Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
Add more filters










Publication year range
1.
EMBO J ; 41(23): e112402, 2022 12 01.
Article in English | MEDLINE | ID: mdl-36321514

ABSTRACT

Partial Retraction of: The EMBO Journal (2010) 29: 3607-3620. DOI: 10.1038/emboj.2010.237 | Published online 24 September 2010 Journal statement The journal contacted the authors in February 2022 about potential image insertions and duplications in Fig 4A and 4E. In the absence of source data, the authors are retracting Fig 4A, the lower panel of Fig 4E (LAMP1 immunoblot), and the following statements in the text that rely on these data: "Quantitative analysis showed that the percentage of Flotillin-1 associated with DRMs was increased in LSD endolysosomal membranes (Figure 4A), indicating an increased amount of cholesterol-enriched regions in these membrane samples." "LAMP1 also displayed a similar distribution profile in WT and LSD cells (Figure 4E)". Author statement The authors could not verify the aberrations in panel A of Fig 4 and the lower immunoblot (LAMP1) of 4E because the original source data are no longer available (12 years after publication, which is beyond the institute's 10-year data retention policy). The authors wish to clarify that the main conclusions of the paper are not affected by the retraction of Figure panels 4A and 4E for the following reasons: Figure panel 4A supports the observation that there are increased cholesterol-enhanced regions in LSD samples. This finding is also supported by data provided in figs 4B, 4C and 4D. Figure panel 4E: The LAMP1 blot in Fig 4E shows that the distribution of protein normally excluded from DRMs is not altered between Wt and LSD samples. This result is also supported by the upper blot in this panel (Transferrin receptor). The authors apologize for these errors and agree with this corrigendum; no response could be obtained from AL.

2.
EMBO J ; 38(3)2019 02 01.
Article in English | MEDLINE | ID: mdl-30591554

ABSTRACT

Transcription factor TFEB is thought to control cellular functions-including in the vascular bed-primarily via regulation of lysosomal biogenesis and autophagic flux. Here, we report that TFEB also orchestrates a non-canonical program that controls the cell cycle/VEGFR2 pathway in the developing vasculature. In endothelial cells, TFEB depletion halts proliferation at the G1-S transition by inhibiting the CDK4/Rb pathway. TFEB-deficient cells attempt to compensate for this limitation by increasing VEGFR2 levels at the plasma membrane via microRNA-mediated mechanisms and controlled membrane trafficking. TFEB stimulates expression of the miR-15a/16-1 cluster, which limits VEGFR2 transcript stability and negatively modulates expression of MYO1C, a regulator of VEGFR2 trafficking to the cell surface. Altered levels of miR-15a/16-1 and MYO1C in TFEB-depleted cells cause increased expression of plasma membrane VEGFR2, but in a manner associated with low signaling strength. An endothelium-specific Tfeb-knockout mouse model displays defects in fetal and newborn mouse vasculature caused by reduced endothelial proliferation and by anomalous function of the VEGFR2 pathway. These previously unrecognized functions of TFEB expand its role beyond regulation of the autophagic pathway in the vascular system.


Subject(s)
Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/physiology , Cell Proliferation , Embryo, Mammalian/cytology , Endothelium, Vascular/cytology , Gene Expression Regulation, Developmental , Neovascularization, Physiologic , Vascular Endothelial Growth Factor Receptor-2/metabolism , Animals , Cells, Cultured , Embryo, Mammalian/physiology , Endothelium, Vascular/physiology , Female , Mice , Mice, Inbred C57BL , Mice, Knockout , Signal Transduction , Vascular Endothelial Growth Factor Receptor-2/genetics
3.
J Appl Genet ; 58(4): 481-486, 2017 Nov.
Article in English | MEDLINE | ID: mdl-29019057

ABSTRACT

Duplications of Yq arm (and AZF) seems to be tolerated by fertile males, while mutations, deletions, duplications or haploinsufficiency of SHOX can originate a wide range of phenotypes, including short stature and skeletal abnormalities. We report a case of non-obstructive azoospermia in a young man with short stature, skeletal anomalies, normal intelligence and hormonal parameters. This male showed a very singular Y-chromosome aberration, consisting of a duplication of Yq and proximal regions of Yp, with a deletion of almost all PAR1 in Yptel, including SHOX. CBA- and RBA-banding and FISH-mapping with telomeric, centromeric, AZF and SHOX probes were used. These results were confirmed by array CGH, which revealed the following karyotype constitution: arr [hg19] Xp22.33 or Yp11.32p11.31 (310,932-2,646,815 or 260,932-2,596,815) ×1, Yp11.2q12 (8,641,183-59,335,913) ×2. We conclude that the haploinsufficience of SHOX may be the cause of short stature and skeletal defects in the patient, while the non-obstructive azoospermia could be related to the lack of X-Y pairing during meiosis originated by the anomalous configuration of this chromosome abnormality and large deletion which occurred in Yp-PAR1.


