Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 33
Filter
1.
Development ; 148(2)2021 01 26.
Article in English | MEDLINE | ID: mdl-33500317

ABSTRACT

Pathogenic mutations in the endocytic receptor LRP2 in humans are associated with severe neural tube closure defects (NTDs) such as anencephaly and spina bifida. Here, we have combined analysis of neural tube closure in mouse and in the African Clawed Frog Xenopus laevis to elucidate the etiology of Lrp2-related NTDs. Lrp2 loss of function impaired neuroepithelial morphogenesis, culminating in NTDs that impeded anterior neural plate folding and neural tube closure in both model organisms. Loss of Lrp2 severely affected apical constriction as well as proper localization of the core planar cell polarity (PCP) protein Vangl2, demonstrating a highly conserved role of the receptor in these processes, which are essential for neural tube formation. In addition, we identified a novel functional interaction of Lrp2 with the intracellular adaptor proteins Shroom3 and Gipc1 in the developing forebrain. Our data suggest that, during neurulation, motifs within the intracellular domain of Lrp2 function as a hub that orchestrates endocytic membrane removal for efficient apical constriction, as well as PCP component trafficking in a temporospatial manner.


Subject(s)
Endocytosis , Intracellular Space/metabolism , Low Density Lipoprotein Receptor-Related Protein-2/metabolism , Neural Tube/embryology , Animals , Cell Membrane/metabolism , Cell Polarity , Low Density Lipoprotein Receptor-Related Protein-2/deficiency , Mice, Inbred C57BL , Models, Biological , Morphogenesis , Neural Tube/metabolism , Neural Tube/ultrastructure , Neuroepithelial Cells/metabolism , Prosencephalon/metabolism , Protein Binding , Xenopus , Xenopus Proteins/metabolism
2.
Am J Physiol Renal Physiol ; 318(5): F1284-F1294, 2020 05 01.
Article in English | MEDLINE | ID: mdl-32200668

ABSTRACT

Proximal tubule (PT) cells express a single saturable albumin-binding site whose affinity matches the estimated tubular concentration of albumin; however, albumin uptake capacity is greatly increased under nephrotic conditions. Deciphering the individual contributions of megalin and cubilin to the uptake of normal and nephrotic levels of albumin is impossible in vivo, as knockout of megalin in mice globally disrupts PT endocytic uptake. We quantified concentration-dependent albumin uptake in an optimized opossum kidney cell culture model and fit the kinetic profiles to identify albumin-binding affinities and uptake capacities. Mathematical deconvolution fit best to a three-component model that included saturable high- and low-affinity uptake sites for albumin and underlying nonsaturable uptake consistent with passive uptake of albumin in the fluid phase. Knockdown of cubilin or its chaperone amnionless selectively reduced the binding capacity of the high-affinity site, whereas knockdown of megalin impacted the low-affinity site. Knockdown of disabled-2 decreased the capacities of both binding sites. Additionally, knockdown of megalin or disabled-2 profoundly inhibited the uptake of a fluid phase marker, with cubilin knockdown having a more modest effect. We propose a novel model for albumin retrieval along the PT in which cubilin and megalin receptors have different functions in recovering filtered albumin in proximal tubule cells. Cubilin binding to albumin is tuned to capture normally filtered levels of the protein. In contrast, megalin binding to albumin is of lower affinity, and its expression is also essential for enabling the recovery of high concentrations of albumin in the fluid phase.


Subject(s)
Albuminuria/metabolism , Kidney Tubules, Proximal/metabolism , Low Density Lipoprotein Receptor-Related Protein-2/metabolism , Nephrosis/metabolism , Receptors, Cell Surface/metabolism , Serum Albumin/metabolism , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Albuminuria/genetics , Albuminuria/physiopathology , Animals , Apoptosis Regulatory Proteins/genetics , Apoptosis Regulatory Proteins/metabolism , Cell Line , Disease Models, Animal , Endocytosis , Female , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Kidney Tubules, Proximal/physiopathology , Kinetics , Low Density Lipoprotein Receptor-Related Protein-2/deficiency , Low Density Lipoprotein Receptor-Related Protein-2/genetics , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice, Knockout , Models, Biological , Nephrosis/genetics , Nephrosis/physiopathology , Opossums , Receptors, Cell Surface/deficiency , Receptors, Cell Surface/genetics
3.
Invest Ophthalmol Vis Sci ; 60(1): 322-330, 2019 01 02.
Article in English | MEDLINE | ID: mdl-30665232

