Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
Add more filters










Publication year range
1.
Eur Thyroid J ; 13(3)2024 Jun 01.
Article in English | MEDLINE | ID: mdl-38838198

ABSTRACT

Objective: Many cases of subacute thyroiditis (SAT) have been described related to SARS-CoV-2 infection, but no prospective data about follow-up are known. This prospective, longitudinal, 3-year, multicentre study aims to explore the clinical peculiarities and outcome of SAT in relation to SARS-CoV-2 infection, ascertained with antibody dosage. Methods: All patients receiving SAT diagnosis from November 2020 to May 2022 were enrolled. Data on anamnesis, physical examination, blood tests (TSH, freeT4, freeT3, thyroglobulin, anti-thyroid antibodies, C-reactive protein, erythrocyte sedimentation rate, complete blood count), and thyroid ultrasound were collected. At baseline, the presence of IgG against the SARS-CoV-2 spike protein or nucleocapsid was investigated. Patients were evaluated after 1, 3, 6, and 12 months. Results: Sixty-six subjects were enrolled. At baseline, 54 presented with pain, 36 (67%) for at least 15 days. Serum SARS-CoV-2 IgG measurements documented that 7 out of 52 subjects (13.5%) had infection before SAT diagnosis (COVID+). No significant differences between the COVID+ and COVID- groups were found at baseline, except for respiratory symptoms and fever, which were more common in COVID+ (P = 0.039 and P = 0.021, respectively). Among the 41 subjects who completed follow-up, COVID+ and COVID- did not differ for therapeutic approach to SAT or outcome, all having an improvement in neck pain, inflammation parameters, and ultrasound features. Conclusion: This is the first prospective study investigating any difference both at diagnosis and at follow-up between SAT presentation in patients with previous SARS-CoV-2 infection and those without. Our data demonstrate that SARS-CoV-2 does not impact on SAT onset, evolution, and outcome.


Subject(s)
COVID-19 , SARS-CoV-2 , Thyroiditis, Subacute , Humans , Thyroiditis, Subacute/diagnosis , Thyroiditis, Subacute/blood , COVID-19/diagnosis , COVID-19/epidemiology , COVID-19/immunology , COVID-19/complications , Male , Female , Prospective Studies , Middle Aged , SARS-CoV-2/immunology , Adult , Immunoglobulin G/blood , Antibodies, Viral/blood , Aged , Longitudinal Studies , Spike Glycoprotein, Coronavirus/immunology
2.
Cell Calcium ; 121: 102896, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38749289

ABSTRACT

Phospholipid scramblases mediate the rapid movement of lipids between membrane leaflets, a key step in establishing and maintaining membrane homeostasis of the membranes of all eukaryotic cells and their organelles. Thus, impairment of lipid scrambling can lead to a variety of pathologies. How scramblases catalyzed the transbilayer movement of lipids remains poorly understood. Despite the availability of direct structural information on three unrelated families of scramblases, the TMEM16s, the Xkrs, and ATG-9, a unifying mechanism has failed to emerge thus far. Among these, the most extensively studied and best understood are the Ca2+ activated TMEM16s, which comprise ion channels and/or scramblases. Early work supported the view that these proteins provided a hydrophilic, membrane-exposed groove through which the lipid headgroups could permeate. However, structural, and functional experiments have since challenged this mechanism, leading to the proposal that the TMEM16s distort and thin the membrane near the groove to facilitate lipid scrambling. Here, we review our understanding of the structural and mechanistic underpinnings of lipid scrambling by the TMEM16s and discuss how the different proposals account for the various experimental observations.


Subject(s)
Anoctamins , Phospholipid Transfer Proteins , Humans , Anoctamins/metabolism , Anoctamins/chemistry , Animals , Phospholipid Transfer Proteins/metabolism , Phospholipid Transfer Proteins/chemistry
3.
FEBS J ; 289(9): 2578-2592, 2022 05.
Article in English | MEDLINE | ID: mdl-34775680

ABSTRACT

Membrane asymmetry is important for cellular physiology and established by energy-dependent unidirectional lipid translocases, which have diverse physiological functions in plants. By contrast, the role of phospholipid scrambling (PLS), the passive bidirectional lipid transfer leading to the break-down of membrane asymmetry, is currently still unexplored. The Arabidopsis thaliana genome contains a single gene (At1g73020) with homology to the eukaryotic TMEM16 family of Ca2+ -activated phospholipid scramblases. Here, we investigated the protein function of this Arabidopsis homolog. Fluorescent AtTMEM16 fusions localized to the ER both in transiently expressing Arabidopsis protoplasts and HEK293 cells. A putative scrambling domain (SCRD) was identified on the basis of sequence conservation and conferred PLS to transfected HEK293 cells, when grafted into the backbone of the non-scrambling plasma membrane-localized TMEM16A chloride channel. Finally, AtTMEM16 'gain-of-function' variants gave rise to cellular phenotypes typical of aberrant scramblase activity, which were reversed by the additional introduction of a 'loss-of-function' mutation into the SCRD. In conclusion, our data suggest AtTMEM16 works as an ER-resident lipid scramblase in Arabidopsis.