Subject(s)
Abnormalities, Multiple/genetics , Azoospermia/genetics , Chromosomes, Human, Y/genetics , Receptor, PAR-1/genetics , Sequence Deletion/genetics , Adult , Chromosome Aberrations , Humans , Male , Young Adult
4.
Sci Immunol ; 2(16)2017 10 27.
Article in English | MEDLINE | ID: mdl-29079589

ABSTRACT

Dendritic cells (DCs) patrol their environment by linking antigen acquisition by macropinocytosis to cell locomotion. DC activation upon bacterial sensing inhibits macropinocytosis and increases DC migration, thus promoting the arrival of DCs to lymph nodes for antigen presentation to T cells. The signaling events that trigger such changes are not fully understood. We show that lysosome signaling plays a critical role in this process. Upon bacterial sensing, lysosomal calcium is released by the ionic channel TRPML1 (transient receptor potential cation channel, mucolipin subfamily, member 1), which activates the actin-based motor protein myosin II at the cell rear, promoting fast and directional migration. Lysosomal calcium further induces the activation of the transcription factor EB (TFEB), which translocates to the nucleus to maintain TRPML1 expression. We found that the TRPML1-TFEB axis results from the down-regulation of macropinocytosis after bacterial sensing by DCs. Lysosomal signaling therefore emerges as a hitherto unexpected link between macropinocytosis, actomyosin cytoskeleton organization, and DC migration.


Subject(s)
Cell Movement , Dendritic Cells/immunology , Lysosomes/metabolism , Signal Transduction , Animals , Antigen Presentation , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/genetics , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/metabolism , Calcium/metabolism , Cell Differentiation , Cell Nucleus/metabolism , Cytoskeleton/metabolism , Dendritic Cells/physiology , Down-Regulation , Lysosomes/immunology , Mice , Myosin Type II/genetics , Myosin Type II/metabolism , Pinocytosis , Transient Receptor Potential Channels/deficiency , Transient Receptor Potential Channels/genetics , Transient Receptor Potential Channels/metabolism
5.
J Prenat Med ; 10(3-4): 20-22, 2016.
Article in English | MEDLINE | ID: mdl-28725342

ABSTRACT

We report a case of a male fetus of 20 weeks of gestation with plurimalformed observed by transonic scan and confirmed by MR. The karyotype was 46, XY. Molecular analysis showed a microdeletion of about 100 kb in the CTNNA3 gene.

6.
Cardiovasc Revasc Med ; 16(3): 141-6, 2015.
Article in English | MEDLINE | ID: mdl-25708003

ABSTRACT

AIM: An innovative xenon-chlorine (excimer) pulsed laser catheter (ELCA X80) has been recently used for the treatment of complex coronary lesions, as calcified stenosis, chronic total occlusions and non-compliant plaques. Such complex lesions are difficult to adequately treat with balloon angioplasty and/or intracoronary stenting. The aim of this study was to examine the acute outcome of this approach on a cohort of patients with coronary lesions. METHODS AND RESULTS: Eighty patients with 100 lesions were enrolled through four centers, and excimer laser coronary angioplasty was performed on 96 lesions (96%). Safety and effectiveness data were compared between patients treated with standard laser therapy and those treated with increased laser therapy. Laser success was obtained in 90 lesions (93.7%), procedural success was reached in 88 lesions (91.7%), and clinical success in was obtained in 87 lesions (90.6%). There was no perforation, major side branch occlusion, spasm, no-reflow phenomenon, dissection nor acute vessel closure. Increased laser parameters were used successfully for 49 resistant lesions without complications. CONCLUSIONS: This study suggests that laser-facilitated coronary angioplasty is a simple, safe and effective device for the management of complex coronary lesions. Furthermore, higher laser energy levels delivered by this catheter improved the device performance without increasing complications.