ABSTRACT

Purpose: Mutations in the megalin-encoding gene, LRP2, cause high myopia as seen in patients suffering from Donnai-Barrow/facio-oculo-acoustico-renal syndrome. Megalin is present in both the nonpigmented epithelium of the ciliary body and in the RPE. In this study, we set out to establish an animal model to study the mechanisms underlying the ocular phenotype and to establish if high myopia/megaophthalmos is induced by postnatal megalin-deficiency in the RPE. Methods: Postnatal RPE-specific deletion of megalin was generated by crossing mice bearing a homozygous loxP-flanked Lrp2 allele with transgenic mice expressing the Cre recombinase driven by the BEST1 promotor. The model was investigated by immunohistologic techniques, and transmission electron microscopy. Results: Mice with postnatal RPE-specific loss of megalin developed a megaophthalmos phenotype with dramatic increase in ocular size and severe retinal thinning associated with compromised vision. This phenotype was present at postnatal day 14, indicating rapid development in the period from onset of BEST1 promotor activity at postnatal day 10. Additionally, RPE melanosomes exhibited abnormal size and morphology, suggested by electron tomography to be caused by fusion events between multiple melanosomes. Conclusions: Postnatal loss of megalin in the RPE induces dramatic and rapid ocular growth and retinal degeneration compatible with the high myopia observed in Donnai-Barrow patients. The morphologic changes of RPE melanosomes, believed to be largely inert and fully differentiated at birth, suggested a continued plasticity of mature melanosomes and a requirement for megalin to maintain their number and morphology.


Subject(s)
Eye Abnormalities/etiology , Low Density Lipoprotein Receptor-Related Protein-2/deficiency , Melanosomes/pathology , Retinal Degeneration/etiology , Retinal Pigment Epithelium/metabolism , Animals , Bestrophins/genetics , Eye Abnormalities/metabolism , Eye Abnormalities/pathology , Female , Integrases/metabolism , Male , Melanosomes/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , Retinal Degeneration/metabolism , Retinal Pigment Epithelium/ultrastructure
4.
Cell Rep ; 25(9): 2457-2469.e8, 2018 11 27.
Article in English | MEDLINE | ID: mdl-30485812

ABSTRACT

Neural stem cells (NSCs) contribute to plasticity and repair of the adult brain. Niches harboring NSCs regulate stem cell self-renewal and differentiation. We used comprehensive and untargeted single-cell RNA profiling to generate a molecular cell atlas of the largest germinal region of the adult mouse brain, the subventricular zone (SVZ). We characterized >20 neural and non-neural cell types and gained insights into the dynamics of neurogenesis by predicting future cell states based on computational analysis of RNA kinetics. Furthermore, we applied our single-cell approach to document decreased numbers of NSCs, reduced proliferation activity of progenitors, and perturbations in Wnt and BMP signaling pathways in mice lacking LRP2, an endocytic receptor required for SVZ maintenance. Our data provide a valuable resource to study adult neurogenesis and a proof of principle for the power of single-cell RNA sequencing to elucidate neural cell-type-specific alterations in loss-of-function models.


Subject(s)
Aging/genetics , Lateral Ventricles/cytology , Neurogenesis/genetics , Single-Cell Analysis , Transcriptome/genetics , Animals , Cell Lineage , Cell Proliferation , Dentate Gyrus/cytology , Gene Expression Regulation , Low Density Lipoprotein Receptor-Related Protein-2/deficiency , Low Density Lipoprotein Receptor-Related Protein-2/metabolism , Mice, Inbred C57BL , Models, Biological , Neural Stem Cells/cytology , Neural Stem Cells/metabolism , RNA/metabolism , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism , Stem Cell Niche/genetics
5.
Kidney Int ; 93(2): 335-342, 2018 02.
Article in English | MEDLINE | ID: mdl-29032953

ABSTRACT

The megalin/cubilin receptor complex is required for proximal tubular endocytosis and degradation of filtered albumin. An additional high-capacity retrieval pathway of intact albumin for the recovery of large amounts of filtered albumin has been proposed, possibly involving cooperation between megalin/cubilin and the neonatal Fc receptor. To clarify the potential role of such a pathway, we examined the effects of megalin/cubilin gene inactivation on tubular albumin uptake and plasma albumin levels in nephrotic, podocin knockout mice. Immunofluorescence microscopy of megalin/cubilin/podocin knockout mouse kidneys demonstrated abolishment of proximal tubule albumin uptake, in contrast to the excessive albumin accumulation observed in podocin knockout mice compared to controls. Correspondingly, urinary albumin excretion was increased 1.4 fold in megalin/cubilin/podocin compared to podocin knockout mice (albumin/creatinine: 226 vs. 157 mg/mg). However, no difference in plasma albumin levels was observed between megalin/cubilin/podocin and podocin knockout mice, as both were reduced to approximately 40% of controls. There were no differences in liver albumin synthesis by mRNA levels and protein abundance. Thus, megalin/cubilin knockout efficiently blocks proximal tubular albumin uptake in nephrotic mice but plasma albumin levels did not differ as a result of megalin/cubilin-deficiency, suggesting no significance of the megalin/cubilin-pathway for albumin homeostasis by retrieval of intact albumin.


