Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 117
Filter
Add more filters










Publication year range
1.
Sci Rep ; 13(1): 17276, 2023 10 12.
Article in English | MEDLINE | ID: mdl-37828038

ABSTRACT

Megalin/LRP2 is a major receptor supporting apical endocytosis in kidney proximal tubular cells. We have previously reported that kidney-specific perinatal ablation of the megalin gene in cystinotic mice, a model of nephropathic cystinosis, essentially blocks renal cystine accumulation and partially preserves kidney tissue integrity. Here, we examined whether inhibition of the megalin pathway in adult cystinotic mice by dietary supplementation (5x-fold vs control regular diet) with the dibasic amino-acids (dAAs), lysine or arginine, both of which are used to treat patients with other rare metabolic disorders, could also decrease renal cystine accumulation and protect cystinotic kidneys. Using surface plasmon resonance, we first showed that both dAAs compete for protein ligand binding to immobilized megalin in a concentration-dependent manner, with identical inhibition curves by L- and D-stereoisomers. In cystinotic mice, 2-month diets with 5x-L-lysine and 5x-L-arginine were overall well tolerated, while 5x-D-lysine induced strong polyuria but no weight loss. All diets induced a marked increase of dAA urinary excretion, most prominent under 5x-D-lysine, without sign of kidney insufficiency. Renal cystine accumulation was slowed down approx. twofold by L-dAAs, and totally suppressed by D-lysine. We conclude that prolonged dietary manipulation of the megalin pathway in kidneys is feasible, tolerable and can be effective in vivo.


Subject(s)
Cystine , Cystinosis , Adult , Humans , Animals , Mice , Cystine/metabolism , Cystinosis/metabolism , Lysine , Low Density Lipoprotein Receptor-Related Protein-2 , Kidney/metabolism , Dietary Supplements
2.
Endocrinology ; 157(4): 1363-71, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26812160

ABSTRACT

Hypothyroidism is the most frequent and earliest endocrine complication in cystinosis, a multisystemic lysosomal storage disease caused by defective transmembrane cystine transporter, cystinosin (CTNS gene). We recently demonstrated in Ctns(-/-) mice that altered thyroglobulin biosynthesis associated with endoplasmic reticulum stress, combined with defective lysosomal processing, caused hypothyroidism. In Ctns(-/-) kidney, hematopoietic stem cell (HSC) transplantation provides long-term functional and structural protection. Tissue repair involves transfer of cystinosin-bearing lysosomes from HSCs differentiated as F4/80 macrophages into deficient kidney tubular cells, via tunneling nanotubes that cross basement laminae. Here we evaluated the benefit of HSC transplantation for cystinotic thyroid and investigated the underlying mechanisms. HSC engraftment in Ctns(-/-) thyroid drastically decreased cystine accumulation, normalized the TSH level, and corrected the structure of a large fraction of thyrocytes. In the thyroid microenvironment, HSCs differentiated into a distinct, mixed macrophage/dendritic cell lineage expressing CD45 and major histocompatibility complex II but low CD11b and F4/80. Grafted HSCs closely apposed to follicles and produced tunneling nanotube-like extensions that crossed follicular basement laminae. HSCs themselves further squeezed into follicles, allowing extensive contact with thyrocytes, but did not transdifferentiate into Nkx2.1-expressing cells. Our observations revealed significant differences of basement lamina porosity between the thyroid and kidney and/or intrinsic macrophage invasive properties once in the thyroid microenvironment. The contrast between extensive thyrocyte protection and low HSC abundance at steady state suggests multiple sequential encounters and/or remanent impact. This is the first report demonstrating the potential of HSC transplantation to correct thyroid disease and supports a major multisystemic benefit of stem cell therapy for cystinosis.


Subject(s)
Cystinosis/therapy , Disease Models, Animal , Hematopoietic Stem Cell Transplantation/methods , Thyroid Gland/physiopathology , Amino Acid Transport Systems, Neutral/genetics , Amino Acid Transport Systems, Neutral/metabolism , Animals , Cell Differentiation , Cystine/metabolism , Cystinosis/genetics , Cystinosis/physiopathology , Female , Hematopoietic Stem Cells/metabolism , Humans , Luminescent Proteins/genetics , Luminescent Proteins/metabolism , Lysosomes/metabolism , Macrophages/metabolism , Male , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Microscopy, Confocal , Thyroid Gland/metabolism , Thyrotropin/metabolism , Transplantation, Homologous
3.
Endocrinology ; 156(6): 2349-64, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25811319