Subject(s)
Angioplasty, Balloon, Laser-Assisted , Atherectomy, Coronary , Coronary Angiography , Stents , Adult , Aged , Aged, 80 and over , Angioplasty, Balloon, Coronary/methods , Angioplasty, Balloon, Laser-Assisted/methods , Atherectomy, Coronary/methods , Coronary Angiography/methods , Female , Humans , Male , Middle Aged , Myocardial Revascularization/methods , Time Factors , Treatment Outcome
7.
J Prenat Med ; 8(3-4): 42-6, 2014.
Article in English | MEDLINE | ID: mdl-26266000

ABSTRACT

OBJECTIVE: the purpose of this paper is to report the first case of megaurethra in a fetus with Meckel syndrome and in a fetus with femal pseudoermaphroditism. RESULTS: the former case refers to a fetus of 13 weeks gestation with the three following prominent anomalies, observed by transonic scan and confirmed by autopsy: congenital megaurethra, anal atresia, single umbelical artery. The latter case refers to a fetus of 18 weeks gestation. Autopsy confirmed penile malformation and revealed ovaries in the abdomen. The karyotype was 46,XX with normal molecular karytype. The megaurethra was discovered by sonography at 18 weeks gestation. Autopsy confirmed penile malformation and revealed ovaries in the abdomen. The karyotype was 46,XX with normal molecular karyotype (Array-CGH, 1 Mb of resolution). METHODS: transonic scan, autopsy, karyotype, array-CGH. CONCLUSIONS: the first prenatal cases of two genetic syndromes with megaurethra have been reported, concening respectively a fetus with Meckel syndrome and a fetus with femal pseudoermaphroditism. The latter was confirmed by both autopsy and the normal female 46,XX karyotype.

8.
Int J Oncol ; 43(6): 1763-70, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24101161

ABSTRACT

Statins are a class of drugs that inhibit the rate-limiting step in the cholesterol biosynthetic pathway and show an anticancer effect, probably through the inhibition of cell proliferation. To date, the exact mechanism of cancer cell growth arrest induced by statins is not known. We report that simvastatin is able to induce apoptosis in melanoma cells but not in normal cells and also able to contrast the growth of tumor in an experimental melanoma murine model. We observed a delay in the tumor development in almost the 50% of the simvastatin administered animals and a strong reduction of the tumor volume with a differences of ~150% compared to the controls. Also the survival rate was significantly higher in mice that received the drug with a survival increase of ~130% compared to the controls. The tumor growth reduction in mice was supported by the results of cell migration assay, confirming that simvastatin clearly reduced cell migration. Moreover, simvastatin induced a strong downregulation of NonO gene expression, an important growth factor involved in the splicing regulation. This result could explain the decrease of melanoma cells proliferation, suggesting a possible action mechanism. The results derived from our experiments may sustain the many reports on the anticancer inhibitory property of statins and encourage new studies on this drug for a possible use in therapy, probably in combination with conventional chemotherapy.


Subject(s)
Apoptosis/drug effects , Hydroxymethylglutaryl-CoA Reductase Inhibitors/therapeutic use , Melanoma, Experimental/drug therapy , Simvastatin/therapeutic use , Skin Neoplasms/drug therapy , 3T3 Cells , Animals , Antineoplastic Agents/therapeutic use , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Cholesterol/biosynthesis , DNA-Binding Proteins/biosynthesis , Disease Progression , Humans , Melanoma, Experimental/mortality , Mice , Mice, Inbred C57BL , RNA-Binding Proteins , Skin Neoplasms/mortality , Survival , Survival Rate , Wound Healing/drug effects
9.
Autophagy ; 9(7): 1117-8, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23669057

ABSTRACT

It is hard to find an area of biology in which autophagy is not involved. In fact, the topic extends beyond scientific research to stimulate intellectual exercise and entertainment-autophagy has found its way into a crossword puzzle (Klionsky, 2013). We have found yet another function of autophagy while searching for a better treatment for Pompe disease, a devastating metabolic myopathy resulting from excessive lysosomal glycogen storage. To relieve this glycogen burden, we stimulated lysosomal exocytosis through upregulation of transcription factor EB (TFEB). Overexpression of TFEB in Pompe muscle clears the cells of enlarged lysosomes, reduces glycogen levels, and alleviates autophagic buildup, the major secondary abnormality in Pompe disease. Unexpectedly, the process of exocytosis does not seem to be a purely lysosomal event; vesicles arranged along the plasma membrane are double-labeled with the lysosomal marker LAMP1 and the autophagosomal marker LC3, indicating that TFEB induces the exocytosis of autolysosomes. Furthermore, the effects of TFEB are almost abrogated in autophagy-deficient Pompe mice, suggesting a previously unrecognized role of autophagy in TFEB-mediated cellular clearance.


