Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 97
Filter
Add more filters

Publication year range
1.
J Am Soc Nephrol ; 30(11): 2177-2190, 2019 11.
Article in English | MEDLINE | ID: mdl-31548351

ABSTRACT

BACKGROUND: Deletions or inactivating mutations of the cystinosin gene CTNS lead to cystine accumulation and crystals at acidic pH in patients with nephropathic cystinosis, a rare lysosomal storage disease and the main cause of hereditary renal Fanconi syndrome. Early use of oral cysteamine to prevent cystine accumulation slows progression of nephropathic cystinosis but it is a demanding treatment and not a cure. The source of cystine accumulating in kidney proximal tubular cells and cystine's role in disease progression are unknown. METHODS: To investigate whether receptor-mediated endocytosis by the megalin/LRP2 pathway of ultrafiltrated, disulfide-rich plasma proteins could be a source of cystine in proximal tubular cells, we used a mouse model of cystinosis in which conditional excision of floxed megalin/LRP2 alleles in proximal tubular cells of cystinotic mice was achieved by a Cre-LoxP strategy using Wnt4-CRE. We evaluated mice aged 6-9 months for kidney cystine levels and crystals; histopathology, with emphasis on swan-neck lesions and proximal-tubular-cell apoptosis and proliferation (turnover); and proximal-tubular-cell expression of the major apical transporters sodium-phosphate cotransporter 2A (NaPi-IIa) and sodium-glucose cotransporter-2 (SGLT-2). RESULTS: Wnt4-CRE-driven megalin/LRP2 ablation in cystinotic mice efficiently blocked kidney cystine accumulation, thereby preventing lysosomal deformations and crystal deposition in proximal tubular cells. Swan-neck lesions were largely prevented and proximal-tubular-cell turnover was normalized. Apical expression of the two cotransporters was also preserved. CONCLUSIONS: These observations support a key role of the megalin/LRP2 pathway in the progression of nephropathic cystinosis and provide a proof of concept for the pathway as a therapeutic target.


Subject(s)
Cystinosis/etiology , Endocytosis , Kidney Tubules, Proximal/pathology , Low Density Lipoprotein Receptor-Related Protein-2/physiology , Animals , Cystine/metabolism , Cystinosis/prevention & control , Disease Progression , Mice , Mice, Inbred C57BL , Signal Transduction/physiology , Wnt4 Protein/physiology
2.
Development ; 143(11): 1958-70, 2016 06 01.
Article in English | MEDLINE | ID: mdl-27068110

ABSTRACT

Thyroid follicles, the functional units of the thyroid gland, are delineated by a monolayer of thyrocytes resting on a continuous basement membrane. The developmental mechanisms of folliculogenesis, whereby follicles are formed by the reorganization of a non-structured mass of non-polarized epithelial cells, are largely unknown. Here we show that assembly of the epithelial basement membrane is crucial for folliculogenesis and is controlled by endothelial cell invasion and by BMP-Smad signaling in thyrocytes. Thyroid-specific Smad1 and Smad5 double-knockout (Smad1/5(dKO)) mice displayed growth retardation, hypothyroidism and defective follicular architecture. In Smad1/5(dKO) embryonic thyroids, epithelial cells remained associated in large clusters and formed small follicles. Although similar follicular defects are found in Vegfa knockout (Vegfa(KO)) thyroids, Smad1/5(dKO) thyroids had normal endothelial cell density yet impaired endothelial differentiation. Interestingly, both Vegfa(KO) and Smad1/5(dKO) thyroids displayed impaired basement membrane assembly. Furthermore, conditioned medium (CM) from embryonic endothelial progenitor cells (eEPCs) rescued the folliculogenesis defects of both Smad1/5(dKO) and Vegfa(KO) thyroids. Laminin α1, ß1 and γ1, abundantly released by eEPCs into CM, were crucial for folliculogenesis. Thus, epithelial Smad signaling and endothelial cell invasion promote folliculogenesis via assembly of the basement membrane.


Subject(s)
Basement Membrane/metabolism , Endothelial Cells/metabolism , Smad1 Protein/metabolism , Smad5 Protein/metabolism , Thyroid Gland/embryology , Animals , Basement Membrane/drug effects , Blood Vessels/drug effects , Blood Vessels/metabolism , Bone Morphogenetic Proteins/metabolism , Collagen Type IV/metabolism , Culture Media, Conditioned/pharmacology , Gene Expression Regulation, Developmental/drug effects , Hypothyroidism/metabolism , Laminin/metabolism , Mice, Knockout , Organogenesis/drug effects , Organogenesis/genetics , Signal Transduction/drug effects , Stem Cells/cytology , Stem Cells/drug effects , Stem Cells/metabolism , Thyroid Epithelial Cells/cytology , Thyroid Epithelial Cells/drug effects , Thyroid Epithelial Cells/metabolism , Thyroid Gland/cytology , Thyroid Gland/drug effects , Vascular Endothelial Growth Factor A/metabolism
3.
Blood ; 127(10): 1325-35, 2016 Mar 10.
Article in English | MEDLINE | ID: mdl-26668133