Subject(s)
Albuminuria/metabolism , Endocytosis , Kidney Tubules, Proximal/metabolism , Nephrotic Syndrome/metabolism , Serum Albumin/metabolism , Albuminuria/blood , Albuminuria/genetics , Albuminuria/urine , Animals , Creatinine/urine , Disease Models, Animal , Female , Intracellular Signaling Peptides and Proteins/deficiency , Intracellular Signaling Peptides and Proteins/genetics , Liver/metabolism , Low Density Lipoprotein Receptor-Related Protein-2/deficiency , Low Density Lipoprotein Receptor-Related Protein-2/genetics , Membrane Proteins/deficiency , Membrane Proteins/genetics , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Knockout , Nephrotic Syndrome/blood , Nephrotic Syndrome/genetics , Nephrotic Syndrome/urine , Receptors, Cell Surface/deficiency , Receptors, Cell Surface/genetics
6.
Am J Respir Cell Mol Biol ; 57(5): 589-602, 2017 11.
Article in English | MEDLINE | ID: mdl-28678521

ABSTRACT

Acute respiratory distress syndrome constitutes a significant disease burden with regard to both morbidity and mortality. Current therapies are mostly supportive and do not address the underlying pathophysiologic mechanisms. Removal of protein-rich alveolar edema-a clinical hallmark of acute respiratory distress syndrome-is critical for survival. Here, we describe a transforming growth factor (TGF)-ß-triggered mechanism, in which megalin, the primary mediator of alveolar protein transport, is negatively regulated by glycogen synthase kinase (GSK) 3ß, with protein phosphatase 1 and nuclear inhibitor of protein phosphatase 1 being involved in the signaling cascade. Inhibition of GSK3ß rescued transepithelial protein clearance in primary alveolar epithelial cells after TGF-ß treatment. Moreover, in a bleomycin-based model of acute lung injury, megalin+/- animals (the megalin-/- variant is lethal due to postnatal respiratory failure) showed a marked increase in intra-alveolar protein and more severe lung injury compared with wild-type littermates. In contrast, wild-type mice treated with the clinically relevant GSK3ß inhibitors, tideglusib and valproate, exhibited significantly decreased alveolar protein concentrations, which was associated with improved lung function and histopathology. Together, we discovered that the TGF-ß-GSK3ß-megalin axis is centrally involved in disturbances of alveolar protein clearance in acute lung injury and provide preclinical evidence for therapeutic efficacy of GSK3ß inhibition.


Subject(s)
Acute Lung Injury/metabolism , Acute Lung Injury/therapy , Low Density Lipoprotein Receptor-Related Protein-2/metabolism , Pulmonary Alveoli/metabolism , Acute Lung Injury/genetics , Animals , Glycogen Synthase Kinase 3 beta/antagonists & inhibitors , Glycogen Synthase Kinase 3 beta/metabolism , Low Density Lipoprotein Receptor-Related Protein-2/deficiency , Lung/metabolism , Mice, Inbred C57BL , Mice, Knockout , Pulmonary Edema/metabolism , Pulmonary Edema/therapy , Respiratory Distress Syndrome/metabolism , Respiratory Distress Syndrome/therapy , Transforming Growth Factor beta/metabolism
7.
J Neuroinflammation ; 14(1): 26, 2017 01 31.
Article in English | MEDLINE | ID: mdl-28143489

ABSTRACT

BACKGROUND: The multiligand receptor megalin controls the brain uptake of a number of ligands, including insulin and leptin. Despite the role of megalin in the transport of these metabolically relevant hormones, the role of megalin at the blood-brain-barrier (BBB) has not yet been explored in the context of metabolic regulation. METHODS: Here we investigate the role of brain endothelial megalin in energy metabolism and leptin signaling using an endothelial cell-specific megalin deficient (EMD) mouse model. RESULTS: We found megalin is important to protect mice from developing obesity and metabolic syndrome when mice are fed a normal chow diet. EMD mice developed neuroinflammation, by triggering several pro-inflammatory cytokines, displayed reduced neurogenesis and mitochondrial deregulation. CONCLUSIONS: These results implicate brain endothelial megalin expression in obesity-related metabolic changes through the leptin signaling pathway proposing a potential link between obesity and neurodegeneration.


Subject(s)
Encephalitis/genetics , Encephalitis/pathology , Endothelial Cells/metabolism , Low Density Lipoprotein Receptor-Related Protein-2/deficiency , Obesity/genetics , Obesity/pathology , Age Factors , Animals , Blood-Brain Barrier/metabolism , Blood-Brain Barrier/pathology , Brain/physiopathology , Cytokines/genetics , Cytokines/metabolism , Disease Models, Animal , Doublecortin Domain Proteins , Eating/genetics , Glial Fibrillary Acidic Protein/metabolism , Low Density Lipoprotein Receptor-Related Protein-2/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Microtubule-Associated Proteins/metabolism , Mitochondria/pathology , Mitochondria/ultrastructure , Neuropeptides/metabolism , Phosphopyruvate Hydratase/metabolism , STAT3 Transcription Factor/metabolism
8.
Dev Dyn ; 245(5): 569-79, 2016 05.
Article in English | MEDLINE | ID: mdl-26872844

ABSTRACT

To fulfill their multiple roles in organ development and adult tissue homeostasis, hedgehog (HH) morphogens act through their receptor Patched (PTCH) on target cells. However, HH actions also require HH binding proteins, auxiliary cell surface receptors that agonize or antagonize morphogen signaling in a context-dependent manner. Here, we discuss recent findings on the LDL receptor-related protein 2 (LRP2), an exemplary HH binding protein that modulates sonic hedgehog activities in stem and progenitor cell niches in embryonic and adult tissues. LRP2 functions are crucial for developmental processes in a number of tissues, including the brain, the eye, and the heart, and defects in this receptor pathway are the cause of devastating congenital diseases in humans. Developmental Dynamics 245:569-579, 2016. © 2016 Wiley Periodicals, Inc.


