renal function, periglomerular inflammation, and fibrosis. Lack of thymosin ß4 in nephrotoxic nephritis led to the redistribution of podocytes from the glomerular tuft toward the Bowman capsule suggesting a role for thymosin ß4 in the migration of these cells. Thymosin ß4 knockdown in cultured podocytes also increased migration in a wound-healing assay, accompanied by F-actin rearrangement and increased RhoA activity. We propose that endogenous thymosin ß4 is a modifier of glomerular injury, likely having a protective role acting as a brake to slow disease progression.


Assuntos
Glomerulonefrite/metabolismo , Podócitos/metabolismo , Timosina/metabolismo , Animais , Movimento Celular , Células Cultivadas , Citoesqueleto/metabolismo , Fibrose , Glomerulonefrite/patologia , Glomérulos Renais/patologia , Macrófagos , Masculino , Camundongos Endogâmicos C57BL , Camundongos Knockout
13.
Am J Physiol Renal Physiol ; 311(4): F722-F730, 2016 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-27465991

RESUMO

Aging-associated kidney disease is usually considered a degenerative process associated with aging. Recently, it has been shown that animals can produce fructose endogenously, and that this can be a mechanism for causing kidney damage in diabetic nephropathy and in association with recurrent dehydration. We therefore hypothesized that low-level metabolism of endogenous fructose might play a role in aging-associated kidney disease. Wild-type and fructokinase knockout mice were fed a normal diet for 2 yr that had minimal (<5%) fructose content. At the end of 2 yr, wild-type mice showed elevations in systolic blood pressure, mild albuminuria, and glomerular changes with mesangial matrix expansion, variable mesangiolysis, and segmental thrombi. The renal injury was amplified by provision of high-salt diet for 3 wk, as noted by the presence of glomerular hypertrophy, mesangial matrix expansion, and alpha smooth muscle actin expression, and with segmental thrombi. Fructokinase knockout mice were protected from renal injury both at baseline and after high salt intake (3 wk) compared with wild-type mice. This was associated with higher levels of active (phosphorylated serine 1177) endothelial nitric oxide synthase in their kidneys. These studies suggest that aging-associated renal disease might be due to activation of specific metabolic pathways that could theoretically be targeted therapeutically, and raise the hypothesis that aging-associated renal injury may represent a disease process as opposed to normal age-related degeneration.


Assuntos
Envelhecimento/metabolismo , Albuminúria/metabolismo , Frutoquinases/metabolismo , Nefropatias/metabolismo , Rim/metabolismo , Envelhecimento/patologia , Albuminúria/genética , Albuminúria/patologia , Animais , Pressão Sanguínea/fisiologia , Creatinina/sangue , Frutoquinases/genética , Rim/patologia , Nefropatias/genética , Nefropatias/patologia , Lipocalina-2/urina , Camundongos , Camundongos Knockout , Óxido Nítrico Sintase Tipo III/metabolismo , Fosforilação
14.
J Am Soc Nephrol ; 27(1): 69-77, 2016 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-26038530

RESUMO

Polycystic kidney diseases (PKD) are genetic disorders characterized by progressive epithelial cyst growth leading to destruction of normally functioning renal tissue. Current therapies have focused on the cyst epithelium, and little is known about how the blood and lymphatic microvasculature modulates cystogenesis. Hypomorphic Pkd1(nl/nl) mice were examined, showing that cystogenesis was associated with a disorganized pericystic network of vessels expressing platelet/endothelial cell adhesion molecule 1 and vascular endothelial growth factor receptor 3 (VEGFR3). The major ligand for VEGFR3 is VEGFC, and there were lower levels of Vegfc mRNA within the kidneys during the early stages of cystogenesis in 7-day-old Pkd1(nl/nl) mice. Seven-day-old mice were treated with exogenous VEGFC for 2 weeks on the premise that this would remodel both the VEGFR3(+) pericystic vascular network and larger renal lymphatics that may also affect the severity of PKD. Treatment with VEGFC enhanced VEGFR3 phosphorylation in the kidney, normalized the pattern of the pericystic network of vessels, and widened the large lymphatics in Pkd1(nl/nl) mice. These effects were associated with significant reductions in cystic disease, BUN and serum creatinine levels. Furthermore, VEGFC administration reduced M2 macrophage pericystic infiltrate, which has been implicated in the progression of PKD. VEGFC administration also improved cystic disease in Cys1(cpk/cpk) mice, a model of autosomal recessive PKD, leading to a modest but significant increase in lifespan. Overall, this study highlights VEGFC as a potential new treatment for some aspects of PKD, with the possibility for synergy with current epithelially targeted approaches.