Subject(s)
Anoctamins , Arabidopsis , Anoctamins/genetics , Anoctamins/metabolism , Arabidopsis/genetics , Arabidopsis/metabolism , Calcium/metabolism , HEK293 Cells , Humans , Phospholipid Transfer Proteins/genetics , Phospholipid Transfer Proteins/metabolism , Phospholipids/metabolism
4.
Front Pharmacol ; 12: 723218, 2021.
Article in English | MEDLINE | ID: mdl-34950024

ABSTRACT

Alexander's disease (AxD) is a rare, usually relentlessly progressive disorder of astroglial cells in the central nervous system related to mutations in the gene encoding the type III intermediate filament protein, glial fibrillary acidic protein (GFAP). The pathophysiology of AxD is only partially understood. Available data indicate that an excessive GFAP gene expression may play a role. In particular, a "threshold hypothesis" has been reported, suggesting that mutant GFAP representing about 20% of the total cellular GFAP should be sufficient to cause disease. Thus, strategies based on reducing cellular mutant GFAP protein levels and/or activating biological processes involved in the correct protein folding could be effective in counteracting the toxic effect of misfolded GFAP. Considering that clomipramine (CLM), which has been selected by a wide small molecules screening as the greatest inhibitory potential drug against GFAP expression, is contraindicated because of its proconvulsant activity in the infantile form of AxD, which is also characterized by the occurrence of epileptic seizures, two powerful antiepileptic agents, carbamazepine (CBZ) and phenytoin (PHT), which share specific stereochemical features in common with CLM, were taken into consideration in a reliable in vitro model of AxD. In the present work, we document for the first time that CBZ and PHT have a definite inhibitory effect on pathological GFAP cellular expression and folding. Moreover, we confirm previous results of a similar beneficial effect of CLM. In addition, we have demonstrated that CBZ and CLM play a refolding effect on mutant GFAP proteins, likely ascribed at the induction of CRYAB expression, resulting in the decrease of mutant GFAP aggregates formation. As CBZ and PHT are currently approved for use in humans, their documented effects on pathological GFAP cellular expression and folding may indicate a potential therapeutic role as disease-modifying agents of these drugs in the clinical management of AxD, particularly in AxD patients with focal epilepsy with and without secondary generalization.

5.
J Bone Miner Res ; 36(3): 531-545, 2021 03.
Article in English | MEDLINE | ID: mdl-33125761

ABSTRACT

ClC-7 is a chloride-proton antiporter of the CLC protein family. In complex with its accessory protein Ostm-1, ClC-7 localizes to lysosomes and to the osteoclasts' ruffled border, where it plays a critical role in acidifying the resorption lacuna during bone resorption. Gene inactivation in mice causes severe osteopetrosis, neurodegeneration, and lysosomal storage disease. Mutations in the human CLCN7 gene are associated with diverse forms of osteopetrosis. The functional evaluation of ClC-7 variants might be informative with respect to their pathogenicity, but the cellular localization of the protein hampers this analysis. Here we investigated the functional effects of 13 CLCN7 mutations identified in 13 new patients with severe or mild osteopetrosis and a known ADO2 mutation. We mapped the mutated amino acid residues in the homology model of ClC-7 protein, assessed the lysosomal colocalization of ClC-7 mutants and Ostm1 through confocal microscopy, and performed patch-clamp recordings on plasma-membrane-targeted mutant ClC-7. Finally, we analyzed these results together with the patients' clinical features and suggested a correlation between the lack of ClC-7/Ostm1 in lysosomes and severe neurodegeneration. © 2020 American Society for Bone and Mineral Research (ASBMR).