ABSTRACT

Mutations in the calreticulin gene (CALR) represented by deletions and insertions in exon 9 inducing a -1/+2 frameshift are associated with a significant fraction of myeloproliferative neoplasms (MPNs). The mechanisms by which CALR mutants induce MPN are unknown. Here, we show by transcriptional, proliferation, biochemical, and primary cell assays that the pathogenic CALR mutants specifically activate the thrombopoietin receptor (TpoR/MPL). No activation is detected with a battery of type I and II cytokine receptors, except granulocyte colony-stimulating factor receptor, which supported only transient and weak activation. CALR mutants induce ligand-independent activation of JAK2/STAT/phosphatydylinositol-3'-kinase (PI3-K) and mitogen-activated protein (MAP) kinase pathways via TpoR, and autonomous growth in Ba/F3 cells. In these transformed cells, no synergy is observed between JAK2 and PI3-K inhibitors in inhibiting cytokine-independent proliferation, thus showing a major difference from JAK2V617F cells where such synergy is strong. TpoR activation was dependent on its extracellular domain and its N-glycosylation, especially at N117. The glycan binding site and the novel C-terminal tail of the mutant CALR proteins were required for TpoR activation. A soluble form of TpoR was able to prevent activation of full-length TpoR provided that it was N-glycosylated. By confocal microscopy and subcellular fractionation, CALR mutants exhibit different intracellular localization from that of wild-type CALR. Finally, knocking down either MPL/TpoR or JAK2 in megakaryocytic progenitors from patients carrying CALR mutations inhibited cytokine-independent megakaryocytic colony formation. Taken together, our study provides a novel signaling paradigm, whereby a mutated chaperone constitutively activates cytokine receptor signaling.


Subject(s)
Calreticulin/metabolism , Hematologic Neoplasms/metabolism , LIM Domain Proteins/metabolism , Muscle Proteins/metabolism , Mutation , Myeloproliferative Disorders/metabolism , Neoplasm Proteins/metabolism , Receptors, Thrombopoietin/metabolism , Animals , Calreticulin/genetics , Cell Line, Tumor , Glycosylation , Hematologic Neoplasms/genetics , Hematologic Neoplasms/pathology , Janus Kinase 2/genetics , Janus Kinase 2/metabolism , LIM Domain Proteins/genetics , Mice , Muscle Proteins/genetics , Myeloproliferative Disorders/genetics , Myeloproliferative Disorders/pathology , Neoplasm Proteins/genetics , Phosphatidylinositol 3-Kinases/genetics , Phosphatidylinositol 3-Kinases/metabolism , Phosphorylation/genetics , Protein Transport/genetics , Receptors, Thrombopoietin/genetics , STAT Transcription Factors/genetics , STAT Transcription Factors/metabolism , Signal Transduction/genetics
4.
Immunity ; 28(3): 414-24, 2008 Mar.
Article in English | MEDLINE | ID: mdl-18342010

ABSTRACT

For several days after antigenic stimulation, human cytolytic T lymphocyte (CTL) clones exhibit a decrease in their effector activity and in their binding to human leukocyte antigen (HLA)-peptide tetramers. We observed that, when in this state, CTLs lose the colocalization of the T cell receptor (TCR) and CD8. Effector function and TCR-CD8 colocalization were restored with galectin disaccharide ligands, suggesting that the binding of TCR to galectin plays a role in the distancing of TCR from CD8. These findings appear to be applicable in vivo, as TCR was observed to be distant from CD8 on human tumor-infiltrating lymphocytes, which were anergic. These lymphocytes recovered effector functions and TCR-CD8 colocalization after ex vivo treatment with galectin disaccharide ligands. The separation of TCR and CD8 molecules could be one major mechanism of anergy in tumors and other chronic stimulation conditions.


Subject(s)
CD8 Antigens/metabolism , Clonal Anergy/immunology , Lymphocytes, Tumor-Infiltrating/metabolism , Receptors, Antigen, T-Cell/metabolism , T-Lymphocytes, Cytotoxic/metabolism , CD8 Antigens/immunology , Cell Line, Tumor , Flow Cytometry , Galectins/metabolism , HLA Antigens/immunology , Humans , Lymphocyte Activation/immunology , Lymphocytes, Tumor-Infiltrating/immunology , Microscopy, Confocal , Microscopy, Electron, Scanning , Peptides/immunology , Receptors, Antigen, T-Cell/immunology , T-Lymphocytes, Cytotoxic/immunology
5.
J Am Soc Nephrol ; 27(6): 1678-88, 2016 06.
Article in English | MEDLINE | ID: mdl-26449607

ABSTRACT

Cystinosis is a rare autosomal recessive storage disorder characterized by defective lysosomal efflux of cystine due to mutations in the CTNS gene encoding the lysosomal cystine transporter, cystinosin. Lysosomal cystine accumulation leads to crystal formation and functional impairment of multiple organs. Moreover, cystinosis is the most common inherited cause of renal Fanconi syndrome in children. Oral cysteamine therapy delays disease progression by reducing intracellular cystine levels. However, because cysteamine does not correct all complications of cystinosis, including Fanconi syndrome, we hypothesized that cystinosin could have novel roles in addition to transporting cystine out of the lysosome. By coimmunoprecipitation experiments and mass spectrometry, we found cystinosin interacts with almost all components of vacuolar H(+)-ATPase and the Ragulator complex and with the small GTPases Ras-related GTP-binding protein A (RagA) and RagC. Furthermore, the mammalian target of rapamycin complex 1 (mTORC1) pathway was downregulated in proximal tubular cell lines derived from Ctns(-/-) mice. Decrease of lysosomal cystine levels by cysteamine did not rescue mTORC1 activation in these cells, suggesting that the downregulation of mTORC1 is due to the absence of cystinosin rather than to the accumulation of cystine. Our results show a dual role for cystinosin as a cystine transporter and as a component of the mTORC1 pathway, and provide an explanation for the appearance of Fanconi syndrome in cystinosis. Furthermore, this study highlights the need to develop new treatments not dependent on lysosomal cystine depletion alone for this devastating disease.