Assuntos
Doenças Renais Policísticas/tratamento farmacológico , Fator C de Crescimento do Endotélio Vascular/uso terapêutico , Animais , Camundongos , Doenças Renais Policísticas/etiologia , Fator C de Crescimento do Endotélio Vascular/fisiologia
15.
Clin Sci (Lond) ; 129(12): 1225-36, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26561594

RESUMO

Kidney glomeruli ultrafilter blood to generate urine and they are dysfunctional in a variety of kidney diseases. There are two key vascular growth factor families implicated in glomerular biology and function, namely the vascular endothelial growth factors (VEGFs) and the angiopoietins (Angpt). We present examples showing not only how these molecules help generate and maintain healthy glomeruli but also how they drive disease when their expression is dysregulated. Finally, we review how manipulating VEGF and Angpt signalling may be used to treat glomerular disease.


Assuntos
Angiopoietinas/metabolismo , Taxa de Filtração Glomerular , Nefropatias/metabolismo , Glomérulos Renais/irrigação sanguínea , Glomérulos Renais/metabolismo , Transdução de Sinais , Fatores de Crescimento do Endotélio Vascular/metabolismo , Angiopoietinas/genética , Animais , Nefropatias Diabéticas/metabolismo , Nefropatias Diabéticas/fisiopatologia , Células Endoteliais/metabolismo , Células Epiteliais/metabolismo , Humanos , Nefropatias/genética , Nefropatias/fisiopatologia , Nefropatias/terapia , Glomérulos Renais/fisiopatologia , Fatores de Crescimento do Endotélio Vascular/genética
16.
J Am Soc Nephrol ; 25(1): 33-42, 2014 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-24009238

RESUMO

Vascular growth factors play an important role in maintaining the structure and integrity of the glomerular filtration barrier. In healthy adult glomeruli, the proendothelial survival factors vascular endothelial growth factor-A (VEGF-A) and angiopoietin-1 are constitutively expressed in glomerular podocyte epithelia. We demonstrate that this milieu of vascular growth factors is altered in streptozotocin-induced type 1 diabetic mice, with decreased angiopoietin-1 levels, VEGF-A upregulation, decreased soluble VEGF receptor-1 (VEGFR1), and increased VEGFR2 phosphorylation. This was accompanied by marked albuminuria, nephromegaly, hyperfiltration, glomerular ultrastructural alterations, and aberrant angiogenesis. We subsequently hypothesized that restoration of angiopoietin-1 expression within glomeruli might ameliorate manifestations of early diabetic glomerulopathy. Podocyte-specific inducible repletion of angiopoietin-1 in diabetic mice caused a 70% reduction of albuminuria and prevented diabetes-induced glomerular endothelial cell proliferation; hyperfiltration and renal morphology were unchanged. Furthermore, angiopoietin-1 repletion in diabetic mice increased Tie-2 phosphorylation, elevated soluble VEGFR1, and was paralleled by a decrease in VEGFR2 phosphorylation and increased endothelial nitric oxide synthase Ser(1177) phosphorylation. Diabetes-induced nephrin phosphorylation was also reduced in mice with angiopoietin-1 repletion. In conclusion, targeted angiopoietin-1 therapy shows promise as a renoprotective tool in the early stages of diabetic kidney disease.