Subject(s)
Bone Resorption , Osteopetrosis , Animals , Chloride Channels/genetics , Humans , Lysosomes , Mice , Mutation/genetics , Osteoclasts , Osteopetrosis/genetics
6.
Hum Mutat ; 41(6): 1157-1170, 2020 06.
Article in English | MEDLINE | ID: mdl-32112655

ABSTRACT

Mutations in the human TMEM16E/ANO5 gene are causative for gnathodiaphyseal dysplasia (GDD), a rare bone malformation and fragility disorder, and for two types of muscular dystrophy (MD). Previous studies have demonstrated that TMEM16E/ANO5 is a Ca2+ -activated phospholipid scramblase and that the mutation c.1538C>T (p.Thr513Ile) causing GDD leads to a gain-of-function phenotype. Here, using established HEK293-based functional assays, we investigated the effects of MD-related and further GDD-related amino acid exchanges on TMEM16E/ANO5 function in the same expression system. These experiments also revealed that the gradual changes in HEK293 cell morphology observed upon expression of TMEM16E/ANO5GDD mutants are a consequence of aberrant protein activity. Our results collectively demonstrate that, on the level of protein function, MD mutations are associated to loss-of-function and GDD mutations to gain-of-function phenotypes, confirming conjectures made on the basis of inheritance modes.


Subject(s)
Anoctamins/genetics , Muscular Dystrophies/genetics , Osteogenesis Imperfecta/genetics , Amino Acid Sequence , Bone Diseases, Developmental/genetics , Gain of Function Mutation , HEK293 Cells , Humans , Loss of Function Mutation , Phenotype , Phospholipids
8.
Cell Mol Life Sci ; 75(9): 1657-1670, 2018 05.
Article in English | MEDLINE | ID: mdl-29124309

ABSTRACT

Mutations in the human TMEM16E (ANO5) gene are associated both with the bone disease gnathodiaphyseal dysplasia (GDD; OMIM: 166260) and muscle dystrophies (OMIM: 611307, 613319). However, the physiological function of TMEM16E has remained unclear. We show here that human TMEM16E, when overexpressed in mammalian cell lines, displayed partial plasma membrane localization and gave rise to phospholipid scrambling (PLS) as well as non-selective ionic currents with slow time-dependent activation at highly depolarized membrane potentials. While the activity of wild-type TMEM16E depended on elevated cytosolic Ca2+ levels, a mutant form carrying the GDD-causing T513I substitution showed PLS and large time-dependent ion currents even at low cytosolic Ca2+ concentrations. Contrarily, mutation of the homologous position in the Ca2+-activated Cl- channel TMEM16B paralog hardly affected its function. In summary, these data provide the first direct demonstration of Ca2+-dependent PLS activity for TMEM16E and suggest a gain-of-function phenotype related to a GDD mutation.


Subject(s)
Anoctamins/genetics , Gain of Function Mutation , Osteogenesis Imperfecta/genetics , Phospholipids/metabolism , Animals , Anoctamins/metabolism , CHO Cells , Cricetinae , Cricetulus , Enzyme Activation/genetics , HEK293 Cells , Humans , Osteogenesis Imperfecta/metabolism , Phospholipid Transfer Proteins/genetics , Phospholipid Transfer Proteins/metabolism , Tumor Cells, Cultured
9.
Hum Mutat ; 39(2): 219-236, 2018 02.
Article in English | MEDLINE | ID: mdl-29098737

ABSTRACT

Heterozygous mutations in the PHOX2B gene are causative of congenital central hypoventilation syndrome (CCHS), a neurocristopathy characterized by defective autonomic control of breathing due to the impaired differentiation of neural crest cells. Among PHOX2B mutations, polyalanine (polyAla) expansions are almost exclusively associated with isolated CCHS, whereas frameshift variants, although less frequent, are often more severe than polyAla expansions and identified in syndromic CCHS. This article provides a complete review of all the frameshift mutations identified in cases of isolated and syndromic CCHS reported in the literature as well as those identified by us and not yet published. These were considered in terms of both their structure, whether the underlying indels induced frameshifts of either 1 or 2 steps ("frame 2" and "frame 3" mutations respectively), and clinical associations. Furthermore, we evaluated the structural and functional effects of one "frame 3" mutation identified in a patient with isolated CCHS, and one "frame 2" mutation identified in a patient with syndromic CCHS, also affected with Hirschsprung's disease and neuroblastoma. The data thus obtained confirm that the type of translational frame affects the severity of the transcriptional dysfunction and the predisposition to isolated or syndromic CCHS.