Subject(s)
Amino Acid Transport Systems, Neutral/physiology , Cystinosis/etiology , Multiprotein Complexes/physiology , Signal Transduction , TOR Serine-Threonine Kinases/physiology , Vacuolar Proton-Translocating ATPases/physiology , Animals , Mechanistic Target of Rapamycin Complex 1 , Mice
6.
Traffic ; 15(4): 401-17, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24548619

ABSTRACT

Matrix metalloproteinase-27 (MMP-27) is poorly characterized. Sequence comparison suggests that a C-terminal extension (CTE) includes a potential transmembrane domain as in some membrane-type (MT)-MMPs. Having noticed that MMP-27 was barely secreted, we investigated its subcellular localization and addressed CTE contribution for MMP-27 retention. Intracellular MMP-27 was sensitive to endoglycosidase H. Subcellular fractionation and confocal microscopy evidenced retention of endogenous MMP-27 or recombinant rMMP-27 in the endoplasmic reticulum (ER) with locked exit across the intermediate compartment (ERGIC). Conversely, truncated rMMP-27 without CTE accessed downstream secretory compartments (ERGIC and Golgi) and was constitutively secreted. CTE addition to rMMP-10 (a secreted MMP) caused ER retention and blocked secretion. Addition of a PKA target sequence to the cytosolic C-terminus of transmembrane MT1-MMP/MMP-14 led to effective phosphorylation upon forskolin stimulation, but not for MMP-27, excluding transmembrane anchorage. Moreover, MMP-27 was protected from digestion by proteinase K. Finally, MT1-MMP/MMP-14 but neither endogenous nor recombinant MMP-27 partitioned in the detergent phase after Triton X-114 extraction, indicating that MMP-27 is not an integral membrane protein. In conclusion, MMP-27 is efficiently retained within the ER due to its unique CTE, which does not lead to stable membrane insertion. This could represent a novel ER retention system.


Subject(s)
Endoplasmic Reticulum/enzymology , Matrix Metalloproteinases/metabolism , Amino Acid Sequence , Humans , Matrix Metalloproteinases/chemistry , Molecular Sequence Data , Sequence Homology, Amino Acid , Subcellular Fractions/enzymology
7.
Kidney Int ; 89(5): 1075-1089, 2016 May.
Article in English | MEDLINE | ID: mdl-27083284

ABSTRACT

Hepatocyte nuclear factor 1α (HNF1α) is a transcription factor expressed in the liver, pancreas, and proximal tubule of the kidney. Mutations of HNF1α cause an autosomal dominant form of diabetes mellitus (MODY-HNF1A) and tubular dysfunction. To gain insights into the role of HNF1α in the proximal tubule, we analyzed Hnf1a-deficient mice. Compared with wild-type littermates, Hnf1a knockout mice showed low-molecular-weight proteinuria and a 70% decrease in the uptake of ß2-microglobulin, indicating a major endocytic defect due to decreased expression of megalin/cubilin receptors. We identified several binding sites for HNF1α in promoters of Lrp2 and Cubn genes encoding megalin and cubilin, respectively. The functional interaction of HNF1α with these promoters was shown in C33 epithelial cells lacking endogenous HNF1α. Defective receptor-mediated endocytosis was confirmed in proximal tubule cells from these knockout mice and could be rescued by transfection of wild-type but not mutant HNF1α. Transfection of human proximal tubule HK2 cells with HNF1α was able to upregulate megalin and cubilin expression and to increase endocytosis of albumin. Low-molecular-weight proteinuria was consistently detected in individuals with HNF1A mutations compared with healthy controls and patients with non-MODY-HNF1A diabetes mellitus. Thus, HNF1α plays a key role in the constitutive expression of megalin and cubilin, hence regulating endocytosis in the proximal tubule of the kidney. These findings provide new insight into the renal phenotype of individuals with mutations of HNF1A.


Subject(s)
Diabetes Mellitus, Type 2/genetics , Diabetic Nephropathies/genetics , Endocytosis , Hepatocyte Nuclear Factor 1-alpha/genetics , Kidney Tubules, Proximal/metabolism , Mutation , Proteinuria/genetics , Adolescent , Adult , Aged , Animals , Binding Sites , Case-Control Studies , Cells, Cultured , Diabetes Mellitus, Type 2/metabolism , Diabetes Mellitus, Type 2/physiopathology , Diabetic Nephropathies/metabolism , Diabetic Nephropathies/physiopathology , Female , Gene Expression Regulation , Genetic Predisposition to Disease , Hepatocyte Nuclear Factor 1-alpha/deficiency , Hepatocyte Nuclear Factor 1-alpha/metabolism , Humans , Kidney Tubules, Proximal/physiopathology , Low Density Lipoprotein Receptor-Related Protein-2/genetics , Low Density Lipoprotein Receptor-Related Protein-2/metabolism , Male , Mice, Inbred C57BL , Mice, Knockout , Middle Aged , Phenotype , Promoter Regions, Genetic , Proteinuria/metabolism , Proteinuria/physiopathology , Receptors, Cell Surface/genetics , Receptors, Cell Surface/metabolism , Signal Transduction , Transfection , Young Adult
8.
Stem Cells ; 33(1): 301-9, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25186209

ABSTRACT

Despite controversies on the potential of hematopoietic stem cells (HSCs) to promote tissue repair, we previously showed that HSC transplantation could correct cystinosis, a multisystemic lysosomal storage disease, caused by a defective lysosomal membrane cystine transporter, cystinosin (CTNS gene). Addressing the cellular mechanisms, we here report vesicular cross-correction after HSC differentiation into macrophages. Upon coculture with cystinotic fibroblasts, macrophages produced tunneling nanotubes (TNTs) allowing transfer of cystinosin-bearing lysosomes into Ctns-deficient cells, which exploited the same route to retrogradely transfer cystine-loaded lysosomes to macrophages, providing a bidirectional correction mechanism. TNT formation was enhanced by contact with diseased cells. In vivo, HSCs grafted to cystinotic kidneys also generated nanotubular extensions resembling invadopodia that crossed the dense basement membranes and delivered cystinosin into diseased proximal tubular cells. This is the first report of correction of a genetic lysosomal defect by bidirectional vesicular exchange via TNTs and suggests broader potential for HSC transplantation for other disorders due to defective vesicular proteins.