Subject(s)
Autophagy , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/metabolism , Exocytosis , Glycogen Storage Disease Type II/metabolism , Glycogen Storage Disease Type II/pathology , Lysosomes/metabolism , Animals , Humans , Lysosomes/pathology , Mice , Phagosomes/metabolism
10.
EMBO Mol Med ; 5(5): 691-706, 2013 May.
Article in English | MEDLINE | ID: mdl-23606558

ABSTRACT

A recently proposed therapeutic approach for lysosomal storage disorders (LSDs) relies upon the ability of transcription factor EB (TFEB) to stimulate autophagy and induce lysosomal exocytosis leading to cellular clearance. This approach is particularly attractive in glycogen storage disease type II [a severe metabolic myopathy, Pompe disease (PD)] as the currently available therapy, replacement of the missing enzyme acid alpha-glucosidase, fails to reverse skeletal muscle pathology. PD, a paradigm for LSDs, is characterized by both lysosomal abnormality and dysfunctional autophagy. Here, we show that TFEB is a viable therapeutic target in PD: overexpression of TFEB in a new muscle cell culture system and in mouse models of the disease reduced glycogen load and lysosomal size, improved autophagosome processing, and alleviated excessive accumulation of autophagic vacuoles. Unexpectedly, the exocytosed vesicles were labelled with lysosomal and autophagosomal membrane markers, suggesting that TFEB induces exocytosis of autophagolysosomes. Furthermore, the effects of TFEB were almost abrogated in the setting of genetically suppressed autophagy, supporting the role of autophagy in TFEB-mediated cellular clearance.


Subject(s)
Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/metabolism , Glycogen Storage Disease Type II/enzymology , Adenoviridae/genetics , Animals , Autophagy , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/chemistry , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/genetics , Cells, Cultured , Disease Models, Animal , Exocytosis , Genetic Vectors/genetics , Genetic Vectors/metabolism , Glycogen/metabolism , Glycogen Storage Disease Type II/pathology , Lysosomes/metabolism , Mice , Mice, Knockout , Muscle, Skeletal/cytology , Muscle, Skeletal/metabolism , Muscle, Skeletal/ultrastructure , alpha-Glucosidases/deficiency , alpha-Glucosidases/genetics , alpha-Glucosidases/metabolism
11.
EMBO Mol Med ; 5(5): 675-90, 2013 May.
Article in English | MEDLINE | ID: mdl-23568409

ABSTRACT

Mucopolysaccharidoses type IIIA (MPS-IIIA) is a neurodegenerative lysosomal storage disorder (LSD) caused by inherited defects of the sulphamidase gene. Here, we used a systemic gene transfer approach to demonstrate the therapeutic efficacy of a chimeric sulphamidase, which was engineered by adding the signal peptide (sp) from the highly secreted iduronate-2-sulphatase (IDS) and the blood-brain barrier (BBB)-binding domain (BD) from the Apolipoprotein B (ApoB-BD). A single intravascular administration of AAV2/8 carrying the modified sulphamidase was performed in adult MPS-IIIA mice in order to target the liver and convert it to a factory organ for sustained systemic release of the modified sulphamidase. We showed that while the IDS sp replacement results in increased enzyme secretion, the addition of the ApoB-BD allows efficient BBB transcytosis and restoration of sulphamidase activity in the brain of treated mice. This, in turn, resulted in an overall improvement of brain pathology and recovery of a normal behavioural phenotype. Our results provide a novel feasible strategy to develop minimally invasive therapies for the treatment of brain pathology in MPS-IIIA and other neurodegenerative LSDs.