Subject(s)
Frameshift Mutation/genetics , Homeodomain Proteins/chemistry , Homeodomain Proteins/genetics , Hypoventilation/congenital , Sleep Apnea, Central/genetics , Transcription Factors/chemistry , Transcription Factors/genetics , Electrophoretic Mobility Shift Assay , HeLa Cells , Humans , Hypoventilation/genetics , Hypoventilation/metabolism , Microscopy, Fluorescence , Mutation , Sleep Apnea, Central/metabolism
10.
Oncotarget ; 8(42): 72133-72146, 2017 09 22.
Article in English | MEDLINE | ID: mdl-29069774

ABSTRACT

The pathogenic role of the PHOX2B gene in neuroblastoma is indicated by heterozygous mutations in neuroblastoma patients and by gene overexpression in both neuroblastoma cell lines and tumor samples. PHOX2B encodes a transcription factor which is crucial for the correct development and differentiation of sympathetic neurons. PHOX2B overexpression is considered a prognostic marker for neuroblastoma and it is also used by clinicians to monitor minimal residual disease. Furthermore, it has been observed that neuronal differentiation in neuroblastoma is dependent on down-regulation of PHOX2B expression, which confirms that PHOX2B expression may be considered a target in neuroblastoma. Here, PHOX2B promoter or 3' untranslated region were used as molecular targets in an in vitro high-throughput approach that led to the identification of molecules able to decrease PHOX2B expression at transcriptional and likely even at post-transcriptional levels. Further functional investigations carried out on PHOX2B mRNA levels and biological consequences, such as neuroblastoma cell apoptosis and growth, showed that chloroquine and mycophenolate mofetil are most promising agents for neuroblastoma therapy based on down-regulation of PHOX2B expression. Finally, a strong correlation between the effect of drugs in terms of down-regulation of PHOX2B expression and of biological consequences in neuroblastoma cells confirms the role of PHOX2B as a potential molecular target in neuroblastoma.

11.
EMBO Rep ; 18(7): 1100-1107, 2017 07.
Article in English | MEDLINE | ID: mdl-28536248

ABSTRACT

Phosphatidylinositol-3,5-bisphosphate (PI(3,5)P2) is a low-abundance signaling lipid associated with endo-lysosomal and vacuolar membranes in eukaryotic cells. Recent studies on Arabidopsis indicated a critical role of PI(3,5)P2 in vacuolar acidification and morphology during ABA-induced stomatal closure, but the molecular targets in plant cells remained unknown. By using patch-clamp recordings on Arabidopsis vacuoles, we show here that PI(3,5)P2 does not affect the activity of vacuolar H+-pyrophosphatase or vacuolar H+-ATPase. Instead, PI(3,5)P2 at low nanomolar concentrations inhibited an inwardly rectifying conductance, which appeared upon vacuolar acidification elicited by prolonged H+ pumping activity. We provide evidence that this novel conductance is mediated by chloride channel a (CLC-a), a member of the anion/H+ exchanger family formerly implicated in stomatal movements in Arabidopsis H+-dependent currents were absent in clc-a knock-out vacuoles, and canonical CLC-a-dependent nitrate/H+ antiport was inhibited by low concentrations of PI(3,5)P2 Finally, using the pH indicator probe BCECF, we show that CLC-a inhibition contributes to vacuolar acidification. These data provide a mechanistic explanation for the essential role of PI(3,5)P2 and advance our knowledge about the regulation of vacuolar ion transport.


Subject(s)
Arabidopsis/metabolism , Chloride Channels/metabolism , Phosphatidylinositol Phosphates/metabolism , Signal Transduction , Anions , Arabidopsis Proteins/metabolism , Biological Transport , Hydrogen-Ion Concentration , Ion Transport , Lysosomes/metabolism , Vacuolar Proton-Translocating ATPases/metabolism , Vacuoles/metabolism
12.
Biochim Biophys Acta Mol Basis Dis ; 1863(7): 1770-1777, 2017 07.
Article in English | MEDLINE | ID: mdl-28433712

ABSTRACT

HSCR is a congenital disorder of the enteric nervous system, characterized by the absence of neurons along a variable length of the gut resulting from loss-of-function RET mutations. Congenital Central Hypoventilation Syndrome (CCHS) is a rare neurocristopathy characterized by impaired response to hypercapnia and hypoxemia caused by heterozygous mutations of the PHOX2B gene, mostly polyalanine (polyA) expansions but also missense, nonsense, and frameshift mutations, while polyA contractions are common in the population and believed neutral. HSCR associated CCHS can present in patients carrying PHOX2B mutations. Indeed, RET expression is orchestrated by different transcriptional factors among which PHOX2B, thus suggesting its possible role in HSCR pathogenesis. Following the observation of HSCR patients carrying in frame trinucleotide deletions within the polyalanine stretch in exon 3 (polyA contractions), we have verified the hypothesis that these PHOX2B variants do reduce its transcriptional activity, likely resulting in a down-regulation of RET expression and, consequently, favouring the development of the HSCR phenotype. Using proper reporter constructs, we show here that the in vitro transactivation of the RET promoter by different HSCR-associated PHOX2B polyA variants has resulted significantly lower compared to the effect of PHOX2B wild type protein. In particular, polyA contractions do induce a reduced transactivation of the RET promoter, milder compared to the severe polyA expansions associated with CCHS+HSCR, and correlated with the length of the deleted trait, with a more pronounced effect when contractions are larger.