ABSTRACT

Thyroid hormones are released from thyroglobulin (Tg) in lysosomes, which are impaired in infantile/nephropathic cystinosis. Cystinosis is a lysosomal cystine storage disease due to defective cystine exporter, cystinosin. Cystinotic children develop subclinical and then overt hypothyroidism. Why hypothyroidism is the most frequent and earliest endocrine complication of cystinosis is unknown. We here defined early alterations in Ctns(-/-) mice thyroid and identified subcellular and molecular mechanisms. At 9 months, T4 and T3 plasma levels were normal and TSH was moderately increased (∼4-fold). By histology, hyperplasia and hypertrophy of most follicles preceded colloid exhaustion. Increased immunolabeling for thyrocyte proliferation and apoptotic shedding indicated accelerated cell turnover. Electron microscopy revealed endoplasmic reticulum (ER) dilation, apical lamellipodia indicating macropinocytic colloid uptake, and lysosomal cystine crystals. Tg accumulation in dilated ER contrasted with mRNA down-regulation. Increased expression of ER chaperones, glucose-regulated protein of 78 kDa and protein disulfide isomerase, associated with alternative X-box binding protein-1 splicing, revealed unfolded protein response (UPR) activation by ER stress. Decreased Tg mRNA and ER stress suggested reduced Tg synthesis. Coordinated increase of UPR markers, activating transcription factor-4 and C/EBP homologous protein, linked ER stress to apoptosis. Hormonogenic cathepsins were not altered, but lysosome-associated membrane protein-1 immunolabeling disclosed enlarged vesicles containing iodo-Tg and impaired lysosomal fusion. Isopycnic fractionation showed iodo-Tg accumulation in denser lysosomes, suggesting defective lysosomal processing and hormone release. In conclusion, Ctns(-/-) mice showed the following alterations: 1) compensated primary hypothyroidism and accelerated thyrocyte turnover; 2) impaired Tg production linked to ER stress/UPR response; and 3) altered endolysosomal trafficking and iodo-Tg processing. The Ctns(-/-) thyroid is useful to study disease progression and evaluate novel therapies.


Subject(s)
Cystinosis/metabolism , Cystinosis/pathology , Endoplasmic Reticulum Stress/physiology , Lysosomes/metabolism , Thyroglobulin/biosynthesis , Unfolded Protein Response/physiology , Amino Acid Transport Systems, Neutral/genetics , Amino Acid Transport Systems, Neutral/metabolism , Animals , Female , Male , Mice
4.
Neurodegener Dis ; 10(1-4): 92-5, 2012.
Article in English | MEDLINE | ID: mdl-22343802

ABSTRACT

BACKGROUND: The two major isoforms of the human amyloid precursor protein (APP) are APP695 and APP751. They differ by the insertion of a Kunitz-type protease inhibitor (KPI) sequence in the extracellular domain of APP751. APP-KPI isoforms are increased in Alzheimer's disease brains, and they could be associated with disease progression. Recent studies have shown that APP processing to Aß is regulated by homodimerization, which involves both extracellular and juxtamembrane/transmembrane (JM/TM) regions. OBJECTIVE: Our aim is to understand the mechanisms controlling APP dimerization and the contribution of the ectodomain and JM/TM regions to this process. METHODS: We used bimolecular fluorescence complementation approaches coupled to fluorescence-activated cell sorting analysis to measure the dimerization level of different APP isoforms and APP C-terminal fragments (C99) mutated in their JM/TM region. RESULTS: APP751 was found to form significantly more homodimers than APP695. Mutation of dimerization motifs in the TM domain of APP or C99 did not significantly affect fluorescence complementation. CONCLUSION: These findings indicate that the KPI domain plays a major role in APP dimerization. They set the basis for further investigation of the relation between dimerization, metabolism and function of APP.


Subject(s)
Amyloid beta-Protein Precursor/metabolism , Dimerization , Protease Inhibitors/metabolism , Protein Multimerization/physiology , Amyloid beta-Protein Precursor/genetics , Animals , Bacterial Proteins/genetics , COS Cells , Chlorocebus aethiops , Flow Cytometry , Humans , Luminescent Proteins/genetics , Mutation/genetics , Protein Isoforms/genetics , Protein Isoforms/metabolism , Protein Multimerization/genetics , Protein Processing, Post-Translational , Protein Structure, Tertiary , Sequence Homology, Amino Acid , Transfection
5.
Exp Cell Res ; 316(19): 3239-53, 2010 Nov 15.
Article in English | MEDLINE | ID: mdl-20832399

ABSTRACT

Src, a non-receptor tyrosine kinase, is a key signal transduction partner of epidermal growth factor (EGF) receptor (EGFR). In human breast cancer, EGFR and Src are frequently over-expressed and/or over-activated. Although reciprocal activation is documented, mechanisms underlying Src:EGFR interactions are incompletely understood. We here exploited ts/v-Src thermo-activation in MDCK monolayers to test whether acute Src activation impacts on signalling and trafficking of non-liganded EGFR. We found that thermo-activation caused rapid Src recruitment to the plasma membrane, concomitant association with EGFR, and its phosphorylation at Y845 and Y1173 predominantly at the cell surface. Like low EGF concentrations, activated Src (i) decreased EGF surface binding without affecting the total EGFR pool; (ii) triggered EGFR endocytosis via clathrin-coated vesicles; (iii) and led to its sequestration in perinuclear/recycling endosomes with avoidance of multivesicular bodies and lysosomal degradation. Combined Src activation and EGF were synergistic for EGFR-Y845 and -Y1173 phosphorylation at some endosomes. We conclude that acute effects of Src in MDCK cells may mimic those of low EGF on EGFR activation and redistribution. Src:EGFR interactions may be sufficient to trigger EGFR activation and might contribute to its local signalling, without requiring either soluble extracellular signal or receptor over-expression.