Subject(s)
Hedgehog Proteins/metabolism , Low Density Lipoprotein Receptor-Related Protein-2/physiology , Animals , Congenital Abnormalities/embryology , Congenital Abnormalities/etiology , Embryonic Development , Humans , Low Density Lipoprotein Receptor-Related Protein-2/deficiency , Morphogenesis , Signal Transduction/physiology
9.
Dis Model Mech ; 9(4): 413-25, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26822476

ABSTRACT

Lipoprotein-related receptor protein 2 (LRP2) is important for development of the embryonic neural crest and brain in both mice and humans. Although a role in cardiovascular development can be expected, the hearts ofLrp2knockout (KO) mice have not yet been investigated. We studied the cardiovascular development ofLrp2KO mice between embryonic day 10.5 (E10.5) and E15.5, applying morphometry and immunohistochemistry, using antibodies against Tfap2α (neural crest cells), Nkx2.5 (second heart field), WT1 (epicardium derived cells), tropomyosin (myocardium) and LRP2. TheLrp2KO mice display a range of severe cardiovascular abnormalities, including aortic arch anomalies, common arterial trunk (persistent truncus arteriosus) with coronary artery anomalies, ventricular septal defects, overriding of the tricuspid valve and marked thinning of the ventricular myocardium. Both the neural crest cells and second heart field, which are essential for the lengthening and growth of the right ventricular outflow tract, are abnormally positioned in theLrp2KO. This explains the absence of the aorto-pulmonary septum, which leads to common arterial trunk and ventricular septal defects. Severe blebbing of the epicardial cells covering the ventricles is seen. Epithelial-mesenchymal transition does occur; however, there are fewer WT1-positive epicardium-derived cells in the ventricular wall as compared to normal, coinciding with the myocardial thinning and deep intertrabecular spaces. LRP2 plays a crucial role in cardiovascular development in mice. This corroborates findings of cardiac anomalies in humans withLRP2mutations. Future studies should reveal the underlying signaling mechanisms in which LRP2 is involved during cardiogenesis.


Subject(s)
Heart Defects, Congenital/embryology , Heart Defects, Congenital/pathology , Heart/embryology , Low Density Lipoprotein Receptor-Related Protein-2/deficiency , Animals , Cell Movement , Embryo, Mammalian/abnormalities , Embryo, Mammalian/pathology , Endothelium, Vascular/embryology , Endothelium, Vascular/pathology , Female , Fluorescent Antibody Technique , Heart Ventricles/embryology , Heart Ventricles/pathology , Imaging, Three-Dimensional , Low Density Lipoprotein Receptor-Related Protein-2/metabolism , Male , Mice, Inbred C57BL , Mice, Knockout , Models, Cardiovascular , Myocardium/pathology , Neural Crest/pathology , Pericardium/embryology , Pericardium/pathology
10.
J Cell Sci ; 127(Pt 10): 2261-8, 2014 May 15.
Article in English | MEDLINE | ID: mdl-24639464

ABSTRACT

The low-density lipoprotein (LDL) receptor-related protein 2 (LRP2) is a multifunctional cell-surface receptor expressed in the embryonic neuroepithelium. Loss of LRP2 in the developing murine central nervous system (CNS) causes impaired closure of the rostral neural tube at embryonic stage (E) 9.0. Similar neural tube defects (NTDs) have previously been attributed to impaired folate metabolism in mice. We therefore asked whether LRP2 might be required for the delivery of folate to neuroepithelial cells during neurulation. Uptake assays in whole-embryo cultures showed that LRP2-deficient neuroepithelial cells are unable to mediate the uptake of folate bound to soluble folate receptor 1 (sFOLR1). Consequently, folate concentrations are significantly reduced in Lrp2(-/-) embryos compared with control littermates. Moreover, the folic-acid-dependent gene Alx3 is significantly downregulated in Lrp2 mutants. In conclusion, we show that LRP2 is essential for cellular folate uptake in the developing neural tube, a crucial step for proper neural tube closure.