Subject(s)
Hematopoietic Stem Cell Transplantation , Hematopoietic Stem Cells/cytology , Lysosomes/metabolism , Macrophages/cytology , Nanotubes , Animals , Cystinosis/metabolism , Cystinosis/pathology , Cystinosis/therapy , Fibroblasts , Hematopoietic Stem Cells/metabolism , Humans , Macrophages/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic
9.
Cell Mol Life Sci ; 72(23): 4633-51, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26077601

ABSTRACT

Although cholesterol is essential for membrane fluidity and deformability, the level of its lateral heterogeneity at the plasma membrane of living cells is poorly understood due to lack of appropriate probe. We here report on the usefulness of the D4 fragment of Clostridium perfringens toxin fused to mCherry (theta*), as specific, non-toxic, sensitive and quantitative cholesterol-labeling tool, using erythrocyte flat membrane. By confocal microscopy, theta* labels cholesterol-enriched submicrometric domains in coverslip-spread but also gel-suspended (non-stretched) fresh erythrocytes, suggesting in vivo relevance. Cholesterol domains on spread erythrocytes are stable in time and space, restricted by membrane:spectrin anchorage via 4.1R complexes, and depend on temperature and sphingomyelin, indicating combined regulation by extrinsic membrane:cytoskeleton interaction and by intrinsic lipid packing. Cholesterol domains partially co-localize with BODIPY-sphingomyelin-enriched domains. In conclusion, we show that theta* is a useful vital probe to study cholesterol organization and demonstrate that cholesterol forms submicrometric domains in living cells.


Subject(s)
Cholesterol/metabolism , Erythrocyte Membrane/metabolism , Membrane Microdomains/metabolism , Animals , Bacterial Toxins/genetics , Bacterial Toxins/metabolism , Boron Compounds/chemistry , Boron Compounds/metabolism , Cell Line , Erythrocyte Membrane/chemistry , Hemolysin Proteins/genetics , Hemolysin Proteins/metabolism , Humans , Membrane Microdomains/chemistry , Mice , Myoblasts/metabolism , Sphingomyelins/chemistry , Sphingomyelins/metabolism , Temperature
10.
Traffic ; 14(8): 933-48, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23621784

ABSTRACT

Recycling is a limiting step for receptor-mediated endocytosis. We first report three in vitro or in vivo evidences that class III PI3K/VPS34 is the key PI3K isoform regulating apical recycling. A substractive approach, comparing in Opossum Kidney (OK) cells a pan-class I/II/III PI3K inhibitor (LY294002) with a class I/II PI3K inhibitor (ZSTK474), suggested that class III PI3K/VPS34 inhibition induced selective apical endosome swelling and sequestration of the endocytic receptor, megalin/LRP-2, causing surface down-regulation. GFP-(FYVE)x2 overexpression to sequester PI(3)P caused undistinguishable apical endosome swelling. In mouse kidney proximal tubular cells, conditional Vps34 inactivation also led to vacuolation and intracellular megalin redistribution. We next report that removal of LY294002 from LY294002-treated OK cells induced a spectacular burst of recycling tubules and restoration of megalin surface pool. Acute triggering of recycling tubules revealed recruitment of dynamin-GFP and dependence of dynamin-GTPase, guidance directionality by microtubules, and suggested that a microfilamentous net constrained endosomal swelling. We conclude that (i) besides its role in endosome fusion, PI3K-III is essential for endosome fission/recycling; and (ii) besides its role in endocytic entry, dynamin also supports tubulation of recycling endosomes. The unleashing of recycling upon acute reversal of PI3K inhibition may help study its dynamics and associated machineries.


Subject(s)
Class III Phosphatidylinositol 3-Kinases/metabolism , Dynamins/metabolism , Endosomes/metabolism , Animals , Cell Culture Techniques , Chromones/pharmacology , Class III Phosphatidylinositol 3-Kinases/antagonists & inhibitors , Class III Phosphatidylinositol 3-Kinases/genetics , Endocytosis , Enzyme Inhibitors/pharmacology , Inositol/analogs & derivatives , Inositol/pharmacology , Low Density Lipoprotein Receptor-Related Protein-2/genetics , Low Density Lipoprotein Receptor-Related Protein-2/metabolism , Mice , Morpholines/pharmacology , Opossums
11.
Hum Reprod ; 30(5): 1156-68, 2015 May.
Article in English | MEDLINE | ID: mdl-25750204