Subject(s)
Base Sequence , Gene Expression Regulation , Genetic Predisposition to Disease , Hirschsprung Disease/metabolism , Homeodomain Proteins/metabolism , Peptides/metabolism , Proto-Oncogene Proteins c-ret/biosynthesis , Sequence Deletion , Transcription Factors/metabolism , Transcription, Genetic , Cell Line, Tumor , Female , Hirschsprung Disease/genetics , Hirschsprung Disease/pathology , Homeodomain Proteins/genetics , Humans , Male , Peptides/genetics , Proto-Oncogene Proteins c-ret/genetics , Transcription Factors/genetics
13.
Biochim Biophys Acta ; 1849(8): 1057-65, 2015 Aug.
Article in English | MEDLINE | ID: mdl-26145533

ABSTRACT

Neuroblastoma (NB) is a rare childhood cancer of the peripheral sympathetic nervous system and accounts for approximately 10% of all pediatric tumors. Heterozygous PHOX2B mutations have been found in association with NB development in familial, sporadic and syndromic cases. In addition, the PHOX2B gene is widely over-expressed both in tumor samples and NB cell lines. Post-transcriptional gene regulation is known to be involved in mRNA stability and, in NB, microRNAs (miRNAs) seem to be responsible for altered expression of genes driving differentiation, apoptosis, and migration. To assess the possible impact of post-transcriptional regulation in NB cell lines, we have focused on the PHOX2B mRNA stability by both in silico analysis and functional studies on its 3'untranslated region (3'UTR). PHOX2B gene expression has resulted under post-transcriptional control, as suggested by: i) instability of PHOX2B mRNA, demonstrated by short mRNA half-life levels in both IMR32 and LAN-1 cell lines, ii) role of the PHOX2B-3'UTR, confirmed by the activity of proper reporter constructs, and iii) miRNA-204, shown to enhance the PHOX2B 3'UTR mediated down-regulation of the reporter construct activity. Finally, miRNA-204 has resulted to decrease the stability of the PHOX2B mRNA at different extents in the presence of different SNP rs1063611 alleles. Therefore, post-transcriptional down-regulation of the PHOX2B gene takes place in NB cell lines and miRNA-204 participates in such a 3'UTR mediated control.


Subject(s)
Brain Neoplasms/genetics , Gene Expression Regulation, Neoplastic , Homeodomain Proteins/genetics , MicroRNAs/physiology , Neuroblastoma/genetics , Transcription Factors/genetics , 3' Untranslated Regions , Base Sequence , Brain Neoplasms/pathology , Cells, Cultured , Child , Down-Regulation/genetics , Humans , Molecular Sequence Data , Neuroblastoma/pathology , RNA Processing, Post-Transcriptional/genetics
14.
Exp Cell Res ; 336(1): 43-57, 2015 Aug 01.
Article in English | MEDLINE | ID: mdl-25882494

ABSTRACT

PHOX2B is a transcription factor involved in the regulation of neurogenesis and in the correct differentiation of the autonomic nervous system. The pathogenetic role of PHOX2B in neuroblastoma (NB) is supported by mutations in familial, sporadic and syndromic cases of NB and overexpression of PHOX2B and its target ALK in tumor samples and NB cell lines. Starting from these observations, we have performed in vitro drug screening approaches targeting PHOX2B overexpression as a potential pharmacological means in NB. In particular, in order to identify molecules able to decrease PHOX2B expression, we have evaluated the effects of 70 compounds in IMR-32 cell line stably expressing the luciferase gene under the control of the PHOX2B promoter. Curcumin, SAHA and trichostatin A showed to down-regulate the PHOX2B promoter activity which resulted in a decrease of both protein and mRNA expressions. In addition, we have observed that curcumin acts by interfering with PBX-1/MEIS-1, NF-κB and AP-1 complexes, in this work demonstrated for the first time to regulate the transcription of the PHOX2B gene. Finally, combined drug treatments showed successful effects in down-regulating the expression of both PHOX2B and its target ALK genes, thus supporting the notion of the effectiveness of molecule combination in tumor therapy.