Subject(s)
Cell Membrane/enzymology , Endocytosis/drug effects , Endosomes/enzymology , Epidermal Growth Factor/pharmacology , ErbB Receptors/metabolism , Signal Transduction/drug effects , src-Family Kinases/metabolism , Animals , Cell Membrane/drug effects , Cell Movement/drug effects , Cell Nucleus/drug effects , Cell Nucleus/metabolism , Clathrin-Coated Vesicles/metabolism , Dogs , Endosomes/drug effects , Enzyme Activation/drug effects , Epithelial Cells/cytology , Epithelial Cells/drug effects , Epithelial Cells/enzymology , Focal Adhesions/drug effects , Focal Adhesions/metabolism , Humans , Intracellular Space/drug effects , Intracellular Space/metabolism , Ligands , Phosphorylation/drug effects , Protein Binding/drug effects , Protein Transport/drug effects , rab4 GTP-Binding Proteins/metabolism
6.
Biochim Biophys Acta ; 1798(5): 909-27, 2010 May.
Article in English | MEDLINE | ID: mdl-20123084

ABSTRACT

Micrometric lipid compartmentation at the plasma membrane is disputed. Using live confocal imaging, we found that three unrelated fluorescent sphingomyelin (SM) analogs spontaneously clustered at the outer leaflet into micrometric domains, contrasting with homogeneous labelling by DiIC18 and TMA-DPH. In erythrocytes, these domains were round, randomly distributed, and reversibly coalesced under hypotonicity. BODIPY-SM and -glucosylceramide showed distinct temperature-dependence, in the same ranking as Tm for corresponding natural lipids, indicating phase behaviour. Scanning electron microscopy excluded micrometric surface structural features. In CHO cells, similar surface micrometric patches were produced by either direct BODIPY-SM insertion or intracellular processing from BODIPY-ceramide, ruling out aggregation artefacts. BODIPY-SM surface micrometric patches were refractory to endocytosis block or actin depolymerization and clustered upon cholesterol deprivation, indicating self-clustering at the plasma membrane. BODIPY-SM excimers further suggested clustering in ordered domains. Segregation of BODIPY-SM and -lactosylceramide micrometric domains showed coexistence of distinct phases. Consistent with micrometric domain boundaries, fluorescence recovery after photobleaching (FRAP) revealed restriction of BODIPY-SM lateral diffusion over long-range, but not short-range, contrasting with comparable high mobile fraction of BODIPY-lactosylceramide in both ranges. Controlled perturbations of endogenous SM pool similarly affected BODIPY-SM domain size by confocal imaging and its mobile fraction by FRAP. The latter evidence supports the hypothesis that, as shown for BODIPY-SM, endogenous SM spontaneously clusters at the plasmalemma outer leaflet of living cells into ordered micrometric domains, defined in shape by liquid-phase coexistence and in size by membrane tension and cholesterol. This proposal remains speculative and calls for further investigations.


Subject(s)
Cell Membrane/chemistry , Membrane Microdomains/chemistry , Sphingomyelins/chemistry , Animals , Boron Compounds/chemistry , CHO Cells , Cell Membrane/ultrastructure , Ceramides/chemistry , Cholesterol/metabolism , Cricetinae , Cricetulus , Erythrocytes/cytology , Erythrocytes/metabolism , Fluorescence Recovery After Photobleaching , Fluorescent Dyes/chemistry , HeLa Cells , Humans , Membrane Lipids/chemistry , Membrane Lipids/metabolism , Membrane Microdomains/ultrastructure
7.
Hum Reprod ; 24(2): 333-40, 2009 Feb.
Article in English | MEDLINE | ID: mdl-19049990

ABSTRACT

BACKGROUND: Menstruation is associated with a striking increase in matrix metalloproteinase (MMP) activity. However, it is still unknown whether the level of MMP activity correlates with the amount of menstrual bleeding. METHODS: We used histochemistry to investigate the degradation of the extracellular matrix (ECM), and immunohistochemical labelling and zymographic analysis to determine the level of expression and activity of MMP-2 and -9, and of their tissue inhibitors (TIMPs) -1, -2 and -3, in endometria sampled during menstruation in 14 women experiencing excessive menstrual bleeding and in 10 women successfully treated for menorrhagia by thermal ablation of the endometrium. RESULTS: After thermal ablation, regenerated menstrual endometria showed reduced areas of collagen fibre lysis and increased content of TIMP-1 and TIMP-2 compared with endometria from non-treated menorrhagic women. Surprisingly, treated endometria contained more latent gelatinase A (proMMP-2) but a lower proportion of the active form of gelatinase B (MMP-9) than non-treated endometria. CONCLUSIONS: These results suggest that ECM degradation is decreased at menstruation in the endometrium regenerated after thermal ablation, mostly because of an increased TIMP expression. This represents the first molecular explanation for the decreased amount of menstrual bleeding.


Subject(s)
Endometrial Ablation Techniques , Matrix Metalloproteinase 2/metabolism , Matrix Metalloproteinase 9/metabolism , Menorrhagia/metabolism , Adult , Extracellular Matrix/metabolism , Female , Humans , Menorrhagia/pathology , Menorrhagia/surgery , Middle Aged , Tissue Inhibitor of Metalloproteinase-1/metabolism , Tissue Inhibitor of Metalloproteinase-2/metabolism , Treatment Outcome
8.
Kidney Int ; 74(1): 52-61, 2008 Jul.
Article in English | MEDLINE | ID: mdl-18322545