Subject(s)
Folic Acid/metabolism , Low Density Lipoprotein Receptor-Related Protein-2/metabolism , Neural Tube/metabolism , Animals , Endocytosis , Folate Receptor 1/metabolism , Homeodomain Proteins/biosynthesis , Homeodomain Proteins/genetics , Low Density Lipoprotein Receptor-Related Protein-2/deficiency , Low Density Lipoprotein Receptor-Related Protein-2/genetics , Mice , Mice, Inbred C57BL , Mice, Transgenic , Neural Tube/embryology , Reduced Folate Carrier Protein/biosynthesis , Reduced Folate Carrier Protein/genetics
11.
J Alzheimers Dis ; 39(4): 711-7, 2014.
Article in English | MEDLINE | ID: mdl-24254699

ABSTRACT

Megalin has been suggested to be involved in Alzheimer's disease (AD), mediating blood-brain barrier (BBB) transport of multiple ligands, including amyloid-ß peptide (Aß), but also neuroprotective factors. Because no transgenic model is currently available to study this concept, we have obtained transgenic mice blocking megalin expression at the BBB. These endothelial megalin deficient (EMD) mice developed increased anxiety behavior and impaired learning ability and recognition memory, similar to symptoms described in AD. Degenerating neurons were also observed in the cerebral cortex of EMD mice. In view of our findings we suggest that, in mice, megalin deficiency at the BBB leads to neurodegeneration.


Subject(s)
Alzheimer Disease/metabolism , Cognition Disorders/metabolism , Endothelial Cells/metabolism , Low Density Lipoprotein Receptor-Related Protein-2/deficiency , Alzheimer Disease/genetics , Alzheimer Disease/pathology , Animals , Cells, Cultured , Cognition Disorders/genetics , Cognition Disorders/pathology , Endothelial Cells/pathology , Humans , Low Density Lipoprotein Receptor-Related Protein-2/genetics , Male , Mice , Mice, Knockout , Mice, Transgenic
12.
Nephrol Dial Transplant ; 28(3): 585-91, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23048173

ABSTRACT

BACKGROUND: The reabsorption of filtered plasma proteins, hormones and vitamins by the renal proximal tubules is vital for body homeostasis. Studies of megalin-deficient mice suggest that the large multi-ligand endocytic receptor megalin plays an essential role in this process. In humans, dysfunctional megalin causes the extremely rare Donnai-Barrow/Facio-Oculo-Acustico-Renal (DB/FOAR) syndrome characterized by a characteristic and multifaceted phenotype including low-molecular-weight proteinuria. In this study, we examined the role of megalin for tubular protein reabsorption in humans through analysis of proximal tubular function in megalin-deficient patients. METHODS: Direct sequencing of the megalin-encoding gene (LRP2) was performed in a family in which three children presented with classical DB/FOAR manifestations. Renal consequences of megalin deficiency were investigated through immunohistochemical analyses of renal biopsy material and immunoblotting of urine samples. RESULTS: In the patients, a characteristic urinary protein profile with increased urinary excretion of vitamin D-binding protein, retinol-binding protein and albumin was associated with absence of, or reduced, proximal tubular endocytic uptake as shown by renal immunohistochemistry. In the absence of tubular uptake, urinary albumin excretion was in the micro-albuminuric range suggesting that limited amounts of albumin are filtered in human glomeruli. CONCLUSIONS: This study demonstrated that megalin plays an essential role for human proximal tubular protein reabsorption and suggests that only limited amounts of albumin is normally filtered in the human glomeruli. Finally, we propose that the characteristic urinary protein profile of DB/FOAR patients may be utilized as a diagnostic marker of megalin dysfunction.


Subject(s)
Agenesis of Corpus Callosum/pathology , Albumins/metabolism , Hearing Loss, Sensorineural/pathology , Kidney Tubules, Proximal/pathology , Low Density Lipoprotein Receptor-Related Protein-2/deficiency , Mutation/genetics , Myopia/pathology , Proteinuria/pathology , Agenesis of Corpus Callosum/genetics , Agenesis of Corpus Callosum/metabolism , Child, Preschool , Female , Hearing Loss, Sensorineural/genetics , Hearing Loss, Sensorineural/metabolism , Hernias, Diaphragmatic, Congenital , Humans , Kidney Tubules, Proximal/metabolism , Low Density Lipoprotein Receptor-Related Protein-2/genetics , Myopia/genetics , Myopia/metabolism , Phenotype , Proteinuria/genetics , Proteinuria/metabolism , Renal Tubular Transport, Inborn Errors
13.
J Am Soc Nephrol ; 23(7): 1181-9, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22518004

ABSTRACT

Angiotensin II content in the kidney is much higher than in the plasma, and it increases more in kidney diseases through an uncertain mechanism. Because the kidney abundantly expresses angiotensinogen mRNA, transcriptional dysregulation of angiotensinogen within the kidney is one potential cause of increased renal angiotensin II in the setting of disease. Here, we observed that kidney-specific angiotensinogen knockout mice had levels of renal angiotensinogen protein and angiotensin II that were similar to those levels of control mice. In contrast, liver-specific knockout of angiotensinogen nearly abolished plasma and renal angiotensinogen protein and renal tissue angiotensin II. Immunohistochemical analysis in mosaic proximal tubules of megalin knockout mice revealed that angiotensinogen protein was incorporated selectively in megalin-intact cells of the proximal tubule, indicating that the proximal tubule reabsorbs filtered angiotensinogen through megalin. Disruption of the filtration barrier in a transgenic mouse model of podocyte-selective injury increased renal angiotensin II content and markedly increased both tubular and urinary angiotensinogen protein without an increase in renal renin activity, supporting the dependency of renal angiotensin II generation on filtered angiotensinogen. Taken together, these data suggest that liver-derived angiotensinogen is the primary source of renal angiotensinogen protein and angiotensin II. Furthermore, an abnormal increase in the permeability of the glomerular capillary wall to angiotensinogen, which characterizes proteinuric kidney diseases, enhances the synthesis of renal angiotensin II.


Subject(s)
Angiotensin II/metabolism , Angiotensinogen/metabolism , Kidney/metabolism , Liver/metabolism , Angiotensin II/deficiency , Angiotensin II/genetics , Angiotensinogen/deficiency , Angiotensinogen/genetics , Animals , Kidney/pathology , Liver/pathology , Low Density Lipoprotein Receptor-Related Protein-2/deficiency , Low Density Lipoprotein Receptor-Related Protein-2/genetics , Low Density Lipoprotein Receptor-Related Protein-2/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Models, Animal , Phenotype , RNA, Messenger/metabolism
14.
J Am Soc Nephrol ; 23(4): 591-6, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22282591

ABSTRACT

Urinary albumin excretion is an important diagnostic and prognostic marker of renal function. Both animal and human urine contain large amounts of albumin fragments, but whether these fragments originate from renal tubular degradation of filtered albumin is unknown. Here, we used mice with kidneys lacking megalin and cubilin, the coreceptors that mediate proximal tubular endocytosis of albumin, to determine whether proximal tubular degradation of albumin forms the detectable urinary albumin fragments. After intravenous administration of (125)I-labeled mouse albumin to knockout and control mice, we examined kidney uptake of albumin and urinary excretion of both intact albumin and its fragments using size exclusion chromatography. In control mice, all labeled albumin eluted as albumin fragments in the urine. In megalin/cubilin-deficient mice, we observed decreased uptake and degradation of albumin and increased urinary excretion of intact albumin; we did not, however, detect a decrease in the excretion of albumin fragments. These results show that the generation of urinary albumin fragments occurs independently of renal tubular uptake and degradation of albumin, suggesting that the pathophysiological implications of changes in urinary albumin fragments require reevaluation.


Subject(s)
Albumins/metabolism , Albuminuria/diagnosis , Kidney Tubular Necrosis, Acute/physiopathology , Kidney Tubules, Proximal/metabolism , Animals , Creatinine/analysis , Disease Models, Animal , Humans , Kidney Function Tests , Low Density Lipoprotein Receptor-Related Protein-2/deficiency , Mice , Mice, Knockout , Random Allocation , Receptors, Cell Surface/deficiency , Sensitivity and Specificity , Urinalysis
16.
Eur J Nucl Med Mol Imaging ; 38(4): 623-32, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21170526

ABSTRACT

PURPOSE: Radiolabelled peptides used for peptide receptor radionuclide therapy are excreted mainly via the kidneys and are partly reabsorbed and retained in the proximal tubular cells. The resulting high renal radiation dose can cause nephrotoxicity, limiting the maximum activity dose and the effectiveness of peptide receptor radionuclide therapy. The mechanisms of kidney reabsorption of these peptides are incompletely understood, but the scavenger receptor megalin has been shown to play a role in the reabsorption of (111)In-octreotide. In this study, the role of megalin in the renal reabsorption of various relevant radiolabelled peptides was investigated. METHODS: Groups of kidney-specific megalin-deficient mice and wild-type mice were injected with (111)In-labelled somatostatin, exendin, neurotensin or minigastrin analogues. Single photon emission computed tomographic (SPECT) images of the kidneys were acquired and analysed quantitatively, or the animals were killed 3 h after injection and the activity concentration in the kidneys was measured. RESULTS: Megalin-deficient mice showed significantly lower uptake of all studied radiolabelled peptides in the kidneys, ranging from 22% ((111)In-octreotide) to 65% ((111)In-exendin) of uptake in wild-type kidneys. Quantitative analysis of renal uptake by SPECT and ex vivo measurements showed a very good correlation. CONCLUSION: Megalin is involved in the renal reabsorption of radiolabelled octreotide, octreotate, exendin, neurotensin and minigastrin. This knowledge may help in the design of strategies to reduce this reabsorption and the resulting nephrotoxicity in peptide receptor radionuclide therapy, enabling more effective therapy. Small-animal SPECT is an accurate tool, allowing in vivo quantification of renal uptake and serial measurements in individual mice.


Subject(s)
Kidney/diagnostic imaging , Kidney/metabolism , Low Density Lipoprotein Receptor-Related Protein-2/deficiency , Low Density Lipoprotein Receptor-Related Protein-2/metabolism , Peptides/metabolism , Peptides/pharmacokinetics , Tomography, Emission-Computed, Single-Photon , Animals , Female , Immunohistochemistry , Isotope Labeling , Male , Mice , Organ Specificity , Peptides/therapeutic use , Protein Transport , Tissue Distribution
17.
J Nucl Med ; 51(6): 973-7, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20484435

ABSTRACT

UNLABELLED: We determined the renal radiation dose of a series of (111)In-labeled peptides using animal SPECT. Because the animals' health deteriorated, renal toxicity was assessed. METHODS: Wild-type and megalin-deficient mice were imaged repeatedly at 3- to 6-wk intervals to quantify renal retention after injection of 40-50 MBq of (111)In-diethylenetriaminepentaacetic acid-labeled peptides (octreotide, exendin, octreotate, neurotensin, and minigastrin analogs), and the absorbed kidney radiation doses were estimated. Body weight, renal function parameters, and renal histology were determined at 16-20 wk after the first scan and compared with those in naive animals. RESULTS: Because of high renal retention, (111)In-diethylenetriaminepentaacetic acid-exendin-4 scans resulted in a 70-Gy kidney radiation dose in wild-type mice. Megalin-deficient kidneys received 20-40 Gy. The other peptides resulted in much lower renal doses. Kidney function monitoring indicated renal damage in imaged animals. CONCLUSION: Micro-SPECT enables longitudinal studies in 1 animal. However, long-term nephrotoxic effects may be induced after high renal radiation doses, even with (111)In-labeled radiotracers.


Subject(s)
Indium Radioisotopes , Kidney/radiation effects , Peptides , Tomography, Emission-Computed, Single-Photon/adverse effects , Animals , Exenatide , Female , Injections , Kidney/pathology , Kidney/physiopathology , Low Density Lipoprotein Receptor-Related Protein-2/deficiency , Low Density Lipoprotein Receptor-Related Protein-2/metabolism , Male , Mice , Organ Specificity , Pentetic Acid/chemistry , Peptides/administration & dosage , Peptides/chemistry , Radiation Dosage , Risk , Time Factors , Tomography, X-Ray Computed , Venoms/chemistry
18.
Am J Physiol Renal Physiol ; 298(6): F1457-64, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20237235

ABSTRACT

Connective tissue growth factor (CTGF) plays a key role in renal fibrosis. Urinary CTGF is elevated in various renal diseases and may have biomarker potential. However, it is unknown which processes contribute to elevated urinary CTGF levels. Thus far, urinary CTGF was considered to reflect renal expression. We investigated how tubular dysfunction affects urinary CTGF levels. To study this, we administered recombinant CTGF intravenously to rodents. We used both full-length CTGF and the NH(2)-terminal fragment, since the NH(2)-fragment is the predominant form detected in urine. Renal CTGF extraction, determined by simultaneous arterial and renal vein sampling, was 18 +/- 3% for full-length CTGF and 21 +/- 1% for the NH(2)-fragment. Fractional excretion was very low for both CTGFs (0.02 +/- 0.006% and 0.10 +/- 0.02%, respectively), indicating that >99% of the extracted CTGF was metabolized by the kidney. Immunohistochemistry revealed extensive proximal tubular uptake of CTGF in apical endocytic vesicles and colocalization with megalin. Urinary CTGF was elevated in megalin- and cubilin-deficient mice but not in cubilin-deficient mice. Inhibition of tubular reabsorption by Gelofusine reduced renal uptake of CTGF and increased urinary CTGF. In healthy volunteers, Gelofusine also induced an increase of urinary CTGF excretion, comparable to the increase of beta(2)-microglobulin excretion (r = 0.99). Furthermore, urinary CTGF correlated with beta(2)-microglobulin (r = 0.85) in renal disease patients (n = 108), and only beta(2)-microglobulin emerged as an independent determinant of urinary CTGF. Thus filtered CTGF is normally reabsorbed almost completely in proximal tubules via megalin, and elevated urinary CTGF may largely reflect proximal tubular dysfunction.


Subject(s)
Connective Tissue Growth Factor/urine , Kidney Diseases/metabolism , Kidney Tubules, Proximal/metabolism , Peptide Fragments/urine , Animals , Biomarkers/blood , Biomarkers/urine , Connective Tissue Growth Factor/administration & dosage , Connective Tissue Growth Factor/blood , Connective Tissue Growth Factor/pharmacokinetics , Cross-Sectional Studies , Endocytosis , Glomerular Filtration Rate , Humans , Infusions, Parenteral , Injections, Intravenous , Kidney Diseases/physiopathology , Kidney Tubules, Proximal/drug effects , Kidney Tubules, Proximal/physiopathology , Low Density Lipoprotein Receptor-Related Protein-2/deficiency , Low Density Lipoprotein Receptor-Related Protein-2/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Peptide Fragments/administration & dosage , Peptide Fragments/blood , Peptide Fragments/pharmacokinetics , Polygeline/administration & dosage , Rats , Rats, Inbred WKY , Receptors, Cell Surface/deficiency , Receptors, Cell Surface/genetics , Recombinant Fusion Proteins/urine , beta 2-Microglobulin/urine
19.
J Neurosci ; 29(10): 3220-32, 2009 Mar 11.
Article in English | MEDLINE | ID: mdl-19279259

ABSTRACT

Mutated transthyretin (TTR) causes familial amyloid polyneuropathy, a neurodegenerative disorder characterized by TTR deposition in the peripheral nervous system (PNS). The origin/reason for TTR deposition in the nerve is unknown. Here we demonstrate that both endogenous mouse TTR and TTR injected intravenously have access to the mouse sciatic nerve. We previously determined that in the absence of TTR, both neurite outgrowth in vitro and nerve regeneration in vivo were impaired. Reinforcing this finding, we now show that local TTR delivery to the crushed sciatic nerve rescues the regeneration phenotype of TTR knock-out (KO) mice. As the absence of TTR was unrelated to neuronal survival, we further evaluated the Schwann cell and inflammatory response to injury, as well as axonal retrograde transport, in the presence/absence of TTR. Only retrograde transport was impaired in TTR KO mice which, in addition to the neurite outgrowth impairment, might account for the decreased regeneration in this strain. Moreover, we show that in vitro, in dorsal root ganglia neurons, clathrin-dependent megalin-mediated TTR internalization is needed for TTR neuritogenic activity. Supporting this observation, we demonstrate that in vivo, decreased levels of megalin lead to decreased nerve regeneration and that megalin's action as a regeneration enhancer is dependent on TTR. In conclusion, our work unravels the mechanism of TTR action during nerve regeneration. Additionally, TTR presence in the nerve, as is here shown, may underlie its preferential deposition in the PNS of familial amyloid polyneuropathy patients.


Subject(s)
Low Density Lipoprotein Receptor-Related Protein-2/physiology , Neurites/metabolism , Neurogenesis/physiology , Prealbumin/metabolism , Sensory Receptor Cells/metabolism , Animals , Cells, Cultured , Endocytosis/genetics , Endocytosis/physiology , Ganglia, Spinal/cytology , Ganglia, Spinal/growth & development , Ganglia, Spinal/metabolism , Humans , Low Density Lipoprotein Receptor-Related Protein-2/biosynthesis , Low Density Lipoprotein Receptor-Related Protein-2/deficiency , Mice , Mice, Knockout , Nerve Regeneration/genetics , Nerve Regeneration/physiology , Neurogenesis/genetics , Prealbumin/deficiency , Prealbumin/genetics , Prealbumin/physiology , Sensory Receptor Cells/cytology
20.
Am J Physiol Renal Physiol ; 296(4): F902-11, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19193726

ABSTRACT

Human glomerulonephritis (GN) is characterized by sustained proteinuria, sodium retention, hypertension, and edema formation. Increasing quantities of filtered protein enter the renal tubule, where they may alter epithelial transport functions. Exaggerated endocytosis and consequent protein overload may affect proximal tubules, but intrinsic malfunction of distal epithelia has also been reported. A straightforward assignment to a particular tubule segment causing salt retention in GN is still controversial. We hypothesized that 1) trafficking and surface expression of major transporters and channels involved in volume regulation were altered in GN, and 2) proximal tubular endocytosis may influence locally as well as downstream expressed tubular transporters and channels. Effects of anti-glomerular basement membrane GN were studied in controls and megalin-deficient mice with blunted proximal endocytosis. Mice displayed salt retention and elevated systolic blood pressure when proteinuria had reached 10-15 mg/24 h. Surface expression of proximal Na(+)-coupled transporters and water channels was in part [Na(+)-P(i) cotransporter IIa (NaPi-IIa) and aquaporin-1 (AQP1)] increased by megalin deficiency alone, but unchanged (Na(+)/H(+) exchanger 3) or reduced (NaPi-IIa and AQP1) in GN irrespective of the endocytosis defect. In distal epithelia, significant increases in proteolytic cleavage products of alpha-epithelial Na(+) channel (ENaC) and gamma-ENaC were observed, suggesting enhanced tubular sodium reabsorption. The effects of glomerular proteinuria dominated over those of blunted proximal endocytosis in contributing to ENaC cleavage. Our data indicate that ENaC-mediated sodium entry may be the rate-limiting step in proteinuric sodium retention. Enhanced proteolytic cleavage of ENaC points to a novel mechanism of channel activation which may involve the action of filtered plasma proteases.


Subject(s)
Endocytosis , Glomerulonephritis/metabolism , Kidney Tubules/metabolism , Membrane Transport Proteins/metabolism , Proteinuria/metabolism , Sodium/metabolism , Aldosterone/blood , Animals , Aquaporin 1/metabolism , Aquaporin 2/metabolism , Autoantibodies , Blood Pressure , Cyclooxygenase 2/blood , Disease Models, Animal , Epithelial Sodium Channels/metabolism , Female , Glomerulonephritis/immunology , Glomerulonephritis/physiopathology , Kidney Tubules/enzymology , Kidney Tubules/immunology , Low Density Lipoprotein Receptor-Related Protein-2/deficiency , Low Density Lipoprotein Receptor-Related Protein-2/genetics , Mice , Mice, Knockout , Protein Transport , Proteinuria/immunology , Proteinuria/physiopathology , Renin/blood , Sodium/blood , Sodium/urine , Sodium-Hydrogen Exchanger 3 , Sodium-Hydrogen Exchangers/metabolism , Sodium-Phosphate Cotransporter Proteins, Type IIa/metabolism , Vitamin D-Binding Protein/urine
SELECTION OF CITATIONS
SEARCH DETAIL
...