Subject(s)
Drug Evaluation, Preclinical , High-Throughput Screening Assays , Homeodomain Proteins/antagonists & inhibitors , Neuroblastoma/drug therapy , Neuroblastoma/metabolism , Signal Transduction/drug effects , Small Molecule Libraries/pharmacology , Transcription Factors/antagonists & inhibitors , Blotting, Western , Cell Differentiation , Curcumin/pharmacology , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Histone Deacetylase Inhibitors/pharmacology , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Humans , Hydroxamic Acids/pharmacology , In Vitro Techniques , Myeloid Ecotropic Viral Integration Site 1 Protein , NF-kappa B/genetics , NF-kappa B/metabolism , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Neuroblastoma/pathology , Pre-B-Cell Leukemia Transcription Factor 1 , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Transcription Factor AP-1/genetics , Transcription Factor AP-1/metabolism , Transcription Factors/genetics , Tumor Cells, Cultured
15.
Ann Rheum Dis ; 72(6): 1044-52, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23117241

ABSTRACT

OBJECTIVES: Tumour necrosis factor (TNF) receptor-associated periodic syndrome (TRAPS) is caused by TNFRSF1A mutations, known to induce intracellular retention of the TNFα receptor 1 (TNFR1) protein, defective TNFα-induced apoptosis, and production of reactive oxygen species. As downregulation of autophagy, the main cellular pathway involved in insoluble aggregate elimination, has been observed to increase the inflammatory response, we investigated whether it plays a role in TRAPS pathogenesis. METHODS: The possible link between TNFRSF1A mutations and inflammation in TRAPS was studied in HEK-293T cells, transfected with expression constructs for wild-type and mutant TNFR1 proteins, and in monocytes derived from patients with TRAPS, by investigating autophagy function, NF-κB activation and interleukin (IL)-1ß secretion. RESULTS: We found that autophagy is responsible for clearance of wild-type TNFR1, but when TNFR1 is mutated, the autophagy process is defective, probably accounting for mutant TNFR1 accumulation as well as TRAPS-associated induction of NF-κB activity and excessive IL-1ß secretion, leading to chronic inflammation. Autophagy inhibition due to TNFR1 mutant proteins can be reversed, as demonstrated by the effects of the antibiotic geldanamycin, which was found to rescue the membrane localisation of mutant TNFR1 proteins, reduce their accumulation and counteract the increased inflammation by decreasing IL-1ß secretion. CONCLUSIONS: Autophagy appears to be an important mechanism in the pathogenesis of TRAPS, an observation that provides a rationale for the most effective therapy in this autoinflammatory disorder. Our findings also suggest that autophagy could be proposed as a novel therapeutic target for TRAPS and possibly other similar diseases.


Subject(s)
Autophagy/genetics , Hereditary Autoinflammatory Diseases/genetics , Receptors, Tumor Necrosis Factor, Type I/genetics , Adolescent , Adult , Autophagy/physiology , Case-Control Studies , Child , Child, Preschool , HEK293 Cells , Humans , Interleukin-1beta/metabolism , Middle Aged , NF-kappa B/physiology , Receptors, Tumor Necrosis Factor, Type I/physiology , Syndrome
16.
Exp Cell Res ; 318(15): 1844-54, 2012 Sep 10.
Article in English | MEDLINE | ID: mdl-22705585

ABSTRACT

Heterozygous mutations of the GFAP gene are responsible for Alexander disease, a neurodegenerative disorder characterized by intracytoplasmic Rosenthal fibers (RFs) in dystrophic astrocytes. In vivo and in vitro models have shown co-localization of mutant GFAP proteins with the small heat shock proteins (sHSPs) HSP27 and alphaB-crystallin, ubiquitin and proteasome components. Results reported by several recent studies agree on ascribing an altered cytoskeletal pattern to mutant GFAP proteins, an effect which induces mutant proteins accumulation, leading to impaired proteasome function and autophagy induction. On the basis of the protective role shown by both these small heat shock proteins (sHSPs), and on the already well established neuroprotective effects of curcumin in several diseases, we have investigated the effects of this compound in an in vitro model of Alexander disease, consisting in U251-MG astrocytoma cells transiently transfected with a construct encoding for GFAP carrying the p.R239C mutation in frame with the reporter green fluorescent protein (GFP). In particular, depending on the dose used, we have observed that curcumin is able to induce both HSP27 and alphaB-crystallin, to reduce expression of both RNA and protein of endogenous GFAP, to induce autophagy and, finally, to rescue the filamentous organization of the GFAP mutant protein, thus suggesting a role of this spice in counteracting the pathogenic effects of GFAP mutations.