ABSTRACT

Dysfunction of the proximal tubule (PT) is associated with variable degrees of solute wasting and low-molecular-weight proteinuria. We measured metabolic consequences and adaptation mechanisms in a model of inherited PT disorders using PT cells of ClC-5-deficient (Clcn5Y/-) mice, a well-established model of Dent's disease. Compared to cells taken from control mice, those from the mutant mice had increased expression of markers of proliferation (Ki67, proliferative cell nuclear antigen (PCNA), and cyclin E) and oxidative scavengers (superoxide dismutase I and thioredoxin). Transcriptome and protein analyses showed fourfold induction of type III carbonic anhydrase in a kidney-specific manner in the knockout mice located in scattered PT cells. Kidney-specific carbonic anhydrase type III (CAIII) upregulation was confirmed in other mice lacking the multiligand receptor megalin and in a patient with Dent's disease due to an inactivating CLCN5 mutation. The type III enzyme was specifically detected in the urine of mice lacking ClC-5 or megalin, patients with Dent's disease, and in PT cell lines exposed to oxidative stress. Our study shows that lack of PT ClC-5 in mice and men is associated with CAIII induction, increased cell proliferation, and oxidative stress.


Subject(s)
Carbonic Anhydrase III/physiology , Chloride Channels/deficiency , Fanconi Syndrome/pathology , Kidney Tubules, Proximal/physiology , Animals , Carbonic Anhydrase III/urine , Cell Proliferation , Cells, Cultured , Disease Models, Animal , Humans , Male , Mice , Mice, Knockout , Oxidative Stress
9.
J Cell Mol Med ; 12(1): 351-4, 2008.
Article in English | MEDLINE | ID: mdl-18081693

ABSTRACT

Understanding renal function requires one to integrate the structural complexity of kidney nephrons and the dynamic nature of their cellular processes. Multi-photon fluorescence microscopy is a state-of-the-art imaging technique for in vivo analysis of kidney tubules structure and function in real time. This study presents visual evidence for several levels of heterogeneity of proximal tubular endocytic uptake in the superficial renal mouse cortex and illustrates the potential of multi-photon microscopy for providing a comprehensive and dynamic portrayal of renal function.


Subject(s)
Endocytosis , Fluorescent Dyes/pharmacokinetics , Kidney Tubules, Proximal/metabolism , Microscopy, Fluorescence, Multiphoton , Animals , Male , Mice , Mice, Inbred C57BL , Tissue Distribution
10.
Biochim Biophys Acta ; 1768(7): 1830-8, 2007 Jul.
Article in English | MEDLINE | ID: mdl-17537401

ABSTRACT

The elastic properties of membrane bilayers are key parameters that control its deformation and can be affected by pharmacological agents. Our previous atomic force microscopy studies revealed that the macrolide antibiotic, azithromycin, leads to erosion of DPPC domains in a fluid DOPC matrix [A. Berquand, M. P. Mingeot-Leclercq, Y. F. Dufrene, Real-time imaging of drug-membrane interactions by atomic force microscopy, Biochim. Biophys. Acta 1664 (2004) 198-205.]. Since this observation could be due to an effect on DOPC cohesion, we investigated the effect of azithromycin on elastic properties of DOPC giant unilamellar vesicles (GUVs). Microcinematographic and morphometric analyses revealed that azithromycin addition enhanced lipid membranes fluctuations, leading to eventual disruption of the largest GUVs. These effects were related to change of elastic moduli of DOPC, quantified by the micropipette aspiration technique. Azithromycin decreased both the bending modulus (k(c), from 23.1+/-3.5 to 10.6+/-4.5 k(B)T) and the apparent area compressibility modulus (K(app), from 176+/-35 to 113+/-25 mN/m). These data suggested that insertion of azithromycin into the DOPC bilayer reduced the requirement level of both the energy for thermal fluctuations and the stress to stretch the bilayer. Computer modeling of azithromycin interaction with DOPC bilayer, based on minimal energy, independently predicted that azithromycin (i) inserts at the interface of phospholipid bilayers, (ii) decreases the energy of interaction between DOPC molecules, and (iii) increases the mean surface occupied by each phospholipid molecule. We conclude that azithromycin inserts into the DOPC lipid bilayer, so as to decrease its cohesion and to facilitate the merging of DPPC into the DOPC fluid matrix, as observed by atomic force microscopy. These investigations, based on three complementary approaches, provide the first biophysical evidence for the ability of an amphiphilic antibiotic to alter lipid elastic moduli. This may be an important determinant for drug: lipid interactions and cellular pharmacology.


Subject(s)
Azithromycin/pharmacology , Lipid Bilayers/chemistry , Phosphatidylcholines/chemistry , Unilamellar Liposomes/chemistry , Elasticity
11.
Cell Mol Life Sci ; 64(5): 610-20, 2007 Mar.
Article in English | MEDLINE | ID: mdl-17310281

ABSTRACT

Matrix metalloproteinase-7 (MMP-7, matrilysin- 1) modulates crucial biological events by processing many epithelial cell surface-associated effectors. We addressed MMP-7 interaction with human epithelial cells and its resulting activity. In human endometrium, a model of controlled tissue remodeling, proMMP-7 was diffusely immunolocalized inside epithelial cells, whereas MMP-7 delineated their entire plasma membrane. Endometrial explants preferentially retained active MMP-7, but not proMMP-7. Endometrial epithelial cells and carcinoma cells from various tissues bound active MMP-7. Endometrial carcinoma-derived Ishikawa cells showed high affinity (K(D) of approximately 2.5 nM) and capacity (approximately 260,000 sites per cell) for MMP-7. MMP-7 binding decreased by extracting membrane sterols or interfering with heparan sulfate proteoglycans, and was abrogated by tissue inhibitors of metalloproteinase-2 (TIMP-2) or synthetic MMP inhibitors. Bound MMP-7 not only remained fully active towards a macromolecular substrate but also became resistant to TIMP-2. We conclude that MMP-7-selective targeting to the plasma membrane of epithelial cells promotes its activity by conferring resistance to TIMP-2.