Assuntos
Angiopoietina-1/metabolismo , Nefropatias Diabéticas/metabolismo , Nefropatias Diabéticas/terapia , Terapia de Alvo Molecular , Angiopoietina-1/deficiência , Angiopoietina-1/genética , Angiopoietina-2/genética , Angiopoietina-2/metabolismo , Animais , Diabetes Mellitus Experimental/metabolismo , Diabetes Mellitus Experimental/patologia , Diabetes Mellitus Experimental/terapia , Nefropatias Diabéticas/patologia , Humanos , Peptídeos e Proteínas de Sinalização Intracelular/deficiência , Peptídeos e Proteínas de Sinalização Intracelular/genética , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Masculino , Proteínas de Membrana/deficiência , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos CBA , Camundongos Mutantes , Podócitos/metabolismo , Podócitos/patologia , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Transdução de Sinais , Receptor 1 de Fatores de Crescimento do Endotélio Vascular/metabolismo , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/metabolismo
17.
PLoS One ; 8(2): e56273, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23409162

RESUMO

Cardiovascular disease (CVD) is increasingly recognised as a complication of childhood chronic kidney disease (CKD) even in the absence of diabetes and hypertension. We hypothesized that an alteration in angiopoietin-1 and -2, growth factors which regulate endothelial and vascular function could be involved. We report that the endothelial survival factor, angiopoietin-1 is low in children with pre-dialysis CKD whereas the pro-inflammatory angiopoietin-2 is elevated in children on dialysis. In dialysis patients, angiopoietin-2 positively correlated with time on dialysis, systolic blood pressure, and carotid artery intima media thickness. Elevated angiopoietin-2 levels in dialysis versus pre-dialysis CKD patients were also associated with an anti-angiogenic (high soluble VEGFR-1 and low VEGF-A) and pro-inflammatory (high urate, E-selectin, P-selectin and VCAM-1) milieu. Ang-2 was immunodetected in arterial biopsy samples whilst the expression of VEGF-A was significantly downregulated in dialysis patients. Serum urate correlated with angiopoietin-2 levels in dialysis patients and addition of uric acid was able to induce rapid release of angiopoietin-2 from cultured endothelial cells. Thus, angiopoietin-2 is a marker for cardiovascular disease in children on chronic dialysis and may act as an anti-angiogenic and pro-inflammatory effector in this context. The possibility that the release of angiopoietin-2 from endothelia is mediated by urate should be explored further.


Assuntos
Angiopoietina-2/sangue , Doenças Cardiovasculares/complicações , Diálise Renal , Insuficiência Renal Crônica/sangue , Insuficiência Renal Crônica/terapia , Adolescente , Angiopoietina-2/metabolismo , Artérias/metabolismo , Artérias/patologia , Biomarcadores/sangue , Moléculas de Adesão Celular/sangue , Moléculas de Adesão Celular/química , Criança , Feminino , Células Endoteliais da Veia Umbilical Humana/efeitos dos fármacos , Células Endoteliais da Veia Umbilical Humana/metabolismo , Humanos , Masculino , Insuficiência Renal Crônica/patologia , Solubilidade , Fatores de Tempo , Ácido Úrico/farmacologia , Fator A de Crescimento do Endotélio Vascular/sangue , Receptor 1 de Fatores de Crescimento do Endotélio Vascular/sangue
18.
Pediatr Nephrol ; 28(9): 1749-55, 2013 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-22990303

RESUMO

Autosomal dominant polycystic kidney disease (ADPKD) is characterized by the growth of multiple cysts that in many cases result in end-stage renal disease. Current strategies to reduce cyst progression in ADPKD focus on modulating cell turnover, fluid secretion, and vasopressin signalling; but an alternative approach may be to target pathways providing "general support" for cyst growth, such as surrounding blood vessels. This could be achieved by altering the expression of growth factors involved in vascular network formation, such as the vascular endothelial growth factor (VEGF) and angiopoietin families. We highlight the evidence that blood vessels and vascular growth factors play a role in ADPKD progression. Recent experiments manipulating VEGF in ADPKD are described, and we discuss how alternative strategies to manipulate angiogenesis may be used in the future as a novel treatment for ADPKD.