Subject(s)
Alexander Disease/drug therapy , Alexander Disease/metabolism , Curcumin/pharmacology , Glial Fibrillary Acidic Protein/genetics , Glial Fibrillary Acidic Protein/metabolism , Alexander Disease/genetics , Base Sequence , Cell Line , DNA Primers/genetics , Down-Regulation/drug effects , Glial Fibrillary Acidic Protein/chemistry , HSP27 Heat-Shock Proteins/genetics , HSP27 Heat-Shock Proteins/metabolism , Heat-Shock Proteins , Humans , Molecular Chaperones , Mutant Proteins/chemistry , Mutant Proteins/genetics , Mutant Proteins/metabolism , Protein Multimerization/drug effects , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Up-Regulation/drug effects , alpha-Crystallin B Chain/genetics , alpha-Crystallin B Chain/metabolism
17.
J Mol Med (Berl) ; 90(9): 1025-35, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22307522

ABSTRACT

Expansions of a polyalanine (polyA) stretch in the coding region of the PHOX2B gene cause congenital central hypoventilation syndrome (CCHS), a neurocristopathy characterized by the absence of adequate control of autonomic breathing. Expansion of polyA in PHOX2B leads to protein misfolding and accumulation into inclusions. The mechanisms that regulate mutant protein degradation and turnover have been poorly elucidated. Here, we investigate the regulation of degradation of wild-type and polyA-expanded PHOX2B. We show that expanded PHOX2B is targeted for degradation through the ubiquitin-proteasome system, resulting in lowered levels of the mutant protein relative to its wild-type counterpart. Moreover, we show that mutant PHOX2B forms ubiquitin-positive inclusions, which sequester wild-type PHOX2B. This sequestration correlates with reduced transcriptional activity of endogenous wild-type protein in neuroblastoma cells. Finally, we show that the E3 ubiquitin ligase TRIM11 plays a critical role in the clearance of mutant PHOX2B through the proteasome. Importantly, clearance of mutant PHOX2B by TRIM11 correlates with a rescue of PHOX2B transcriptional activity. We propose that CCHS is partially caused by a dominant-negative effect of expanded PHOX2B due to the retention of the wild-type protein in pathogenic aggregates. Our results demonstrate that TRIM11 is a novel modifier of mutant PHOX2B toxicity and represents a potential therapeutic target for CCHS.


Subject(s)
Homeodomain Proteins/metabolism , Hypoventilation/congenital , Peptides/metabolism , Sleep Apnea, Central/metabolism , Transcription Factors/metabolism , Ubiquitin-Protein Ligases/metabolism , Cell Line, Tumor , HeLa Cells , Homeodomain Proteins/analysis , Homeodomain Proteins/genetics , Humans , Hypoventilation/genetics , Hypoventilation/metabolism , Mutant Proteins/analysis , Mutant Proteins/genetics , Mutant Proteins/metabolism , Mutation , Peptides/genetics , Proteasome Endopeptidase Complex/metabolism , Sleep Apnea, Central/genetics , Transcription Factors/analysis , Transcription Factors/genetics , Transcriptional Activation , Tripartite Motif Proteins , Ubiquitin/metabolism , Ubiquitin-Protein Ligases/analysis
18.
Neurobiol Dis ; 45(1): 508-18, 2012 Jan.
Article in English | MEDLINE | ID: mdl-21964250

ABSTRACT

Heterozygous in frame duplications of the PHOX2B gene, leading to polyalanine (polyAla) expansions ranging from +5 to +13 residues of a 20-alanine stretch, have been identified in the vast majority of patients affected with Congenital Central Hypoventilation Syndrome (CCHS), a rare neurocristopathy characterized by absence of adequate autonomic control of respiration with decreased sensitivity to hypoxia and hypercapnia. Ventilatory supports such as tracheostomy, nasal mask or diaphragm pacing represent the only options available for affected. We have already shown that the severity of the CCHS phenotype correlates with the length of polyAla expansions, ultimately leading to formation of toxic intracytoplasmic aggregates and impaired PHOX2B mediated transactivation of target gene promoters, such as DBH. At present, there is no specific treatment to reduce cell aggregates and to ameliorate patients' respiration. In this work, we have undertaken in vitro analyses aimed at assessing the effects of molecules on the cellular response to polyAla PHOX2B aggregates. In particular, we tested 17-AAG, ibuprofen, 4-PBA, curcumin, trehalose, congo red and chrysamine G for their ability to i) recover the nuclear localisation of polyAla expanded PHOX2B, ii) rescue of PHOX2B mediated transactivation of the DBH promoter, and iii) clearance of PHOX2B (+13 Ala) aggregates. Our data have suggested that 17-AAG and curcumin are effective in vitro in both rescuing the nuclear localization and transactivation activity of PHOX2B carrying the largest expansion of polyAla and promoting the clearance of aggregates of these mutant proteins inducing molecular mechanisms such as ubiquitin-proteasome (UPS), autophagy and heat shock protein (HSP) systems.


Subject(s)
Apoptosis/drug effects , Homeodomain Proteins/genetics , Hypoventilation/congenital , Peptides/genetics , Sleep Apnea, Central/genetics , Transcription Factors/genetics , Animals , Benzoates/pharmacology , Benzoquinones/pharmacology , Biphenyl Compounds/pharmacology , COS Cells , Cells, Cultured , Chlorocebus aethiops , Congo Red/pharmacology , Curcumin/pharmacology , HeLa Cells , Homeodomain Proteins/metabolism , Humans , Hypoventilation/genetics , Hypoventilation/metabolism , Ibuprofen/pharmacology , Lactams, Macrocyclic/pharmacology , Peptides/metabolism , Promoter Regions, Genetic/drug effects , Proteasome Endopeptidase Complex/drug effects , Proteasome Endopeptidase Complex/genetics , Sleep Apnea, Central/metabolism , Transcription Factors/metabolism , Trehalose/pharmacology
19.
Eur J Paediatr Neurol ; 15(5): 449-52, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21388845

ABSTRACT

Molecular pathogenesis of congenital disorders associated with polyalanine expansions has been investigated for several years. Despite different pathological hallmarks characterize each polyalanine disease, they share common features, mainly represented by aggregates containing the mutant proteins, usually mislocated inside the cellular compartments, along with ubiquitin and proteasome components. Recently, particular interest has been raised by investigations on molecules able to restore both correct localization and function of the expanded proteins. Here we report a list of drugs whose effects have been assayed both in in vitro and in vivo models of polyalanine disorders, such as the oculopharyingeal muscular dystrophy, congenital central hypoventilation syndrome, synpolydactyly and in cell and animal models carrying specific artificial mutations. In particular, we have reviewed, for each polyalanine mutant protein, the molecules tested, cellular models under investigation, drugs effects on aggregation and underlying mechanisms.


Subject(s)
DNA Repeat Expansion/genetics , Drug Evaluation, Preclinical/methods , Genetic Diseases, Inborn/drug therapy , Genetic Diseases, Inborn/genetics , Mutant Proteins/genetics , Peptides/genetics , Animals , COS Cells , Cell Culture Techniques/methods , Chlorocebus aethiops , Disease Models, Animal , Genetic Diseases, Inborn/pathology , HeLa Cells , Humans , Mutant Proteins/antagonists & inhibitors
20.
Ann Hum Genet ; 74(6): 506-15, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20946255

ABSTRACT

The Glial Fibrillary Acidic Protein (GFAP) gene encodes a cytoskeletal protein belonging to the intermediate filament family whose expression is considered as a marker of astrocytes differentiation. GFAP expression, shown to be upregulated as a consequence of brain gliosis, depends on hormones, growth factors, cytokine, and transcription factors and, among these latters, activator protein 1 (AP-1) has been demonstrated to play a crucial role. In this study, we have focused on a 2.2 kb sequence of the regulatory region located upstream of the GFAP gene, searching in a panel of control individuals for single-nucleotide polymorphisms (SNPs) that could modulate GFAP transcription. Among four SNPs of the GFAP promoter whose alleles have been predicted by in silico analysis to induce differences in the pattern of binding transcription factors, we have identified a new AP-1 binding site lying at -250 bp upstream from the GFAP transcriptional start site. The two alleles of this polymorphic locus have shown to bind the AP-1 complex to different extents, thus promoting variable transcriptional activities of the GFAP promoter. Therefore, these SNP alleles may, among others, mediate the effects of GFAP mutations, thus explaining the phenotypic heterogeneity of Alexander disease.


Subject(s)
Glial Fibrillary Acidic Protein/genetics , Promoter Regions, Genetic , Transcription Factor AP-1/metabolism , Transcriptional Activation , Alleles , Astrocytes/metabolism , Binding Sites , Cell Line , Gene Frequency , Glial Fibrillary Acidic Protein/metabolism , Humans , Italy , Transcription, Genetic
SELECTION OF CITATIONS
SEARCH DETAIL
...