Subject(s)
Epithelial Cells/physiology , Matrix Metalloproteinase 7/metabolism , Catalysis , Cell Line , Cell Membrane/metabolism , Cells, Cultured , Endometrium/physiology , Female , Humans , Immunohistochemistry , Protein Precursors/metabolism , Tissue Inhibitor of Metalloproteinase-2/metabolism
12.
Bull Mem Acad R Med Belg ; 160(3-4): 193-200; discussion 200-1, 2005.
Article in French | MEDLINE | ID: mdl-16392373

ABSTRACT

Endocytosis is a distinctive property of all eukaryotic cells. Polarized cells face two different worlds by membranes of distinct composition: the basolateral membrane is exposed to the constant internal medium, whereas the apical membrane is exposed to variable environments. Endocytosis on both aspects also depends on different machineries. This short review illustrates the molecular basis and physiopathological implications of apical endocytosis. In a cultured epithelial cell line, Src selectively triggers apical macropinocytosis by activating the actin cytocortex via signalling membrane lipids generated by an amplification cascade involving phosphoinositide 3-kinase, phospholipase C and phospholipase D. Several actors of Src response are also activated by enteroinvasive bacteria, to trigger their entry into enterocytes. In the thyroid gland, the rates of thyroglobulin apical micropinocytosis and transfer to lysosomes determine the level of thyroid hormone production, by controlling the encounter of the prohormone with converting hydrolases. TSH selectively promotes the encounter, by inducing the expression of rate-limiting catalysts, the small GTPases Rab5 and Rab7, and of their exchange factor(s). This induction is constitutive in autonomous adenomas. In kidney proximal tubular cells, apical receptor-mediated endocytosis ensures full recapture of ultrafiltrated proteins. Inactivating mutations of the endosomal chloride channel, ClC-5, that are responsible for Dent's disease, cause a loss of surface receptors leading to proteinuria. These examples illustrate how three levels of regulation of apical endocytosis, namely the mode of entry, the rate of vesicular trafficking and the subcellular addressing account for a variety of human diseases.


Subject(s)
Endocytosis/physiology , Biological Transport , GTP Phosphohydrolases/physiology , Oncogenes/physiology
13.
J Membr Biol ; 192(3): 203-15, 2003 Apr 01.
Article in English | MEDLINE | ID: mdl-12820665

ABSTRACT

The macrolide antibiotic azithromycin was shown to markedly inhibit endocytosis. Here we investigate the interaction of azithromycin with biomembranes and its effects on membrane biophysics in relation to endocytosis. Equilibrium dialysis and 31P NMR revealed that azithromycin binds to lipidic model membranes and decreases the mobility of phospholipid phosphate heads. In contrast, azithromycin had no effect deeper in the bilayer, based on fluorescence polarization of TMA-DPH and DPH, compounds that, respectively, explore the interfacial and hydrophobic domains of bilayers, and it did not induce membrane fusion, a key event of vesicular trafficking. Atomic force microscopy showed that azithromycin perturbed lateral phase separation in Langmuir-Blodgett monolayers, indicating a perturbation of membrane organization in lateral domains. The consequence of azithromycin/ phospholipid interaction on membrane endocytosis was next evaluated in J774 macrophages by using three tracers with different insertion preferences inside the biological membranes and intracellular trafficking: C6-NBD-SM, TMA-DPH and N-Rh-PE. Azithromycin differentially altered their insertion into the plasma membrane, slowed down membrane trafficking towards lysosomes, as evaluated by the rate of N-Rh-PE self-quenching relief, but did not affect bulk membrane internalization of C6-NBD-SM and TMA-DPH. Azithromycin also decreased plasma membrane fluidity, as shown by TMA-DPH fluorescence polarization and confocal microscopy after labeling by fluorescent concanavalin A. We conclude that azithromycin directly interacts with phospholipids, modifies biophysical properties of membrane and affects membrane dynamics in living cells. This antibiotic may therefore help to elucidate the physico-chemical properties underlying endocytosis.


Subject(s)
1,2-Dipalmitoylphosphatidylcholine/chemistry , Azithromycin/chemistry , Azithromycin/pharmacology , Cell Membrane/metabolism , Cell Membrane/ultrastructure , Liposomes/chemistry , Membrane Fluidity/drug effects , Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/pharmacology , Cell Line , Cell Membrane/chemistry , Endocytosis/drug effects , Endocytosis/physiology , Hydrogen-Ion Concentration , Lipids/chemistry , Macromolecular Substances , Macrophages/chemistry , Macrophages/cytology , Macrophages/metabolism , Temperature
14.
J Endocrinol ; 173(1): 177-85, 2002 Apr.
Article in English | MEDLINE | ID: mdl-11927397