ABSTRACT

STUDY QUESTION: Does the endometrial functionalis have the potential to undergo self-renewal after menstruation and how is this process controlled by ovarian steroids? SUMMARY ANSWER: Endometrial xenografts subjected to withdrawal of estradiol and progesterone shrink but also show signs of proliferation and tissue repair; new estradiol supply prevents atrophy but is not sufficient to increase graft volume. WHAT IS KNOWN ALREADY: Menstruation, i.e. cyclic proteolysis of the extracellular matrix of endometrial functionalis, is induced by a fall in estrogen and progesterone concentration and is followed by tissue regeneration. However, there is debate about whether regenerating cells must originate from the basalis or from stem cells and whether new estrogen supply is required for the early repair concomitant with menstruation. STUDY DESIGN, SIZE, DURATION: Fragments from human endometrial functionalis (from 24 hysterectomy specimens) were xenografted in ovariectomized SCID mice and submitted to a 4-day estradiol and progesterone withdrawal (to mimic menstruation) followed by re-exposure to estradiol (to mimic the proliferative phase). We measured signs of proliferation and changes in graft volume. PARTICIPANTS/MATERIALS, SETTING, METHODS: Endometrium was collected from spontaneously cycling women. Cell proliferation was examined by immunolabeling Ki-67, cyclin D1 and phosphorylated-histone H3. Xenograft volume was measured by magnetic resonance imaging. Xenograft histomorphometry was performed to determine how the different tissue compartments contributed to volume change. MAIN RESULTS AND THE ROLE OF CHANCE: Hormone withdrawal induced a rapid decrease in graft volume mainly attributable to stroma condensation and breakdown, concomitant with an increase of proliferation markers. Reinsertion of estradiol pellets after induced menstruation blocked volume decrease and stimulated epithelial and stromal growth, but, surprisingly, did not induce graft enlargement. Reinsertion of both estradiol and progesterone pellets blocked apoptosis. LIMITATIONS, REASONS FOR CAUTION: Mechanisms of endometrial remodeling are different in women and mice and the contribution of circulating inflammatory cells in both species remains to be clarified. Moreover, during human menstruation, endometrial fragments resulting from tissue proteolysis can be expelled by the menstrual flow, unlike in this model. WIDER IMPLICATIONS OF THE FINDINGS: Menstruation is a multifocal event within the functionalis. This is the first evidence that endometrial fragments that are not shed after menstrual tissue breakdown can support endometrial regeneration. Endometriosis is commonly thought to result from the retrograde migration of menstrual fragments of the degraded functionalis into the peritoneal cavity. Our study supports their potential to regenerate as ectopic endometrium. STUDY FUNDING/COMPETING INTERESTS: This work was supported by the Fonds de la Recherche Scientifique Médicale, Concerted Research Actions, Communauté Française de Belgique, Région wallonne, Région bruxelloise and Loterie nationale. P.H. and B.F.J. are research associates of the Belgian Fonds de la Recherche Scientifique (F.R.S.-F.N.R.S.). E.M. is Associate Editor at Human Reproduction. There is no conflict of interest to declare.


Subject(s)
Endometrium/physiology , Endometrium/transplantation , Ovary/metabolism , Steroids/chemistry , Animals , Apoptosis , Cell Proliferation , Cyclin D1/metabolism , Endometriosis/metabolism , Estradiol/metabolism , Estrogen Receptor alpha/metabolism , Female , Heterografts/metabolism , Humans , Hysterectomy , Ki-67 Antigen/metabolism , Magnetic Resonance Imaging , Mice , Mice, SCID , Postmenopause , Progesterone/metabolism , Regeneration , Transplantation, Heterologous
12.
J Am Soc Nephrol ; 25(6): 1256-69, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24525030

ABSTRACT

Cystinosis, a main cause of Fanconi syndrome, is reproduced in congenic C57BL/6 cystinosin knockout (KO) mice. To identify the sequence of pathogenic and adaptation mechanisms of nephropathic cystinosis, we defined the onset of Fanconi syndrome in KO mice between 3 and 6 months of age and analyzed the correlation with structural and functional changes in proximal tubular cells (PTCs), with focus on endocytosis of ultrafiltrated disulfide-rich proteins as a key source of cystine. Despite considerable variation between mice at the same age, typical event sequences were delineated. At the cellular level, amorphous lysosomal inclusions preceded cystine crystals and eventual atrophy without crystals. At the nephron level, lesions started at the glomerulotubular junction and then extended distally. In situ hybridization and immunofluorescence revealed progressive loss of expression of megalin, cubilin, sodium-glucose cotransporter 2, and type IIa sodium-dependent phosphate cotransporter, suggesting apical dedifferentiation accounting for Fanconi syndrome before atrophy. Injection of labeled proteins revealed that defective endocytosis in S1 PTCs led to partial compensatory uptake by S3 PTCs, suggesting displacement of endocytic load and injury by disulfide-rich cargo. Increased PTC apoptosis allowed luminal shedding of cystine crystals and was partially compensated for by tubular proliferation. We conclude that lysosomal storage triggered by soluble cystine accumulation induces apical PTC dedifferentiation, which causes transfer of the harmful load of disulfide-rich proteins to more distal cells, possibly explaining longitudinal progression of swan-neck lesions. Furthermore, our results suggest that subsequent adaptation mechanisms include lysosomal clearance of free and crystalline cystine into urine and ongoing tissue repair.


Subject(s)
Adaptation, Physiological/physiology , Amino Acid Transport Systems, Neutral/genetics , Cystinosis/physiopathology , Fanconi Syndrome/physiopathology , Kidney Tubules, Proximal/physiopathology , Animals , Apoptosis/physiology , Cell Proliferation , Crystallization , Cystine/chemistry , Cystine/metabolism , Cystinosis/genetics , Cystinosis/pathology , Disease Models, Animal , Disease Progression , Endocytosis/physiology , Fanconi Syndrome/genetics , Fanconi Syndrome/pathology , Kidney Tubules, Proximal/pathology , Kidney Tubules, Proximal/physiology , Low Density Lipoprotein Receptor-Related Protein-2/genetics , Lysosomes/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Proteinuria/genetics , Proteinuria/pathology , Proteinuria/physiopathology , Receptors, Cell Surface/genetics , Vacuoles/pathology
13.
J Lipid Res ; 55(7): 1331-42, 2014 07.
Article in English | MEDLINE | ID: mdl-24826836

ABSTRACT

We recently reported that trace insertion of exogenous fluorescent (green BODIPY) analogs of sphingomyelin (SM) into living red blood cells (RBCs), partially spread onto coverslips, labels submicrometric domains, visible by confocal microscopy. We here extend this feature to endogenous SM, upon binding of a SM-specific nontoxic (NT) fragment of the earthworm toxin, lysenin, fused to the red monomeric fluorescent protein, mCherry [construct named His-mCherry-NT-lysenin (lysenin*)]. Specificity of lysenin* binding was verified with composition-defined liposomes and by loss of (125)I-lysenin* binding to erythrocytes upon SM depletion by SMase. The (125)I-lysenin* binding isotherm indicated saturation at 3.5 × 10(6) molecules/RBC, i.e., ∼3% of SM coverage. Nonsaturating lysenin* concentration also labeled sub-micrometric domains on the plasma membrane of partially spread erythrocytes, colocalizing with inserted green BODIPY-SM, and abrogated by SMase. Lysenin*-labeled domains were stable in time and space and were regulated by temperature and cholesterol. The abundance, size, positioning, and segregation of lysenin*-labeled domains from other lipids (BODIPY-phosphatidylcholine or -glycosphingolipids) depended on membrane tension. Similar lysenin*-labeled domains were evidenced in RBCs gently suspended in 3D-gel. Taken together, these data demonstrate submicrometric compartmentation of endogenous SM at the membrane of a living cell in vitro, and suggest it may be a genuine feature of erythrocytes in vivo.


Subject(s)
Erythrocyte Membrane/metabolism , Membrane Microdomains/metabolism , Sphingomyelins/pharmacology , Humans , Sphingomyelins/metabolism , Toxins, Biological/pharmacology
14.
Dev Biol ; 381(1): 227-40, 2013 Sep 01.
Article in English | MEDLINE | ID: mdl-23707896

ABSTRACT

The thyroid is a highly vascularized endocrine gland, displaying a characteristic epithelial organization in closed spheres, called follicles. Here we investigate how endothelial cells are recruited into the developing thyroid and if they control glandular organization as well as thyrocytes and C-cells differentiation. We show that endothelial cells closely surround, and then invade the expanding thyroid epithelial cell mass to become closely associated with nascent polarized follicles. This close and sustained endothelial:epithelial interaction depends on epithelial production of the angiogenic factor, Vascular Endothelial Growth Factor-A (VEGF-A), as its thyroid-specific genetic inactivation reduced the endothelial cell pool of the thyroid by > 90%. Vegfa KO also displayed decreased C-cells differentiation and impaired organization of the epithelial cell mass into follicles. We developed an ex vivo model of thyroid explants that faithfully mimicks bilobation of the thyroid anlagen, endothelial and C-cells invasion, folliculogenesis and differentiation. Treatment of thyroid explants at e12.5 with a VEGFR2 inhibitor ablated the endothelial pool and reproduced ex vivo folliculogenesis defects observed in conditional Vegfa KO. In the absence of any blood supply, rescue by embryonic endothelial progenitor cells restored folliculogenesis, accelerated lumen expansion and stimulated calcitonin expression by C-cells. In conclusion, our data demonstrate that, in developing mouse thyroid, epithelial production of VEGF-A is necessary for endothelial cells recruitment and expansion. In turn, endothelial cells control epithelial reorganization in follicles and C-cells differentiation.


Subject(s)
Endothelial Cells/cytology , Epithelial Cells/cytology , Gene Expression Regulation, Developmental , Thyroid Gland/embryology , Animals , Calcitonin/metabolism , Cell Differentiation , Endothelium/metabolism , Epithelium/metabolism , Female , Male , Mice , Mice, Knockout , Stem Cells/cytology , Thyroid Gland/blood supply , Thyroid Gland/growth & development , Vascular Endothelial Growth Factor A/metabolism , Vascular Endothelial Growth Factor Receptor-2/metabolism
15.
Biochim Biophys Acta ; 1833(12): 3076-3092, 2013 Dec.
Article in English | MEDLINE | ID: mdl-23994617

ABSTRACT

Trypanosomatids contain peroxisome-like organelles called glycosomes. Peroxisomal biogenesis involves a cytosolic receptor, PEX5, which, after its insertion into the organellar membrane, delivers proteins to the matrix. In yeasts and mammalian cells, transient PEX5 monoubiquitination at the membrane serves as the signal for its retrieval from the organelle for re-use. When its recycling is impaired, PEX5 is polyubiquitinated for proteasomal degradation. Stably monoubiquitinated TbPEX5 was detected in cytosolic fractions of Trypanosoma brucei, indicative for its role as physiological intermediate in receptor recycling. This modification's resistance to dithiothreitol suggests ubiquitin conjugation of a lysine residue. T. brucei PEX4, the functional homologue of the ubiquitin-conjugating (UBC) enzyme responsible for PEX5 monoubiquitination in yeast, was identified. It is associated with the cytosolic face of the glycosomal membrane, probably anchored by an identified putative TbPEX22. The involvement of TbPEX4 in TbPEX5 ubiquitination was demonstrated using procyclic ∆PEX4 trypanosomes. Surprisingly, glycosomal matrix protein import was only mildly affected in this mutant. Since other UBC homologues were upregulated, it might be possible that these have partially rescued PEX4's function in PEX5 ubiquitination. In addition, the altered expression of UBCs, notably of candidates involved in cell-cycle control, could be responsible for observed morphological and motility defects of the ∆PEX4 mutant.


Subject(s)
Microbodies/metabolism , Protozoan Proteins/metabolism , Trypanosoma brucei brucei/metabolism , Ubiquitination , Animals , Cell Line , Cytosol/metabolism , Gene Expression Regulation , Gene Knockout Techniques , Intracellular Membranes/metabolism , Life Cycle Stages , Models, Biological , Protein Transport , Protozoan Proteins/genetics , Reproducibility of Results , Trypanosoma brucei brucei/genetics , Trypanosoma brucei brucei/growth & development , Trypanosoma brucei brucei/ultrastructure , Ubiquitin-Conjugating Enzymes/metabolism
16.
BMC Mol Biol ; 15: 9, 2014 May 16.
Article in English | MEDLINE | ID: mdl-24885929

ABSTRACT

BACKGROUND: YBX3/ZONAB/CSDA is an epithelial-specific transcription factor acting in the density-based switch between proliferation and differentiation. Our laboratory reported overexpression of YBX3 in clear cell renal cell arcinoma (ccRCC), as part of a wide study of YBX3 regulation in vitro and in vivo. The preliminary data was limited to 5 cases, of which only 3 could be compared to paired normal tissue, and beta-Actin was used as sole reference to normalize gene expression. We thus decided to re-evaluate YBX3 expression by real-time-PCR in a larger panel of ccRCC samples, and their paired healthy tissue, with special attention on experimental biases such as inter-individual variations, primer specificity, and reference gene for normalization. RESULTS: Gene expression was measured by RT-qPCR in 16 ccRCC samples, each compared to corresponding healthy tissue to minimize inter-individual variations. Eight potential housekeeping genes were evaluated for expression level and stability among the 16-paired samples. Among tested housekeeping genes, PPIA and RPS13, especially in combination, proved best suitable to normalize gene expression in ccRCC tissues as compared to classical reference genes such as beta-Actin, GAPDH, 18S or B2M. Using this pair as reference, YBX3 expression level among a collection of 16 ccRCC tumors was not significantly increased as compared to normal adjacent tissues. However, stratification according to Fuhrman grade disclosed higher YBX3 expression levels in low-grade tumors and lower in high-grade tumors. Immunoperoxidase confirmed homogeneous nuclear staining for YBX3 in low-grade but revealed nuclear heterogeneity in high-grade tumors. CONCLUSIONS: This paper underlines that special attention to reference gene products in the design of real-time PCR analysis of tumoral tissue is crucial to avoid misleading conclusions. Furthermore, we found that global YBX3/ZONAB/CSDA mRNA expression level may be considered within a "signature" of RCC grading.


Subject(s)
CCAAT-Enhancer-Binding Proteins/genetics , Carcinoma, Renal Cell/genetics , Gene Expression Regulation, Neoplastic/genetics , Gene Expression/genetics , Genes, Essential/genetics , Heat-Shock Proteins/genetics , Kidney Neoplasms/genetics , Actins/genetics , Adult , Aged , DNA-Binding Proteins/genetics , Female , Humans , Male , Middle Aged , Ribosomal Proteins/genetics
17.
Mol Hum Reprod ; 20(8): 767-75, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24810263

ABSTRACT

Matrix metalloproteinases (MMPs) are key enzymes involved in extracellular matrix remodelling. In the human endometrium, the expression and activity of several MMPs are maximal during the menstrual phase. Moreover, MMPs are thought to be involved in the pathogenesis of endometriosis and cancers, in particular with invasion and metastasis. We recently reported that MMP-27 is a unique MMP with an intracellular retention motif. We investigated the expression and cellular localization of MMP-27 in the cycling human endometrium and in endometriotic lesions. MMP-27 mRNA was detected throughout the menstrual cycle. Despite large interpatient variations, mRNA levels increased from the proliferative to the secretory phase, to peak during the menstrual phase. MMP-27 was immunolocalized in large isolated cells scattered throughout the stroma and around blood vessels: these cells were most abundant at menstruation and were identified by immunofluorescence as CD45(+), CD163(+) and CD206(+) macrophages. CD163(+) macrophages were also abundant in endometriotic lesions, but showed different patterns in ovarian or peritoneal endometriotic lesions (co-labelling for CD206 and MMP-27) and rectovaginal lesions (no co-labelling). In conclusion, MMP-27 is expressed in a subset of endometrial macrophages related to menstruation and in ovarian and peritoneal endometriotic lesions.


Subject(s)
Antigens, CD/metabolism , Antigens, Differentiation, Myelomonocytic/metabolism , Endometriosis/metabolism , Endometrium/metabolism , Lectins, C-Type/metabolism , Macrophages/metabolism , Mannose-Binding Lectins/metabolism , Matrix Metalloproteinases, Secreted/metabolism , Receptors, Cell Surface/metabolism , Endometriosis/genetics , Female , Fluorescent Antibody Technique , Humans , Immunohistochemistry , In Vitro Techniques , Mannose Receptor , Matrix Metalloproteinases, Secreted/genetics , Menstrual Cycle/genetics , Menstrual Cycle/metabolism , Real-Time Polymerase Chain Reaction
18.
Blood ; 119(20): 4625-35, 2012 May 17.
Article in English | MEDLINE | ID: mdl-22378845

ABSTRACT

The constitutively active JAK2 V617F mutant is the major determinant of human myeloproliferative neoplasms (MPNs). We show that coexpression of murine JAK2 V617F and the murine thrombopoietin (Tpo) receptor (TpoR, c-MPL) in hematopoietic cell lines or heterozygous knock-in of JAK2 V617F in mice leads to down-modulation of TpoR levels. Enhanced TpoR ubiquitinylation, proteasomal degradation, reduced recycling, and maturation are induced by the constitutive JAK2 V617F activity. These effects can be prevented in cell lines by JAK2 and proteasome inhibitors. Restoration of TpoR levels by inhibitors could be detected in platelets from JAK2 inhibitor-treated myelofibrosis patients that express the JAK2 V617F mutant, and in platelets from JAK2 V617F knock-in mice that were treated in vivo with JAK2 or proteasome inhibitors. In addition, we show that Tpo can induce both proliferative and antiproliferative effects via TpoR at low and high JAK2 activation levels, respectively, or on expression of JAK2 V617F. The antiproliferative signaling and receptor down-modulation by JAK2 V617F were dependent on signaling via TpoR cytosolic tyrosine 626. We propose that selection against TpoR antiproliferative signaling occurs by TpoR down-modulation and that restoration of down-modulated TpoR levels could become a biomarker for the treatment of MPNs.


Subject(s)
Janus Kinase 2/antagonists & inhibitors , Janus Kinase 2/physiology , Proteasome Inhibitors , Protein Kinase Inhibitors/pharmacology , Receptors, Thrombopoietin/genetics , Amino Acid Substitution/genetics , Amino Acid Substitution/physiology , Animals , Cell Membrane/drug effects , Cell Membrane/metabolism , Cells, Cultured , Down-Regulation/drug effects , Drug Evaluation, Preclinical , HEK293 Cells , Humans , Janus Kinase 2/genetics , Janus Kinase 2/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , Mutation, Missense/physiology , Phenylalanine/genetics , Receptor Cross-Talk/drug effects , Receptor Cross-Talk/physiology , Receptors, Thrombopoietin/metabolism , Signal Transduction/drug effects , Valine/genetics
19.
FASEB J ; 27(9): 3711-9, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23729593

ABSTRACT

Menstrual endometrial breakdown induced by estradiol and progesterone withdrawal is regularly attributed to vasospasm of spiral arteries causing ischemia and hypoxia. We investigated whether hypoxia actually occurred in an in vivo model of menstruation. Three complementary approaches were used to look for signs of hypoxia in fragments of human functionalis xenografted to ovariectomized immunodeficient mice bearing pellets-releasing estradiol and progesterone, and then deprived of ovarian steroids. Hormone withdrawal 21 d after grafting induced menstrual breakdown and MMP expression within 4 d. Local partial oxygen pressure (pO2) was measured by electron paramagnetic resonance using implanted lithium phtalocyanine crystals. In mice with hormone maintenance until sacrifice, pO2 was low one week after grafting (14.8±3.4 mmHg) but increased twofold from the second week when tissue was largely revascularized. After 3 wk, pO2 was not modified by hormone withdrawal but was slightly increased on hormone reimpregnation 4 d after removal (34.7±6.1 mmHg) by comparison with hormone maintenance (27.1±8.6 mmHg). These results were confirmed using fluorescence quenching-based OxyLite measurements. In a further search for signs of hypoxia, we did not find significant HIF1-α immunostaining, nor pimonidazole adducts after hormone withdrawal. We conclude that hypoxia is not needed to trigger menstrual-like tissue breakdown or repair in human endometrial xenograft.


Subject(s)
Endometrium/metabolism , Hypoxia/metabolism , Menstrual Cycle/metabolism , Transplantation, Heterologous , Animals , Female , Humans , Hypoxia/genetics , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Matrix Metalloproteinases/genetics , Menstrual Cycle/genetics , Mice , Ovariectomy
20.
Biochem J ; 455(2): 195-206, 2013 Oct 15.
Article in English | MEDLINE | ID: mdl-23905686

ABSTRACT

PIKfyve (FYVE domain-containing phosphatidylinositol 3-phosphate 5-kinase), the lipid kinase that phosphorylates PtdIns3P to PtdIns(3,5)P2, has been implicated in insulin-stimulated glucose uptake. We investigated whether PIKfyve could also be involved in contraction/AMPK (AMP-activated protein kinase)-stimulated glucose uptake in skeletal muscle. Incubation of rat epitrochlearis muscles with YM201636, a selective PIKfyve inhibitor, reduced contraction- and AICAriboside (5-amino-4-imidazolecarboxamide riboside)-stimulated glucose uptake. Consistently, PIKfyve knockdown in C2C12 myotubes reduced AICAriboside-stimulated glucose transport. Furthermore, muscle contraction increased PtdIns(3,5)P2 levels and PIKfyve phosphorylation. AMPK phosphorylated PIKfyve at Ser307 both in vitro and in intact cells. Following subcellular fractionation, PIKfyve recovery in a crude intracellular membrane fraction was increased in contracting versus resting muscles. Also in opossum kidney cells, wild-type, but not S307A mutant, PIKfyve was recruited to endosomal vesicles in response to AMPK activation. We propose that PIKfyve activity is required for the stimulation of skeletal muscle glucose uptake by contraction/AMPK activation. PIKfyve is a new AMPK substrate whose phosphorylation at Ser307 could promote PIKfyve translocation to endosomes for PtdIns(3,5)P2 synthesis to facilitate GLUT4 (glucose transporter 4) translocation.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Glucose/metabolism , Muscle Contraction/physiology , Muscle, Skeletal/enzymology , Muscle, Skeletal/metabolism , Phosphatidylinositol 3-Kinase/metabolism , AMP-Activated Protein Kinases/genetics , Aminoimidazole Carboxamide/analogs & derivatives , Aminoimidazole Carboxamide/metabolism , Animals , Cell Line , Glucose Transporter Type 4/metabolism , Humans , Insulin/metabolism , Male , Opossums , Phosphatidylinositol 3-Kinase/genetics , Phosphatidylinositol Phosphates/metabolism , Phosphorylation , Rats , Rats, Wistar
SELECTION OF CITATIONS
SEARCH DETAIL