Subject(s)
Blood-Brain Barrier/metabolism , Brain/physiology , Iduronate Sulfatase/metabolism , Mucopolysaccharidosis III/enzymology , Animals , Apolipoproteins B/chemistry , Apolipoproteins B/metabolism , Brain/pathology , Cell Line , Dependovirus/genetics , Disease Models, Animal , Gene Transfer Techniques , Genetic Vectors/genetics , Genetic Vectors/metabolism , Iduronate Sulfatase/genetics , Liver/metabolism , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice , Mice, Inbred C57BL , Mucopolysaccharidosis III/genetics , Mucopolysaccharidosis III/pathology , Phenotype , Protein Engineering , Protein Structure, Tertiary , Recombinant Fusion Proteins/biosynthesis , Recombinant Fusion Proteins/genetics , Serine Endopeptidases/genetics , Serine Endopeptidases/metabolism , Transcytosis
12.
Int J Oncol ; 40(4): 935-41, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22134829

ABSTRACT

The statins (3-hydroxy-3-methylglutaryl coenzyme A reductase inhibitors) have been proven to be effective in lowering cholesterol and as anti-lipid agents against cardiovascular disease. Recent reports demonstrate an anticancer effect induced by the statins through inhibition of cell proliferation. Probably, these effects are due to suppression of the mevalonate pathway leading to the depletion of various downstream products that play an essential role in cell cycle progression, cell signaling and membrane integrity. To date, although many hypotheses have been proposed, the exact mechanism at the basis of cancer cell growth arrest induced by statins is not known. In this study, we have demonstrated that simvastatin, at a dose of 20 µM for 24-72 h, induced in cancer cells but not in normal cells precise features of apoptosis including increased DNA fragmentation while, at the molecular level simvastatin induced overexpression of the pro-apoptotic gene Bax together with an inhibition of BCL-2, the gene that has the well-known function of protecting cells from apoptosis. The simvastatin-mediated induction of apoptosis in similar cancer cells but not in normal cells is very interesting and may be at the basis of cancer therapy using statins, usually in combination with chemotherapy or to be used as a cancer protective drug. Simvastatin may, thus, play a dual prophylactic role as a lipid-lowering drug for the prevention of heart disease and as an anticancer agent to prevent certain types of cancers.


Subject(s)
Apoptosis/drug effects , Genes, bcl-2/drug effects , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology , Neoplasms/drug therapy , Proto-Oncogene Proteins c-bcl-2/biosynthesis , Simvastatin/pharmacology , bcl-2-Associated X Protein/metabolism , Apoptosis/genetics , Blotting, Western , Cell Line, Tumor , DNA Fragmentation/drug effects , Down-Regulation/drug effects , Gene Expression/drug effects , Hep G2 Cells , Humans , In Situ Nick-End Labeling , Neoplasms/pathology , Proto-Oncogene Proteins c-bcl-2/genetics , Proto-Oncogene Proteins c-bcl-2/metabolism , Real-Time Polymerase Chain Reaction , bcl-2-Associated X Protein/biosynthesis , bcl-2-Associated X Protein/genetics
13.
Dev Cell ; 21(3): 421-30, 2011 Sep 13.
Article in English | MEDLINE | ID: mdl-21889421

ABSTRACT

Lysosomes are cellular organelles primarily involved in degradation and recycling processes. During lysosomal exocytosis, a Ca²âº-regulated process, lysosomes are docked to the cell surface and fuse with the plasma membrane (PM), emptying their content outside the cell. This process has an important role in secretion and PM repair. Here we show that the transcription factor EB (TFEB) regulates lysosomal exocytosis. TFEB increases the pool of lysosomes in the proximity of the PM and promotes their fusion with PM by raising intracellular Ca²âº levels through the activation of the lysosomal Ca²âº channel MCOLN1. Induction of lysosomal exocytosis by TFEB overexpression rescued pathologic storage and restored normal cellular morphology both in vitro and in vivo in lysosomal storage diseases (LSDs). Our data indicate that lysosomal exocytosis may directly modulate cellular clearance and suggest an alternative therapeutic strategy for disorders associated with intracellular storage.


Subject(s)
Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/metabolism , Exocytosis/genetics , Lysosomes/metabolism , TRPM Cation Channels/genetics , Transcriptional Activation , Animals , Apoptosis , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/genetics , COS Cells , Calcium/metabolism , Cell Membrane/physiology , Chlorocebus aethiops , Disease Models, Animal , HeLa Cells , Humans , Lysosomes/genetics , Membrane Fusion , Mice , Multiple Sulfatase Deficiency Disease/genetics , Multiple Sulfatase Deficiency Disease/metabolism , Multiple Sulfatase Deficiency Disease/pathology , Transient Receptor Potential Channels , Up-Regulation/drug effects
14.
Mol Ther ; 19(5): 860-9, 2011 May.
Article in English | MEDLINE | ID: mdl-21326216

ABSTRACT

Multiple sulfatase deficiency (MSD), a severe autosomal recessive disease is caused by mutations in the sulfatase modifying factor 1 gene (Sumf1). We have previously shown that in the Sumf1 knockout mouse model (Sumf1(-/-)) sulfatase activities are completely absent and, similarly to MSD patients, this mouse model displays growth retardation and early mortality. The severity of the phenotype makes MSD unsuitable to be treated by enzyme replacement or bone marrow transplantation, hence the importance of testing the efficacy of novel treatment strategies. Here we show that recombinant adeno-associated virus serotype 9 (rAAV9) vector injected into the cerebral ventricles of neonatal mice resulted in efficient and widespread transduction of the brain parenchyma. In addition, we compared a combined, intracerebral ventricles and systemic, administration of an rAAV9 vector encoding SUMF1 gene to the single administrations-either directly in brain, or systemic alone -in MSD mice. The combined treatment resulted in the global activation of sulfatases, near-complete clearance of glycosaminoglycans (GAGs) and decrease of inflammation in both the central nervous system (CNS) and visceral organs. Furthermore, behavioral abilities were improved by the combined treatment. These results underscore that the "combined" mode of rAAV9 vector administration is an efficient option for the treatment of severe whole-body disorders.


Subject(s)
Genetic Therapy , Multiple Sulfatase Deficiency Disease/genetics , Multiple Sulfatase Deficiency Disease/therapy , Sulfatases/metabolism , Animals , Blotting, Western , Central Nervous System/immunology , Central Nervous System/pathology , Cerebral Ventricles/virology , Dependovirus/genetics , Disease Models, Animal , Fluorescent Antibody Technique , Gene Transfer Techniques , Genes, Transgenic, Suicide , Genetic Vectors , Glycosaminoglycans/metabolism , Inflammation/therapy , Mice , Mice, Inbred C57BL , Oxidoreductases Acting on Sulfur Group Donors , Sulfatases/deficiency
15.
EMBO J ; 29(21): 3607-20, 2010 Nov 03.
Article in English | MEDLINE | ID: mdl-20871593

ABSTRACT

The function of lysosomes relies on the ability of the lysosomal membrane to fuse with several target membranes in the cell. It is known that in lysosomal storage disorders (LSDs), lysosomal accumulation of several types of substrates is associated with lysosomal dysfunction and impairment of endocytic membrane traffic. By analysing cells from two severe neurodegenerative LSDs, we observed that cholesterol abnormally accumulates in the endolysosomal membrane of LSD cells, thereby reducing the ability of lysosomes to efficiently fuse with endocytic and autophagic vesicles. Furthermore, we discovered that soluble N-ethylmaleimide-sensitive factor attachment protein (SNAP) receptors (SNAREs), which are key components of the cellular membrane fusion machinery are aberrantly sequestered in cholesterol-enriched regions of LSD endolysosomal membranes. This abnormal spatial organization locks SNAREs in complexes and impairs their sorting and recycling. Importantly, reducing membrane cholesterol levels in LSD cells restores normal SNARE function and efficient lysosomal fusion. Our results support a model by which cholesterol abnormalities determine lysosomal dysfunction and endocytic traffic jam in LSDs by impairing the membrane fusion machinery, thus suggesting new therapeutic targets for the treatment of these disorders.


Subject(s)
Cholesterol/metabolism , Lysosomal Storage Diseases/metabolism , Lysosomes/metabolism , Membrane Fusion/physiology , SNARE Proteins/metabolism , Animals , Autophagy , Blotting, Western , Cells, Cultured , Embryo, Mammalian/cytology , Embryo, Mammalian/metabolism , Endocytosis/physiology , ErbB Receptors/metabolism , Fibroblasts/cytology , Fibroblasts/metabolism , Fluorescent Antibody Technique , Immunoenzyme Techniques , Immunoprecipitation , Lysosomal Storage Diseases/pathology , Lysosomal Membrane Proteins/genetics , Lysosomal Membrane Proteins/metabolism , Mice , Phospholipids/metabolism , RNA, Messenger/genetics , Reverse Transcriptase Polymerase Chain Reaction
16.
Hum Mol Genet ; 17(17): 2610-21, 2008 Sep 01.
Article in English | MEDLINE | ID: mdl-18508857

ABSTRACT

Sulfatase modifying factor 1 (SUMF1) encodes for the formylglicine generating enzyme, which activates sulfatases by modifying a key cysteine residue within their catalytic domains. SUMF1 is mutated in patients affected by multiple sulfatase deficiency, a rare recessive disorder in which all sulfatase activities are impaired. Despite the absence of canonical retention/retrieval signals, SUMF1 is largely retained in the endoplasmic reticulum (ER), where it exerts its enzymatic activity on nascent sulfatases. Part of SUMF1 is secreted and paracrinally taken up by distant cells. Here we show that SUMF1 interacts with protein disulfide isomerase (PDI) and ERp44, two thioredoxin family members residing in the early secretory pathway, and with ERGIC-53, a lectin that shuttles between the ER and the Golgi. Functional assays reveal that these interactions are crucial for controlling SUMF1 traffic and function. PDI couples SUMF1 retention and activation in the ER. ERGIC-53 and ERp44 act downstream, favoring SUMF1 export from and retrieval to the ER, respectively. Silencing ERGIC-53 causes proteasomal degradation of SUMF1, while down-regulating ERp44 promotes its secretion. When over-expressed, each of three interactors favors intracellular accumulation. Our results reveal a multistep control of SUMF1 trafficking, with sequential interactions dynamically determining ER localization, activity and secretion.


Subject(s)
Mannose-Binding Lectins/metabolism , Membrane Proteins/metabolism , Molecular Chaperones/metabolism , Protein Disulfide-Isomerases/metabolism , Sulfatases/metabolism , HeLa Cells , Humans , Oxidoreductases Acting on Sulfur Group Donors , Polysaccharides/metabolism , Proteasome Endopeptidase Complex/metabolism , Protein Transport , Sulfatases/analysis
17.
Hum Mol Genet ; 17(1): 119-29, 2008 Jan 01.
Article in English | MEDLINE | ID: mdl-17913701

ABSTRACT

Most lysosomal storage disorders (LSDs) are caused by deficiencies of lysosomal hydrolases. While LSDs were among the first inherited diseases for which the underlying biochemical defects were identified, the mechanisms from enzyme deficiency to cell death are poorly understood. Here we show that lysosomal storage impairs autophagic delivery of bulk cytosolic contents to lysosomes. By studying the mouse models of two LSDs associated with severe neurodegeneration, multiple sulfatase deficiency (MSD) and mucopolysaccharidosis type IIIA (MPSIIIA), we observed an accumulation of autophagosomes resulting from defective autophagosome-lysosome fusion. An impairment of the autophagic pathway was demonstrated by the inefficient degradation of exogenous aggregate-prone proteins (i.e. expanded huntingtin and mutated alpha-synuclein) in cells from LSD mice. This impairment resulted in massive accumulation of polyubiquitinated proteins and of dysfunctional mitochondria which are the putative mediators of cell death. These data identify LSDs as 'autophagy disorders' and suggest the presence of common mechanisms in the pathogenesis of these and other neurodegenerative diseases.


Subject(s)
Autophagy/physiology , Lysosomal Storage Diseases/pathology , Animals , Autophagy/genetics , Base Sequence , Cells, Cultured , DNA Primers/genetics , Humans , Lysosomal Storage Diseases/genetics , Lysosomal Storage Diseases/physiopathology , Lysosomal Storage Diseases, Nervous System/genetics , Lysosomal Storage Diseases, Nervous System/pathology , Lysosomal Storage Diseases, Nervous System/physiopathology , Lysosomes/pathology , Membrane Fusion , Mice , Microtubule-Associated Proteins/genetics , Microtubule-Associated Proteins/metabolism , Mitochondria/pathology , Mucopolysaccharidosis III/genetics , Mucopolysaccharidosis III/pathology , Mucopolysaccharidosis III/physiopathology , Multiple Sulfatase Deficiency Disease/genetics , Multiple Sulfatase Deficiency Disease/pathology , Multiple Sulfatase Deficiency Disease/physiopathology , Nerve Degeneration/genetics , Nerve Degeneration/pathology , Nerve Degeneration/physiopathology , Phagosomes/pathology , Transfection , Ubiquitination
18.
Hum Mol Genet ; 16(14): 1699-707, 2007 Jul 15.
Article in English | MEDLINE | ID: mdl-17517693

ABSTRACT

PRPF3 is an element of the splicing machinery ubiquitously expressed, yet mutations in this gene are associated with a tissue-specific phenotype: autosomal dominant retinitis pigmentosa (RP). Here, we studied the subcellular localization of endogenous- and mutant-transfected PRPF3. We found that (i) subcellular distribution of the endogenous wild-type protein co-localizes with small nuclear ribonucleoproteins, partially with a nucleolar marker and accumulates in speckles labeled by SC35; (ii) in human retinas, PRPF3 does not show a distinctive abundance in photoreceptors, the cells affected in RP and (iii) the RP causing mutant PRPF3, differently from the wild-type protein, forms abnormally big aggregates in transfected photoreceptor cells. Aggregation of T494M mutant PRPF3 inside the nucleus triggers apoptosis only in photoreceptor cells. On the basis of the observation that mutant PRPF3 accumulates in the nucleolus and that transcriptional, translational and proteasome inhibition can induce this phenomenon in non-photoreceptor cells, we hypothesize that mutation affects splicing factor recycling. Noteworthy, accumulation of the mutant protein in big aggregates also affects distribution of some other splicing factors. Our data suggest that the mutant protein has a cell-specific dominant effect in rod photoreceptors while appears not to be harmful to epithelial and fibroblast cells.


Subject(s)
Mutation , Nuclear Proteins/genetics , Nuclear Proteins/physiology , Retinal Degeneration/genetics , Ribonucleoprotein, U4-U6 Small Nuclear/genetics , Ribonucleoprotein, U4-U6 Small Nuclear/physiology , Active Transport, Cell Nucleus , Alternative Splicing , Epithelial Cells/metabolism , Fibroblasts/metabolism , Gene Expression Profiling , Genes, Dominant , HeLa Cells , Humans , Phenotype , Proteasome Endopeptidase Complex/metabolism , Retina/metabolism , Retinal Rod Photoreceptor Cells/metabolism
19.
Proc Natl Acad Sci U S A ; 104(11): 4506-11, 2007 Mar 13.
Article in English | MEDLINE | ID: mdl-17360554

ABSTRACT

Sulfatases are involved in several biological functions such as degradation of macromolecules in the lysosomes. In patients with multiple sulfatase deficiency, mutations in the SUMF1 gene cause a reduction of sulfatase activities because of a posttranslational modification defect. We have generated a mouse line carrying a null mutation in the Sumf1 gene. Sulfatase activities are completely absent in Sumf1(-/-) mice, indicating that Sumf1 is indispensable for sulfatase activation and that mammals, differently from bacteria, have a single sulfatase modification system. Similarly to multiple sulfatase deficiency patients, Sumf1(-/-) mice display frequent early mortality, congenital growth retardation, skeletal abnormalities, and neurological defects. All examined tissues showed progressive cell vacuolization and significant lysosomal storage of glycosaminoglycans. Sumf1(-/-) mice showed a generalized inflammatory process characterized by a massive presence of highly vacuolated macrophages, which are the main site of lysosomal storage. Activated microglia were detected in the cerebellum and brain cortex associated with remarkable astroglyosis and neuronal cell loss. Between 4 and 6 months of age, we detected a strong increase in the expression levels of inflammatory cytokines and of apoptotic markers in both the CNS and liver, demonstrating that inflammation and apoptosis occur at the late stage of disease and suggesting that they play an important role in both the systemic and CNS phenotypes observed in lysosomal disorders. This mouse model, in which the function of an entire protein family has been silenced, offers a unique opportunity to study sulfatase function and the mechanisms underlying lysosomal storage diseases.


Subject(s)
Inflammation , Neurodegenerative Diseases/genetics , Sulfatases/deficiency , Sulfatases/genetics , Animals , Apoptosis , Cell Line , Disease Models, Animal , Fibroblasts/metabolism , Glycosaminoglycans/chemistry , Macrophages/metabolism , Mice , Mice, Transgenic , Mutation , Neurons/cytology , Oxidoreductases Acting on Sulfur Group Donors , Phenotype , Sulfatases/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...