ABSTRACT

Nitric oxide (NO) is a well-known mediator of autoimmune processes. In the thyroid gland, it is produced in response to interleukin 1 (IL-1) and may mediate cytokine action at an early stage of autoimmune thyroiditis. In this study, we have investigated whether NO is involved in cytokine-induced cytotoxic effects and epithelial barrier alterations in thyrocytes. Human thyroid epithelial cells were cultured as tight polarised monolayers on a permeable support and exposed or not to IL-1alpha (100 U/ml), alone or in combination with interferon-gamma (IFN-gamma; 100 U/ml) added to the basal compartment. NO production was not detected in control thyrocytes, but was significantly induced by the combination of IL-1alpha with IFN-gamma, in a time-dependent fashion. Similarly, expression of the inducible isoform of nitric oxide synthase (NOSII), determined by immunoblot and immunofluorescence confocal microscopy, was not detected in control cells, but was markedly induced after 48-h exposure to both cytokines. This treatment significantly increased the release of cytosolic lactate dehydrogenase (LDH) in the apical and basolateral media and decreased transepithelial electrical resistance. Although IFN-gamma was not sufficient to induce NO production, it could by itself decrease transepithelial resistance and synergised the IL-1alpha effect on LDH release. The NOS inhibitor, L-nitro-arginine-methyl ester, suppressed the cytokine-induced NO production and decreased the LDH release, but failed to prevent the loss of transepithelial resistance. These results indicated that human thyrocytes express NOSII and produce NO in response to IL-1alpha+IFN-gamma and suggest that NO acts as a mediator of cytokine-induced cytotoxicity in the thyroid gland and may promote the exposure of autoantigens to the immune system. In contrast, NO does not appear to mediate the cytokine-induced disruption of the thyroid epithelial barrier.


Subject(s)
Interleukin-1/pharmacology , L-Lactate Dehydrogenase/metabolism , Nitric Oxide/physiology , Thyroid Gland/metabolism , Cell Death , Cell Polarity , Cells, Cultured , Epithelium/drug effects , Epithelium/metabolism , Humans , Interferon-gamma/pharmacology , Microscopy, Confocal , NG-Nitroarginine Methyl Ester/pharmacology , Nitric Oxide Synthase/antagonists & inhibitors , Nitric Oxide Synthase/metabolism , Nitric Oxide Synthase Type II , Thyroid Gland/drug effects
15.
Am J Kidney Dis ; 38(6): 1421-9, 2001 Dec.
Article in English | MEDLINE | ID: mdl-11728985

ABSTRACT

The most common form of autosomal dominant polycystic kidney disease (PKD) results from mutation of the PKD1 gene on chromosome 16p13.3. The gene encodes a 14-kb messenger RNA that is predicted to express a 462-kd membrane protein. The gene product, polycystin-1, has a large extracellular portion composed of a novel combination of protein-protein interacting domains and is postulated to be a plasma membrane receptor involved in cell-cell/matrix interactions. However, slow progress has been made in the characterization of polycystin-1 or the determination of its function. In fact, the protein is expressed at very low levels in tissues and cell lines and previous efforts directed at expression of recombinant protein had been largely unsuccessful. We have recently developed constructs of full-length human PKD1 complementary (cDNA) that can be expressed in both a stable and transient fashion in mammalian cells. We used these systems to characterize our antibodies and to track the protein in vivo. We report here the first biochemical characterization of recombinant polycystin-1 and show that the protein is a 520-kd glycosylated polypeptide with an unglycosylated core of 460 kd. Subcellular fractionation as well as biotinylation studies confirmed that the protein is plasma-membrane associated. Furthermore, we show that the recombinant protein localizes to cell-cell junctions in polarized madin darby canine kidney cells as revealed by indirect immunofluorescence. Our data represent the first characterization of polycystin-1 performed under highly controlled conditions.


Subject(s)
DNA, Complementary/isolation & purification , Polycystic Kidney, Autosomal Dominant/genetics , Proteins/genetics , Animals , Blotting, Western/standards , Cell Membrane/chemistry , DNA, Complementary/chemistry , Dogs , Gene Expression , Glycosylation , Humans , In Vitro Techniques , Intercellular Junctions/chemistry , Kidney/chemistry , Molecular Weight , Proteins/isolation & purification , RNA, Messenger/isolation & purification , TRPP Cation Channels
16.
Eur J Cell Biol ; 80(7): 466-78, 2001 Jul.
Article in English | MEDLINE | ID: mdl-11499789

ABSTRACT

The dicationic macrolide antibiotic azithromycin inhibits the uptake of horseradish peroxidase (HRP) by fluid-phase pinocytosis in fibroblasts in a time- and concentration-dependent fashion without affecting its decay (regurgitation and/or degradation). The azithromycin effect is additive to that of nocodazole, known to impair endocytic uptake and transport of solutes along the endocytic pathway. Cytochemistry (light and electron microscopy) shows a major reduction by azithromycin in the number of HRP-labeled endocytic vesicles at 5 min (endosomes) and 2 h (lysosomes). Within 3 h of exposure, azithromycin also causes the appearance of large and light-lucentlelectron-lucent vacuoles, most of which can be labeled by lucifer yellow when this tracer is added to culture prior to azithromycin exposure. Three days of treatment with azithromycin result in the accumulation of very large vesicles filled with pleiomorphic content, consistent with phospholipidosis. These vesicles are accessible to fluorescein-labeled bovine serum albumin (FITC-BSA) and intensively stained with filipin, indicating a mixed storage with cholesterol. The impairment of HRP pinocytosis directly correlates with the amount of azithromycin accumulated by the cells, but not with the phospholipidosis induced by the drug. The proton ionophore monensin, which completely suppresses azithromycin accumulation, also prevents inhibition of HRP uptake. Erythromycylamine, another dicationic macrolide, also inhibits HRP pinocytosis in direct correlation with its cellular accumulation and is as potent as azithromycin at equimolar cellular concentrations. We suggest that dicationic macrolides inhibit fluid-phase pinocytosis by impairing the formation of pinocytic vacuoles and endosomes.


Subject(s)
Anti-Bacterial Agents/pharmacology , Azithromycin/pharmacology , Erythromycin/analogs & derivatives , Lysosomes/metabolism , Pinocytosis/drug effects , Adenosine Triphosphate/metabolism , Animals , Antineoplastic Agents/pharmacology , Cell Membrane/drug effects , Cell Membrane/metabolism , Cell Membrane/ultrastructure , Cells, Cultured , Coloring Agents , DNA/biosynthesis , Erythromycin/pharmacology , Fetus/cytology , Fibroblasts/cytology , Horseradish Peroxidase/pharmacokinetics , Humans , Ionophores/pharmacology , Lysosomes/drug effects , Lysosomes/ultrastructure , Microscopy, Electron , Monensin/pharmacology , Nocodazole/pharmacology , Phospholipids/metabolism , Protein Binding/drug effects , Rats , Rats, Wistar , Tolonium Chloride , Transferrin/metabolism , Transport Vesicles/drug effects , Transport Vesicles/metabolism , Transport Vesicles/ultrastructure , Vacuoles/drug effects , Vacuoles/metabolism , Vacuoles/ultrastructure
17.
Biochem J ; 358(Pt 1): 275-80, 2001 Aug 15.
Article in English | MEDLINE | ID: mdl-11485578

ABSTRACT

Most matrix metalloproteinases (MMPs) are secreted as inactive proenzymes. Their expression is well documented in several human tissues, but their activators in vivo are still unknown. To address this question, the activation of progelatinase B (proMMP-9) in the human endometrium was selected as a model system. ProMMP-9 was detected by gelatin zymography in homogenates of fresh endometrial tissue sampled during all phases of the menstrual cycle, whereas its active form was observed only during the late secretory and menstrual phases. Furthermore, proMMP-9 was expressed and activated in endometrial explants sampled outside the perimenstrual phase and cultured in the absence of both progesterone and oestradiol, mimicking the menstrual condition in vivo. Analysis of such tissue cultures by gelatin zymography and Western blotting showed that activation of proMMP-9 depended on a secreted factor and was selectively inhibited by either a synthetic inhibitor of stromelysin 1 (MMP-3) or a monoclonal antibody that specifically blocks MMP-3, thus providing strong evidence for the activation of proMMP-9 in vivo by MMP-3. The activation of proMMP-3 was itself inhibited by a broad-range MMP inhibitor in most cultures, but seemed to involve multiple pathways, implying both serine proteinases and metalloproteinases, which could operate in parallel or sequentially.


Subject(s)
Enzyme Precursors/metabolism , Gelatinases/metabolism , Matrix Metalloproteinase 3/chemistry , Menstrual Cycle , Metalloendopeptidases/metabolism , Animals , Antibodies, Monoclonal/metabolism , Blotting, Western , Culture Techniques , Dose-Response Relationship, Drug , Endometrium/enzymology , Endometrium/metabolism , Enzyme Activation , Enzyme Inhibitors/pharmacology , Enzyme Precursors/antagonists & inhibitors , Enzyme Precursors/biosynthesis , Estradiol/metabolism , Female , Gelatinases/antagonists & inhibitors , Gelatinases/biosynthesis , Humans , Matrix Metalloproteinase 3/biosynthesis , Matrix Metalloproteinase Inhibitors , Metalloendopeptidases/antagonists & inhibitors , Metalloendopeptidases/biosynthesis , Mice , Progesterone/metabolism , Protein Binding , Time Factors
18.
Scand J Immunol ; 53(1): 56-64, 2001 Jan.
Article in English | MEDLINE | ID: mdl-11169207

ABSTRACT

The intracellular pathway of polymeric immunoglobulin receptor (pIgR) is governed by multiple signals that lead to constitutive transcytosis. In addition, in transfected polarized MDCK cells, polymeric immunoglobulin A (pIgA) binding stimulates rabbit pIgR-transcytosis, owing to phospholipase-C gamma 1 activation and increase of intracellular calcium. Transcytosis of rat pIgR across hepatocytes is similarly accelerated by pIgA injection. In contrast we show here that human Madrin-Darby Canine Kidney (pIgR)-transcytosis, in human Calu-3 and human pIgR-transfected MDCK cells, is not promoted by pIgA, as monitored by a continuous apical release of its secreted ectodomain. However, the incubation of cells expressing human or rabbit pIgR with pIgA induces a comparable IP3 production, and pIgR-transcytosis of either species is accelerated by the protein kinase C (PKC)-activator phorbol myristate acetate. Without pIgA, mimicking phospholipase-C activation by combining low concentrations of phorbol myristate acetate with ionomycin, or high concentrations of ionomycin alone, stimulates the rabbit, but not the human, pIgR transcytosis. These data suggest that the species difference in pIgA-induced pIgR-transcytosis does not stem from the defective production of second messengers, but from a different sensitivity of pIgR to intracellular calcium. Our results outline the danger of extrapolating to humans the abundant data obtained from mucosal vaccination of laboratory animals.


Subject(s)
Immunoglobulin A/metabolism , Receptors, Polymeric Immunoglobulin/metabolism , Signal Transduction/physiology , Adenocarcinoma/pathology , Animals , Calcium Signaling/drug effects , Cell Line/drug effects , DNA, Complementary/genetics , Dogs , Enzyme Activation/drug effects , Humans , Immunoglobulin A/genetics , Immunoglobulin A/immunology , Inositol 1,4,5-Trisphosphate/metabolism , Ionomycin/pharmacology , Ionophores/pharmacology , Kidney Tubules, Proximal/cytology , Lung Neoplasms/pathology , Protein Kinase C/drug effects , Protein Kinase C/physiology , Protein Transport/drug effects , Rabbits , Rats , Receptors, Polymeric Immunoglobulin/genetics , Receptors, Polymeric Immunoglobulin/physiology , Recombinant Fusion Proteins/immunology , Recombinant Fusion Proteins/metabolism , Second Messenger Systems/physiology , Species Specificity , Tetradecanoylphorbol Acetate/pharmacology , Transfection , Tumor Cells, Cultured , Vaccination
19.
Diabetes ; 50(1): 143-9, 2001 Jan.
Article in English | MEDLINE | ID: mdl-11147780

ABSTRACT

Bcl-2 mRNA expression was detected in rat blastocysts by in situ hybridization. The distribution of mRNA expression was rather heterogenous, with approximately 2% of high-expressing cells. In vitro exposure to 28 mmol/l D-glucose for 24 h resulted in a significant increase in the proportion of these cells compared with control embryos in either 6 mmol/l D-glucose or 28 mmol/l D+L-glucose. Heterogeneity in the expression of Bcl-2 was also observed at the protein level by immunocytochemistry. Exposure to 28 mmol/l D-glucose significantly increased the incidence of chromatin degradation (karyolysis) and nuclear fragmentation (karyorhexis), two nuclear markers of apoptosis in rat blastocysts. When two different antisense oligodeoxynucleotides designed to block Bcl-2 expression were added to 28 mmol/1 D-glucose, the incidence of karyolysis (but not karyorhexis) was increased compared with embryos in 28 mmol/l D-glucose alone. These data suggest that Bcl-2 is involved in the protective response against the induction of karyolysis in blastocysts on their exposure to high concentrations of D-glucose in vitro, whereas karyorhexis appears to result from the activation of an intracellular pathway that is independent of Bcl-2.


Subject(s)
Blastocyst/drug effects , Blastocyst/physiology , Glucose/pharmacology , Proto-Oncogene Proteins c-bcl-2/physiology , Animals , Blastocyst/metabolism , Culture Techniques , Dose-Response Relationship, Drug , Female , Oligonucleotides, Antisense/pharmacology , Proto-Oncogene Proteins c-bcl-2/antagonists & inhibitors , Proto-Oncogene Proteins c-bcl-2/biosynthesis , Proto-Oncogene Proteins c-bcl-2/genetics , Proto-Oncogene Proteins c-bcl-2/metabolism , RNA, Messenger/metabolism , Rats , Rats, Wistar
20.
Hum Mol Genet ; 9(20): 2937-45, 2000 Dec 12.
Article in English | MEDLINE | ID: mdl-11115837

ABSTRACT

Nephrolithiasis (kidney stones) affects 5-10% of adults and is most commonly associated with hypercalciuria, which may be due to monogenic renal tubular disorders. One such hypercalciuric disorder is Dent's disease, which is characterized by renal proximal tubular defects that include low molecular weight proteinuria, aminoaciduria and glycosuria, together with rickets in some patients. Dent's disease is due to inactivating mutations of the renal-specific voltage-gated chloride channel, CLC-5, which is expressed in the proximal tubule, thick ascending limb and collecting duct. The subcellular localization of CLC-5 to the proximal tubular endosomes has suggested a role in endocytosis, and to facilitate in vivo investigations of CLC-5 in Dent's disease we generated mice lacking CLC-5 by targeted gene disruption. CLC-5-deficient mice developed renal tubular defects which included low molecular weight (<70 kDa) proteinuria, generalized aminoaciduria that was more pronounced for neutral and polar amino acids, and glycosuria. They also developed hypercalciuria and renal calcium deposits and some had deformities of the spine. Furthermore, endocytosis as assessed by horseradish peroxidase uptake in the proximal tubule was severely impaired in CLC-5-deficient mice, thereby demonstrating a role for CLC-5 in endosomal uptake of low molecular weight proteins. Thus, CLC-5-deficient mice provide a model for Dent's disease and this will help in elucidating the function of this chloride channel in endocytosis and renal calcium homeostasis.


Subject(s)
Chloride Channels/genetics , Endocytosis/physiology , Kidney Calculi/genetics , Animals , Calcium/metabolism , Cells, Cultured , Chloride Channels/deficiency , Chloride Channels/metabolism , Disease Models, Animal , Female , Homeostasis , Immunoenzyme Techniques , Kidney Calculi/metabolism , Kidney Calculi/pathology , Male , Mice , Mice, Inbred C57BL
SELECTION OF CITATIONS
SEARCH DETAIL
...