Assuntos
Neovascularização Patológica/patologia , Rim Policístico Autossômico Dominante/patologia , Inibidores da Angiogênese/uso terapêutico , Vasos Sanguíneos/patologia , Criança , Cistos/patologia , Endotélio Vascular/patologia , Humanos , Neovascularização Patológica/tratamento farmacológico , Neovascularização Patológica/genética , Rim Policístico Autossômico Dominante/tratamento farmacológico , Rim Policístico Autossômico Dominante/genética , Transdução de Sinais/fisiologia , Fator A de Crescimento do Endotélio Vascular/fisiologia
19.
J Am Soc Nephrol ; 24(2): 179-89, 2013 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-23138485

RESUMO

Cardiovascular disease begins early in the course of renal decline and is a life-limiting problem in patients with CKD. The increased burden of cardiovascular disease is due, at least in part, to calcification of the vessel wall. The uremic milieu provides a perfect storm of risk factors for accelerated calcification, but elevated calcium and phosphate levels remain key to the initiation and progression of vascular smooth muscle cell calcification in CKD. Vascular calcification is a highly regulated process that involves a complex interplay between promoters and inhibitors of calcification and has many similarities to bone ossification. Here, we discuss current understanding of the process of vascular calcification, focusing specifically on the discrete and synergistic effects of calcium and phosphate in mediating vascular smooth muscle cell apoptosis, osteochondrocytic differentiation, vesicle release, calcification inhibitor expression, senescence, and death. Using our model of intact human vessels, factors initiating vascular calcification in vivo and the role of calcium and phosphate in driving accelerated calcification ex vivo are described. This work allows us to link clinical and basic research into a working theoretical model to explain the pathway of development of vascular calcification in CKD.


Assuntos
Calcinose/patologia , Insuficiência Renal Crônica/patologia , Uremia/patologia , Doenças Vasculares/patologia , Animais , Calcinose/metabolismo , Humanos , Insuficiência Renal Crônica/metabolismo , Uremia/metabolismo , Doenças Vasculares/metabolismo
20.
Nat Rev Nephrol ; 8(4): 244-50, 2012 02 07.
Artigo em Inglês | MEDLINE | ID: mdl-22310952

RESUMO

Chronic kidney disease is characterized by progressive loss of the renal microvasculature, which leads to local areas of hypoxia and induction of profibrotic responses, scarring and deterioration of renal function. Revascularization alone might be sufficient to restore kidney function and regenerate the structure of the diseased kidney. For revascularization to be successful, however, the underlying disease process needs to be halted or alleviated and there must remain a sufficient number of surviving nephron units that can serve as a scaffold for kidney regeneration. This Perspectives article describes how revascularization might be achieved using vascular growth factors or adoptive transfer of endothelial progenitor cells and provides a brief outline of the studies performed to date. An overview of how therapeutic strategies targeting the microvasculature could be enhanced in the future is also presented.


Assuntos
Indutores da Angiogênese/uso terapêutico , Microcirculação/fisiologia , Recuperação de Função Fisiológica/fisiologia , Circulação Renal/fisiologia , Insuficiência Renal Crônica/tratamento farmacológico , Insuficiência Renal Crônica/fisiopatologia , Transferência Adotiva , Humanos , Microcirculação/efeitos dos fármacos , Neovascularização Fisiológica/efeitos dos fármacos , Neovascularização Fisiológica/fisiologia , Recuperação de Função Fisiológica/efeitos dos fármacos , Regeneração/efeitos dos fármacos , Regeneração/fisiologia , Circulação Renal/efeitos dos fármacos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA