Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 20
1.
Cell Rep ; 42(9): 113070, 2023 09 26.
Article En | MEDLINE | ID: mdl-37659079

The TMEM127 gene encodes a transmembrane protein of poorly known function that is mutated in pheochromocytomas, neural crest-derived tumors of adrenomedullary cells. Here, we report that, at single-nucleus resolution, TMEM127-mutant tumors share precursor cells and transcription regulatory elements with pheochromocytomas carrying mutations of the tyrosine kinase receptor RET. Additionally, TMEM127-mutant pheochromocytomas, human cells, and mouse knockout models of TMEM127 accumulate RET and increase its signaling. TMEM127 contributes to RET cellular positioning, trafficking, and lysosome-mediated degradation. Mechanistically, TMEM127 binds to RET and recruits the NEDD4 E3 ubiquitin ligase for RET ubiquitination and degradation via TMEM127 C-terminal PxxY motifs. Lastly, increased cell proliferation and tumor burden after TMEM127 loss can be reversed by selective RET inhibitors in vitro and in vivo. Our results define TMEM127 as a component of the ubiquitin system and identify aberrant RET stabilization as a likely mechanism through which TMEM127 loss-of-function mutations cause pheochromocytoma.


Adrenal Gland Neoplasms , Pheochromocytoma , Humans , Animals , Mice , Pheochromocytoma/genetics , Pheochromocytoma/metabolism , Pheochromocytoma/pathology , Germ-Line Mutation , Adrenal Gland Neoplasms/genetics , Adrenal Gland Neoplasms/metabolism , Adrenal Gland Neoplasms/pathology , Mutation/genetics , Ubiquitination , Proto-Oncogene Proteins c-ret/genetics , Proto-Oncogene Proteins c-ret/metabolism , Membrane Proteins/genetics , Membrane Proteins/metabolism
2.
Am J Physiol Renal Physiol ; 320(1): F114-F129, 2021 01 01.
Article En | MEDLINE | ID: mdl-33283642

Chronic kidney disease results in high serum urea concentrations leading to excessive protein carbamylation, primarily albumin. This is associated with increased cardiovascular disease and mortality. Multiple methods were used to address whether carbamylation alters albumin metabolism. Intravital two-photon imaging of the Munich Wistar Frömter (MWF) rat kidney and liver allowed us to characterize filtration and proximal tubule uptake and liver uptake. Microscale thermophoresis enabled quantification of cubilin (CUB7,8 domain) and FcRn binding. Finally, multiple biophysical methods including dynamic light scattering, small-angle X-ray scattering, LC-MS/MS and in silico analyses were used to identify the critical structural alterations and amino acid modifications of rat albumin. Carbamylation of albumin reduced binding to CUB7,8 and FcRn in a dose-dependent fashion. Carbamylation markedly increased vascular clearance of carbamylated rat serum albumin (cRSA) and altered distribution of cRSA in both the kidney and liver at 16 h post intravenous injection. By evaluating the time course of carbamylation and associated charge, size, shape, and binding parameters in combination with in silico analysis and mass spectrometry, the critical binding interaction impacting carbamylated albumin's reduced FcRn binding was identified as K524. Carbamylation of RSA had no effect on glomerular filtration or proximal tubule uptake. These data indicate urea-mediated time-dependent carbamylation of albumin lysine K524 resulted in reduced binding to CUB7,8 and FcRn that contribute to altered albumin transport, leading to increased vascular clearance and increased liver and endothelial tissue accumulation.


Histocompatibility Antigens Class I/metabolism , Kidney Tubules, Proximal/metabolism , Liver/metabolism , Receptors, Fc/metabolism , Renal Insufficiency, Chronic/metabolism , Serum Albumin/metabolism , Animals , Chromatography, Liquid , Disease Models, Animal , Glomerular Filtration Rate , Kidney Tubules, Proximal/physiopathology , Lysine , Male , Microscopy, Fluorescence, Multiphoton , Protein Binding , Protein Carbamylation , Rats, Inbred Strains , Rats, Sprague-Dawley , Receptors, Cell Surface/metabolism , Renal Insufficiency, Chronic/pathology , Renal Insufficiency, Chronic/physiopathology , Scattering, Small Angle , Tandem Mass Spectrometry , Time Factors , X-Ray Diffraction
3.
J Clin Endocrinol Metab ; 105(9)2020 09 01.
Article En | MEDLINE | ID: mdl-32575117

CONTEXT: TMEM127 is a poorly known tumor suppressor gene associated with pheochromocytomas, paragangliomas, and renal carcinomas. Our incomplete understanding of TMEM127 function has limited our ability to predict variant pathogenicity. PURPOSE: To better understand the function of the transmembrane protein TMEM127 we undertook cellular and molecular evaluation of patient-derived germline variants. DESIGN: Subcellular localization and steady-state levels of tumor-associated, transiently expressed TMEM127 variants were compared to the wild-type protein using immunofluorescence and immunoblot analysis, respectively, in cells genetically modified to lack endogenous TMEM127. Membrane topology and endocytic mechanisms were also assessed. RESULTS: We identified 3 subgroups of mutations and determined that 71% of the variants studied are pathogenic or likely pathogenic through loss of membrane-binding ability, stability, and/or internalization capability. Investigation into an N-terminal cluster of missense variants uncovered a previously unrecognized transmembrane domain, indicating that TMEM127 is a 4- transmembrane, not a 3-transmembrane domain-containing protein. Additionally, a C-terminal variant with predominant plasma membrane localization revealed an atypical, extended acidic, dileucine-based motif required for TMEM127 internalization through clathrin-mediated endocytosis. CONCLUSION: We characterized the functional deficits of several germline TMEM127 variants and identified novel structure-function features of TMEM127. These findings will assist in determining pathogenicity of TMEM127 variants and will help guide future studies investigating the cellular role of TMEM127.


Cell Membrane/metabolism , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mutation , Adrenal Gland Neoplasms/genetics , Adrenal Gland Neoplasms/metabolism , Amino Acid Substitution , Gene Knockdown Techniques , Genetic Predisposition to Disease , Germ-Line Mutation , HEK293 Cells , Humans , Membrane Proteins/chemistry , Mutagenesis, Site-Directed , Mutation/physiology , Paraganglioma/genetics , Paraganglioma/metabolism , Pheochromocytoma/genetics , Pheochromocytoma/metabolism , Protein Transport/genetics , Tissue Distribution
4.
Hum Mol Genet ; 27(10): 1794-1808, 2018 05 15.
Article En | MEDLINE | ID: mdl-29547888

The TMEM127 tumor suppressor gene encodes a transmembrane protein of unknown function mutated in pheochromocytomas and, rarely, in renal cancers. Tumors with inactivating TMEM127 mutations have increased mTORC1 signaling by undefined mechanisms. Here we report that TMEM127 interacts with the lysosome-anchored complex comprised of Rag GTPases, the LAMTOR pentamer (or 'ragulator') and vATPase, which controls amino acid-mediated mTORC1 activation. We found that under nutrient-rich conditions TMEM127 expression reduces mTORC1 recruitment to Rags. In addition, TMEM127 interacts with LAMTOR in an amino acid-dependent manner and decreases the LAMTOR1-vATPase association, while TMEM127-vATPase binding requires intact lysosomal acidification but is amino acid independent. Conversely, both murine and human cells lacking TMEM127 accumulate LAMTOR proteins in the lysosome. Consistent with these findings, pheochromocytomas with TMEM127 mutations have increased levels of LAMTOR proteins. These results suggest that TMEM127 interactions with ragulator and vATPase at the lysosome contribute to restrain mTORC1 signaling in response to amino acids, thus explaining the increased mTORC1 activation seen in TMEM127-deficient tumors.


Adrenal Gland Neoplasms/genetics , Carrier Proteins/genetics , Membrane Proteins/genetics , Pheochromocytoma/genetics , Adrenal Gland Neoplasms/metabolism , Adrenal Gland Neoplasms/pathology , Amino Acids/genetics , Animals , Gene Expression Regulation , Genes, Tumor Suppressor , Humans , Intracellular Signaling Peptides and Proteins , Lysosomes/genetics , Lysosomes/metabolism , Mechanistic Target of Rapamycin Complex 1/genetics , Mice , Multiprotein Complexes/genetics , Mutation , Pheochromocytoma/metabolism , Pheochromocytoma/pathology , Signal Transduction
5.
Langmuir ; 34(15): 4673-4680, 2018 04 17.
Article En | MEDLINE | ID: mdl-29363972

Artificial lipid membranes incorporating proteins have frequently been used as models for the dynamic organization of biological structures in living cells as well as in the development of biology-inspired technologies. We report here on the experimental demonstration and characterization of a pattern-forming process that occurs in a lipid bilayer membrane adhered via biotin-avidin binding to a second lipid membrane that is supported by a solid substrate. Adhesion regions are roughly circular with a diameter of about 25 µm. Using confocal fluorescence microscopy, we record time series of dynamic fingering patterns that grow in the upper lipid membrane and intermembrane biotin-avidin bonds. The fingers are micrometer-scale elongated pores that grow from the edge of an already-stabilized hole. Finger growth is saltatory on the scale of tens of seconds. We find that as the fingers grow and the density of adhesion proteins increases, the rate of finger growth decreases exponentially and the width of newly formed fingers decreases linearly. We show that these findings are consistent with a thermodynamic description of dynamic pore formation and stabilization.


Lipid Bilayers/chemistry , Membranes, Artificial , Avidin/metabolism , Biotin/metabolism , Protein Binding , Thermodynamics
6.
Matrix Biol ; 52-54: 426-441, 2016.
Article En | MEDLINE | ID: mdl-26780725

For more than 100years, cells and tissues have been studied in vitro using glass and plastic surfaces. Over the last 10-20years, a great body of research has shown that cells are acutely sensitive to their local environment (extracellular matrix, ECM) which contains both chemical and physical cues that influence cell behavior. These observations suggest that modern cell culture systems, using tissue culture polystyrene (TCP) surfaces, may fail to reproduce authentic cell behavior in vitro, resulting in "artificial outcomes." In the current study, we use bone marrow (BM)- and adipose (AD)-derived stromal cells to prepare BM-ECM and AD-ECM, which are decellularized after synthesis by the cells, to mimic the cellular niche for each of these tissues. Each ECM was characterized for its ability to affect BM- and AD-mesenchymal stem cell (MSC) proliferation, as well as proliferation of three cancer cell lines (HeLa, MCF-7, and MDA-MB-231), modulate cell spreading, and direct differentiation relative to standard TCP surfaces. We found that both ECMs promoted the proliferation of MSCs, but that this effect was enhanced when the tissue-origin of the cells matched that of the ECM (i.e. BM-ECM promoted the proliferation of BM-MSCs over AD-MSCs, and vice versa). Moreover, BM- and AD-ECM were shown to preferentially direct MSC differentiation towards either osteogenic or adipogenic lineage, respectively, suggesting that the effects of the ECM were tissue-specific. Further, each ECM influenced cell morphology (i.e. circularity), irrespective of the origin of the MSCs, lending more support to the idea that effects were tissue specific. Interestingly, unlike MSCs, these ECMs did not promote the proliferation of the cancer cells. In an effort to further understand how these three culture substrates influence cell behavior, we evaluated the chemical (protein composition) and physical properties (architecture and mechanical) of the two ECMs. While many structural proteins (e.g. collagen and fibronectin) were found at equivalent levels in both BM- and AD-ECM, the architecture (i.e. fiber orientation; surface roughness) and physical properties (storage modulus, surface energy) of each were unique. These results, demonstrating differences in cell behavior when cultured on the three different substrates (BM- and AD-ECM and TCP) with differences in chemical and physical properties, provide evidence that the two ECMs may recapitulate specific elements of the native stem cell niche for bone marrow and adipose tissues. More broadly, it could be argued that ECMs, elaborated by cells ex vivo, serve as an ideal starting point for developing tissue-specific culture environments. In contrast to TCP, which relies on the "one size fits all" paradigm, native tissue-specific ECM may be a more rational model to approach engineering 3D tissue-specific culture systems to replicate the in vivo niche. We suggest that this approach will provide more meaningful information for basic research studies of cell behavior as well as cell-based therapeutics.


Adipose Tissue/cytology , Bone Marrow Cells/cytology , Cell Culture Techniques/methods , Extracellular Matrix/metabolism , Mesenchymal Stem Cells/cytology , Adipose Tissue/metabolism , Bone Marrow Cells/metabolism , Cell Differentiation , Cell Line, Tumor , Cell Proliferation , HeLa Cells , Humans , MCF-7 Cells , Mesenchymal Stem Cells/metabolism , Polystyrenes/chemistry , Stem Cell Niche
7.
Intravital ; 2(1)2014 Mar 01.
Article En | MEDLINE | ID: mdl-25313346

Maximizing 2-photon parameters used in acquiring images for quantitative intravital microscopy, especially when high sensitivity is required, remains an open area of investigation. Here we present data on correctly setting the black level of the photomultiplier tube amplifier by adjusting the offset to allow for accurate quantitation of low intensity processes. When the black level is set too high some low intensity pixel values become zero and a nonlinear degradation in sensitivity occurs rendering otherwise quantifiable low intensity values virtually undetectable. Initial studies using a series of increasing offsets for a sequence of concentrations of fluorescent albumin in vitro revealed a loss of sensitivity for higher offsets at lower albumin concentrations. A similar decrease in sensitivity, and therefore the ability to correctly determine the glomerular permeability coefficient of albumin, occurred in vivo at higher offset. Finding the offset that yields accurate and linear data are essential for quantitative analysis when high sensitivity is required.

8.
Apoptosis ; 19(5): 816-28, 2014 May.
Article En | MEDLINE | ID: mdl-24375173

The signaling pathways via mTOR (mammalian target of rapamycin) and AMPK (AMP-activated protein kinase) play key roles in transcription, translation and carcinogenesis, and may be activated by light exposure. These pathways can be modulated by naturally occurring compounds, such as the triterpenoid, ursolic acid (UA). Previously, the transcription factors p53 and NF-κB, which transactivate mitochondrial apoptosis-related genes, were shown to be differentially modulated by UA. UA-modulated apoptosis, following exposure to UV-VIS radiation (ultraviolet to visible light broadband radiation, hereafter abbreviated to UVR), is observed to correspond to differential levels of oxidative stress in retinal pigment epithelial (RPE) and skin melanoma (SM) cells. The cellular response to this phytochemical was characterized using western blot, flow cytometry, microscopy with reactive oxidative species probes MitoTracker and dihydroethidium, and membrane permeability assay. UA pretreatment potentiated cell cycle arrest and UVR-induced apoptosis selectively in SM cells while reducing photo-oxidative stress in the DNA of RPE cells presumably by antioxidant activity of UA. Mechanistically, the nuclear transportation of p65 and p53 was reduced by UA administration prior to UVR exposure while the levels of p65 and p53 nuclear transportation in SM cells were sustained at a substantially higher level. Finally, the mitochondrial functional assay showed that UVR induced the collapse of the mitochondrial membrane potential, and this effect was exacerbated by rapamycin or UA pretreatment in SM preferentially. These results were consistent with reduced proliferation observed in the clonogenic assay, indicating that UA treatment enhanced the phototoxicity of UVR, by modulating the activation of p53 and NF-κB and initiating a mitogenic response to optical radiation that triggered mitochondria-dependent apoptosis, particularly in skin melanoma cells. The study indicates that this compound has multiple actions with the potential for protecting normal cells while sensitizing skin melanoma cells to UV irradiation.


Antineoplastic Agents, Phytogenic/pharmacology , Antioxidants/pharmacology , Apoptosis/drug effects , Light/adverse effects , Melanoma/metabolism , Retinal Pigment Epithelium/drug effects , Skin Neoplasms/metabolism , Triterpenes/pharmacology , Antibiotics, Antineoplastic/pharmacology , Antineoplastic Agents, Phytogenic/toxicity , Antioxidants/toxicity , Apoptosis/radiation effects , Cell Cycle Checkpoints , Cell Line , Cell Proliferation/drug effects , Cell Proliferation/radiation effects , Humans , Melanoma/pathology , Mitochondria/metabolism , Reactive Oxygen Species/metabolism , Retinal Pigment Epithelium/cytology , Retinal Pigment Epithelium/radiation effects , Sirolimus/pharmacology , Skin Neoplasms/pathology , Transcription Factor RelA/metabolism , Triterpenes/toxicity , Tumor Suppressor Protein p53/metabolism , Ultraviolet Rays/adverse effects , Ursolic Acid , Melanoma, Cutaneous Malignant
9.
J Am Soc Nephrol ; 23(3): 447-57, 2012 Mar.
Article En | MEDLINE | ID: mdl-22223875

Different laboratories recently reported incongruous results describing the quantification of albumin filtration using two-photon microscopy. We investigated the factors that influence the glomerular sieving coefficient for albumin (GSC(A)) in an effort to explain these discordant reports and to develop standard operating procedures for determining GSC(A). Multiple factors influenced GSC(A), including the kidney depth of image acquisition (10-20 µm was appropriate), the selection of fluorophore (probes emitting longer wavelengths were superior), the selection of plasma regions for fluorescence measurements, the size and molecular dispersion characteristics of dextran polymers if used, dietary status, and the genetic strain of rat. Fasting reduced the GSC(A) in Simonsen Munich Wistar rats from 0.035±0.005 to 0.016±0.004 (P<0.01). Frömter Munich Wistar rats had a much lower GSC(A) in both the fed and the fasted states. Finally, we documented extensive albumin transcytosis with vesicular and tubular delivery to and fusion with the basolateral membrane in S1 proximal tubule cells. In summary, these results help explain the previously conflicting microscopy and micropuncture data describing albumin filtration and highlight the dynamic nature of glomerular albumin permeability.


Albumins/metabolism , Cell Membrane Permeability/physiology , Kidney Glomerulus/cytology , Kidney Glomerulus/physiology , Kidney Tubules, Proximal/cytology , Kidney Tubules, Proximal/physiology , Animals , Dextrans , Diet , Female , Fluorescent Dyes , Male , Microscopy, Fluorescence, Multiphoton , Models, Animal , Punctures , Rats , Rats, Sprague-Dawley , Rats, Wistar , Time Factors
10.
Mol Pharm ; 9(2): 310-7, 2012 Feb 06.
Article En | MEDLINE | ID: mdl-22171616

Due to the overexpression of a folate receptor (FR) on many malignant cells, folate-targeted drugs have been developed to improve the cancer specificity of chemotherapeutic agents. Therapeutic index is further enhanced with the use of self-immolative linkers that efficiently release the attached drug upon cellular internalization of the folate-drug conjugate. Because FR is also abundant in normal kidney proximal tubule (PT) cells, we sought to examine in real time the trafficking and release of folate-targeted drugs in the kidney in vivo. Thus, we conducted two-photon kidney imaging studies in mice utilizing a Förster resonance energy transfer (FRET) based folate conjugate that undergoes a color shift from red to green upon reduction of the disulfide bond linking folate to a surrogate drug molecule. Following infusion via intravenous injection, folate-FRET reached the kidney in its intact unreduced form. The folate-FRET conjugate was then filtered into the lumen of PT, where it was efficiently captured by FR. As FR transcytosed across PT, some disulfide reduction occurred, with reduced folate-FRET detectable in PT vesicles 30 min postinjection. Prolonged monitoring of folate-FRET in mice showed modest progression of reduction in PT cells over time. Moreover, inhibition of FR trafficking in PT cells by colchicine did not significantly affect the rate or extent of folate-FRET reduction. Finally, the lack of cytosolic accumulation of released drug surrogate in the PT suggests that drug release via disulfide bond reduction should cause little kidney toxicity.


Antineoplastic Agents/pharmacokinetics , Boron Compounds/pharmacokinetics , Drug Carriers/pharmacokinetics , Folate Receptors, GPI-Anchored/metabolism , Folic Acid/analogs & derivatives , Kidney Tubules, Proximal/metabolism , Rhodamines/pharmacokinetics , Animals , Antineoplastic Agents/chemistry , Boron Compounds/chemistry , Disulfides/chemistry , Drug Carriers/chemistry , Fluorescence Resonance Energy Transfer , Folate Receptors, GPI-Anchored/drug effects , Folic Acid/chemistry , Folic Acid/pharmacokinetics , Kidney/metabolism , Male , Mice , Mice, Nude , Oxidation-Reduction , Protein Transport , Rhodamines/chemistry
11.
Kidney Int ; 81(1): 112-7, 2012 Jan.
Article En | MEDLINE | ID: mdl-21881552

Measurement of the glomerular filtration rate (GFR) is the gold standard for precise assessment of kidney function. A rapid, point-of-care determination of the GFR may provide advantages in the clinical setting over currently available assays. Here we demonstrate a proof of principle for such an approach in a pig and dogs, two species that approximate the vascular access and GFR results expected in humans. In both animal models, a sub-millimeter optical fiber that delivered excitation light and collected fluorescent emissions was inserted into a peripheral vein (dog) or central venous access (pig) by means of commercial intravenous catheters. A mixture of fluorescent chimeras of a small freely filterable reporter and large non-filterable plasma volume marker were infused as a bolus, excited by light-emitting diodes, and the in vivo signals detected and quantified by photomultiplier tubes in both species in less than 60 min. Concurrent standardized 6-h iohexol plasma kidney clearances validated the accuracy of our results for both physiologic and a chronic kidney disease setting. Thus, our ratiometric technique allows for both measurement of plasma vascular volume and highly accurate real-time GFR determinations, enabling clinical decision making in real time.


Glomerular Filtration Rate , Kidney Function Tests/veterinary , Optical Fibers , Animals , Dogs , Equipment Design , Fluorescent Dyes , Iohexol , Kidney Function Tests/instrumentation , Point-of-Care Systems , Radiometry/instrumentation , Radiometry/veterinary , Swine
12.
Am J Physiol Renal Physiol ; 299(5): F1048-55, 2010 Nov.
Article En | MEDLINE | ID: mdl-20685826

The rapid diagnosis and quantification of acute kidney injury (AKI) severity remain high clinical priorities. By combining intravital fluorescent ratiometric two-photon kidney imaging and the two-compartment pharmacokinetics model, we demonstrate that rapid quantification of glomerular filtration rate (GFR) can be achieved in physiologic and AKI rat kidney models. Using a bolus infusion of a mixture of FITC-inulin and a 500-kDa Texas Red dextran, a full spectrum of GFR values, ranging from 0.17 to 1.12 ml·min(-1)·100 g(-1), was obtained. The GFR values thus determined correlated well with values obtained by the standard 2-h inulin infusion clearance method with a Pearson's correlation coefficient of 0.85. In addition, postischemia deterioration was studied by measuring GFR using the two-photon approach during 24 h following a 45-min bilateral ischemia clamp model. The GFR was found to decline sharply during the initial 4 h followed by a nadir with little sign of rising over the ensuing 24-h period. Moreover, a FITC-labeled 5-kDa dextran was identified as having nearly identical filtration characteristics as FITC-inulin, but had markedly increased fluorescent intensity, thus minimizing the quantity needed for individual studies. The technique reported allows for very rapid GFR determinations, within 10-15 min, based on plasma clearance of a freely filtered fluorescence probe, instead of a prolonged one-compartment interstitial space reporter molecule clearance employed by other technologies.


Acute Kidney Injury/diagnosis , Glomerular Filtration Rate/physiology , Kidney Function Tests/methods , Acute Kidney Injury/chemically induced , Acute Kidney Injury/pathology , Algorithms , Animals , Data Interpretation, Statistical , Female , Fluorescein-5-isothiocyanate , Gentamicins , Image Processing, Computer-Assisted , Inulin/metabolism , Ischemia/pathology , Lipopolysaccharides , Male , Microscopy, Confocal , Protein Synthesis Inhibitors , Rats , Rats, Sprague-Dawley
13.
J Biol Chem ; 283(51): 35579-89, 2008 Dec 19.
Article En | MEDLINE | ID: mdl-18922801

Glomerular injury is often characterized by the effacement of podocytes, loss of slit diaphragms, and proteinuria. Renal ischemia or the loss of blood flow to the kidneys has been widely associated with tubular and endothelial injury but rarely has been shown to induce podocyte damage and disruption of the slit diaphragm. In this study, we have used an in vivo rat ischemic model to demonstrate that renal ischemia induces podocyte effacement with loss of slit diaphragm and proteinuria. Biochemical analysis of the ischemic glomerulus shows that ischemia induces rapid loss of interaction between slit diaphragm junctional proteins Neph1 and ZO-1. To further understand the effect of ischemia on molecular interactions between slit diaphragm proteins, a cell culture model was employed to study the binding between Neph1 and ZO-1. Under physiologic conditions, Neph1 co-localized with ZO-1 at cell-cell contacts in cultured human podocytes. Induction of injury by ATP depletion resulted in rapid loss of Neph1 and ZO-1 binding and redistribution of Neph1 and ZO-1 proteins from cell membrane to the cytoplasm. Recovery resulted in increased Neph1 tyrosine phosphorylation, restoring Neph1 and ZO-1 binding and their localization at the cell membrane. We further demonstrate that tyrosine phosphorylation of Neph1 mediated by Fyn results in significantly increased Neph1 and ZO-1 binding, suggesting a critical role for Neph1 tyrosine phosphorylation in reorganizing the Neph1-ZO-1 complex. This study documents that renal ischemia induces dynamic changes in the molecular interactions between slit diaphragm proteins, leading to podocyte damage and proteinuria.


Ischemia/metabolism , Kidney Diseases/metabolism , Membrane Proteins/metabolism , Multiprotein Complexes/metabolism , Phosphoproteins/metabolism , Podocytes/metabolism , Animals , Cell Line , Cell Membrane/metabolism , Disease Models, Animal , Humans , Male , Phosphorylation , Podocytes/pathology , Protein Binding , Proteinuria/metabolism , Proto-Oncogene Proteins c-fyn/metabolism , Rats , Rats, Sprague-Dawley , Zonula Occludens-1 Protein
14.
Traffic ; 8(7): 932-49, 2007 Jul.
Article En | MEDLINE | ID: mdl-17547697

The Rho family of GTPases is implicated in the control of endocytic and biosynthetic traffic of many cell types; however, the cellular distribution of RhoB remains controversial and its function is not well understood. Using confocal microscopy, we found that endogenous RhoB and green fluorescent protein-tagged wild-type RhoB were localized to early endosomes, and to a much lesser extent to recycling endosomes, late endosomes or Golgi complex of fixed or live polarized Madin-Darby canine kidney cells. Consistent with RhoB localization to early endosomes, we observed that expression of dominant-negative RhoBN19 or dominant-active RhoBV14 altered postendocytic traffic of ligand-receptor complexes that undergo recycling, degradation or transcytosis. In vitro assays established that RhoB modulated the basolateral-to-apical transcytotic pathway by regulating cargo exit from basolateral early endosomes. Our results indicate that RhoB is localized, in part, to early endosomes where it regulates receptor egress through the early endocytic system.


Endosomes/metabolism , rhoB GTP-Binding Protein/metabolism , rhoB GTP-Binding Protein/physiology , Actins/metabolism , Animals , Biological Transport , Cloning, Molecular , Cytoskeleton/metabolism , Cytosol/metabolism , DNA/metabolism , Dogs , Endocytosis , GTP Phosphohydrolases/metabolism , Immunoglobulin A/metabolism , Mutation , Time Factors
15.
Am J Physiol Renal Physiol ; 292(2): F599-606, 2007 Feb.
Article En | MEDLINE | ID: mdl-16968889

Fetuin-A is a known inhibitor of vascular calcification in vitro. In arteries with calcification, there is increased immunostaining for fetuin-A. However, vascular smooth muscle cells (VSMC) do not synthesize fetuin-A, suggesting fetuin-A may be endocytosed to exert its inhibitory effects. To examine the mechanism by which fetuin-A is taken up in bovine VSMC (BVSMC), we examined living cells by confocal microscopy and determined the uptake of Cy5-labeled fetuin-A. The results demonstrated that fetuin-A was taken up in BVSMC only in the presence of extracellular calcium, whereas phosphorus had no effect. Additional studies demonstrated the calcium-dependent uptake was specific for fetuin-A and only observed in BVSMC and osteoblasts, but not epithelial, endothelial, or adipose cells. The uptake was dose dependent, but could not be inhibited by excess unlabeled fetuin-A, suggesting a fluid phase rather than a receptor-mediated process. Fetuin-A also induced a sustained increase in intracellular calcium in BVSMC in the presence of extracellular calcium, whereas there was no increase in the absence of extracellular calcium. To further characterize the uptake, we utilized an inhibitor of annexin calcium channel activity, demonstrating inhibition of both fetuin-A uptake and intracellular calcium increase. Finally, we demonstrate that fetuin-A binds to annexin II at the cell membrane of BVSMC. In summary, our study demonstrates calcium- and annexin-dependent uptake of fetuin-A that leads to a sustained rise in intracellular calcium. This regulated uptake may be a mechanism by which fetuin-A inhibits VSMC calcification in the presence of excess calcium.


Annexins/physiology , Calcium/physiology , Muscle, Smooth, Vascular/metabolism , alpha-Fetoproteins/metabolism , Animals , Calcium Channels/drug effects , Cattle , Cells, Cultured , Endocytosis , Ionomycin/pharmacology , Muscle, Smooth, Vascular/cytology , Thiazepines/pharmacology
17.
Ann Biomed Eng ; 34(10): 1600-6, 2006 Oct.
Article En | MEDLINE | ID: mdl-17029032

Mechanical loading is well known to stimulate bone remodeling. Load-driven interstitial fluid flow and molecular transport have been postulated to play a role in the enhancement of bone formation. In order to evaluate load-driven molecular transport in a lacunocanalicular network, we conducted fluorescence recovery after photobleaching (FRAP) experiments using lacunae stained with uranine (376 Da). Loads were applied to a mouse femur ex vivo with a novel knee-loading modality, where the distal epiphysis was loaded with a sinusoidal force at 2 Hz. The lacunae in the diaphysis located 25% (approximately 4 mm) proximal to the loading site were photobleached and sequentially imaged, and a time constant for fluorescence recovery was determined both with and without knee loading. The time constant was estimated as the period to recover 63% of fluorescent intensity using a best-fit exponential curve. The results reveal that the applied loads shortened the time constant from 33 +/- 9 s with non-loading control to 25 +/- 11 s with knee loading (p = 0.0014). The strain in the measurement site was <100 microstain along the femoral midshaft, which was an order of magnitude smaller than the minimum effective strain threshold for bone remodeling. Taken together, the current study supports the notion that molecular transport in cortical bone is enhanced by the loads applied to the epiphysis without inducing significant in situ strain in the diaphysis.


Femur/anatomy & histology , Femur/physiology , Animals , Biological Transport, Active , Biomechanical Phenomena , Biomedical Engineering , Diffusion , Epiphyses/anatomy & histology , Epiphyses/physiology , Female , Fluorescence Recovery After Photobleaching , In Vitro Techniques , Mice , Mice, Inbred C57BL , Stress, Mechanical
18.
Mol Biol Cell ; 17(7): 3156-75, 2006 Jul.
Article En | MEDLINE | ID: mdl-16641372

Rab10, a protein originally isolated from Madin-Darby Canine Kidney (MDCK) epithelial cells, belongs to a family of Rab proteins that includes Rab8 and Rab13. Although both Rab8 and Rab13 have been found to mediate polarized membrane transport, the function of Rab10 in mammalian cells has not yet been established. We have used quantitative confocal microscopy of polarized MDCK cells expressing GFP chimeras of wild-type and mutant forms of Rab10 to analyze the function of Rab10 in polarized cells. These studies demonstrate that Rab10 is specifically associated with the common endosomes of MDCK cells, accessible to endocytic probes internalized from either the apical or basolateral plasma membrane domains. Expression of mutant Rab10 defective for either GTP hydrolysis or GTP binding increased recycling from early compartments on the basolateral endocytic pathway without affecting recycling from later compartments or the apical recycling pathway. These results suggest that Rab10 mediates transport from basolateral sorting endosomes to common endosomes.


Cell Membrane/metabolism , Endosomes/metabolism , rab GTP-Binding Proteins/metabolism , Animals , Cell Polarity , Dogs , Endosomes/chemistry , Green Fluorescent Proteins/analysis , Green Fluorescent Proteins/genetics , Guanosine Triphosphate/metabolism , Hydrolysis , Immunoglobulin A/metabolism , Intracellular Membranes/chemistry , Intracellular Membranes/metabolism , Kidney/metabolism , Kidney/ultrastructure , Microscopy, Confocal , Mutation , Protein Structure, Tertiary , Protein Transport , Transport Vesicles/metabolism , rab GTP-Binding Proteins/analysis , rab GTP-Binding Proteins/genetics , trans-Golgi Network/chemistry , trans-Golgi Network/metabolism
19.
Virology ; 310(1): 100-8, 2003 May 25.
Article En | MEDLINE | ID: mdl-12788634

Major histocompatibility complex (MHC) class II antigens are expressed on human foreskin keratinocytes (HFKs) following exposure to interferon gamma. The expression of MHC class II proteins on the cell surface may allow keratinocytes to function as antigen-presenting cells and induce a subsequent immune response to virus infection. Invariant chain (Ii) is a chaperone protein which plays an important role in the maturation of MHC class II molecules. The sequential degradation of Ii within acidic endocytic compartments is a key process required for the successful loading of antigenic peptide onto MHC class II molecules. Since human papillomavirus (HPV) 16 E5 can inhibit the acidification of late endosomes in HFKs, the E5 protein may be able to affect proper peptide loading onto the MHC class II molecule. To test this hypothesis, HFKs were infected with either control virus or a recombinant virus expressing HPV16 E5 and the infected cells were subsequently treated with interferon-gamma. ELISAs revealed a decrease of MHC class II expression on the surface of E5-expressing cells compared with control virus-infected cells after interferon treatment. Western blot analysis showed that, in cells treated with interferon gamma, E5 could prevent the breakdown of Ii and block the formation of peptide-loaded, SDS-stable mature MHC class II dimers, correlating with diminished surface MHC class II expression. These data suggest that HPV16 E5 may be able to decrease immune recognition of infected keratinocytes via disruption of MHC class II protein function.


Histocompatibility Antigens Class II/metabolism , Interferon-gamma/pharmacology , Keratinocytes/immunology , Oncogene Proteins, Viral/physiology , Antigens, Differentiation, B-Lymphocyte/metabolism , Cells, Cultured , Dimerization , Down-Regulation , Histocompatibility Antigens Class II/analysis , Histocompatibility Antigens Class II/chemistry , Humans
20.
Am J Physiol Cell Physiol ; 284(6): C1654-68, 2003 Jun.
Article En | MEDLINE | ID: mdl-12584115

The ability to image calcium signals at subcellular levels within the intact depolarizing heart could provide valuable information toward a more integrated understanding of cardiac function. Accordingly, a system combining two-photon excitation with laser-scanning microscopy was developed to monitor electrically evoked [Ca(2+)](i) transients in individual cardiomyocytes within noncontracting Langendorff-perfused mouse hearts. [Ca(2+)](i) transients were recorded at depths

Calcium Signaling/physiology , Calcium/metabolism , Diacetyl/analogs & derivatives , Diagnostic Imaging/methods , Egtazic Acid/analogs & derivatives , Heart/physiology , Animals , Chelating Agents/metabolism , Cytochalasin D/metabolism , Diacetyl/metabolism , Egtazic Acid/metabolism , Enzyme Inhibitors/metabolism , Fluorescent Dyes/metabolism , Fura-2/metabolism , Heterocyclic Compounds, 3-Ring , In Vitro Techniques , Mice , Mice, Inbred Strains , Mice, Transgenic , Muscle Cells/cytology , Muscle Cells/metabolism , Muscle Contraction/physiology , Nucleic Acid Synthesis Inhibitors/metabolism , Perfusion , Photons , Transforming Growth Factor beta/genetics , Transforming Growth Factor beta/metabolism , Transforming Growth Factor beta1
...