Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters











Publication year range
1.
J Allergy Clin Immunol ; 145(1): 368-378.e13, 2020 01.
Article in English | MEDLINE | ID: mdl-31194989

ABSTRACT

BACKGROUND: Cryopyrin-associated periodic syndromes (CAPS) are a group of autoinflammatory diseases linked to gain-of-function mutations in the NOD-like receptor family, pyrin domain containing 3 (NLRP3) gene, which cause uncontrolled IL-1ß secretion. Proton pump inhibitors (PPIs), which are commonly used as inhibitors of gastric acid production, also have anti-inflammatory properties, protect mice from sepsis, and prevent IL-1ß secretion by monocytes from patients with CAPS. OBJECTIVE: We sought to develop a novel Nlrp3 knock-in (KI) mouse model of CAPS to study amyloidosis, a severe CAPS complication, and test novel therapeutic approaches. METHODS: We generated KI mice by engineering the N475K mutation, which is associated with the CAPS phenotype, into the mouse Nlrp3 gene. KI and wild-type mice received PPIs or PBS intraperitoneally and were analyzed for survival, inflammation, cytokine secretion, and amyloidosis development. RESULTS: Mutant Nlrp3 KI mice displayed features that recapitulate the immunologic and clinical phenotype of CAPS. They showed systemic inflammation with high levels of serum proinflammatory cytokines, inflammatory infiltrates in various organs, and amyloid deposits in the spleen, liver, and kidneys. Toll-like receptor stimulated macrophages from KI mice secreted high levels of IL-1ß, IL-18, and IL-1α but low amounts of IL-1 receptor antagonist. Treatment of KI mice with PPIs had a clear clinical effect, showing a reduction in inflammatory manifestations, regression of amyloid deposits, and normalization of proinflammatory and anti-inflammatory cytokine production by macrophages. CONCLUSION: Nlrp3 KI mice displayed a CAPS phenotype with many characteristics of autoinflammation, including amyloidosis. The therapeutic effectiveness of PPIs associated with a lack of toxicity indicates that these drugs could represent relevant adjuvants to the anti-IL-1 drugs in patients with CAPS and other IL-1-driven diseases.


Subject(s)
Amyloidosis , Cryopyrin-Associated Periodic Syndromes , NLR Family, Pyrin Domain-Containing 3 Protein , Proton Pump Inhibitors/pharmacology , Amyloidosis/drug therapy , Amyloidosis/genetics , Amyloidosis/immunology , Animals , Cryopyrin-Associated Periodic Syndromes/drug therapy , Cryopyrin-Associated Periodic Syndromes/genetics , Cryopyrin-Associated Periodic Syndromes/immunology , Cryopyrin-Associated Periodic Syndromes/pathology , Disease Models, Animal , Gene Knock-In Techniques , Humans , Interleukin-1beta/genetics , Interleukin-1beta/immunology , Mice , Mice, Mutant Strains , NLR Family, Pyrin Domain-Containing 3 Protein/genetics , NLR Family, Pyrin Domain-Containing 3 Protein/immunology
2.
J Rheumatol ; 44(11): 1667-1673, 2017 Nov.
Article in English | MEDLINE | ID: mdl-28916543

ABSTRACT

OBJECTIVE: To evaluate the rate of somatic NLRP3 mosaicism in an Italian cohort of mutation-negative patients with cryopyrin-associated periodic syndrome (CAPS). METHODS: The study enrolled 14 patients with a clinical phenotype consistent with CAPS in whom Sanger sequencing of the NLRP3 gene yielded negative results. Patients' DNA were subjected to amplicon-based NLRP3 deep sequencing. RESULTS: Low-level somatic NLRP3 mosaicism has been detected in 4 patients, 3 affected with chronic infantile neurological cutaneous and articular syndrome and 1 with Muckle-Wells syndrome. Identified nucleotide substitutions encode for 4 different amino acid exchanges, with 2 of them being novel (p.Y563C and p.G564S). In vitro functional studies confirmed the deleterious behavior of the 4 somatic NLRP3 mutations. Among the different neurological manifestations detected, 1 patient displayed mild loss of white matter volume on brain magnetic resonance imaging. CONCLUSION: The allele frequency of somatic NLRP3 mutations occurs generally under 15%, considered the threshold of detectability using the Sanger method of DNA sequencing. Consequently, routine genetic diagnostic of CAPS should be currently performed by next-generation techniques ensuring high coverage to identify also low-level mosaicism, whose actual frequency is yet unknown and probably underestimated.


Subject(s)
Brain/diagnostic imaging , Cryopyrin-Associated Periodic Syndromes/genetics , Mosaicism , NLR Family, Pyrin Domain-Containing 3 Protein/genetics , Child , Child, Preschool , Cryopyrin-Associated Periodic Syndromes/diagnostic imaging , Female , High-Throughput Nucleotide Sequencing , Humans , Infant , Infant, Newborn , Italy , Magnetic Resonance Imaging , Male , White Matter/diagnostic imaging
3.
Clin Exp Rheumatol ; 34(6 Suppl 102): S121-S128, 2016.
Article in English | MEDLINE | ID: mdl-27310036

ABSTRACT

OBJECTIVES: Tumour necrosis factor (TNF) receptor-associated periodic syndrome (TRAPS) is a multisystemic autoinflammatory condition associated with heterozygous TNFRSF1A mutations, presenting with a variety of clinical symptoms, many of which yet unexplained. In this work, we aimed at deepening into TRAPS pathogenic mechanisms sustained by monocytes. METHODS: Microarray experiments were conducted to identify genes whose expression results altered in patients compared to healthy individuals, both under basal condition and following LPS stimulation. RESULTS: An inflammatory state baseline, characterised by constitutive overexpression of IL1ß and IL1R1 receptor, has been shown in TRAPS patients compared to controls, including in non-active disease phases. Following LPS stimulation, IL1RN up-regulation is stronger in controls than in patients and inflammatory pathways and microRNAs undergo differential regulation. Genes involved in post-translational modifications, protein folding and ubiquitination result constitutively up-regulated in TRAPS, while response to interferon types I and II is defective, failing to be up-regulated by LPS. TGFß pathway is down-regulated in untreated TRAPS monocytes, while genes involved in redox regulation result constitutively over-expressed. Finally, additional molecular alterations seem to reflect organ failures sometime complicating the disease. CONCLUSIONS: Gene expression profile in resting TRAPS monocytes has confirmed the patients' chronic inflammatory condition. In addition, pathways not yet associated with the disease have been disclosed, such as interferon types I and II response to LPS stimulation and a downregulation of the TGFß pathway in basal condition. The role of miRNA, suggested by our results, deserves in-depth analyses in light of the possible development of targeted therapies.


Subject(s)
Fever/genetics , Gene Expression Regulation , Hereditary Autoinflammatory Diseases/genetics , Inflammation Mediators/metabolism , Monocytes/metabolism , Adolescent , Case-Control Studies , Child , Child, Preschool , Female , Fever/diagnosis , Fever/immunology , Fever/metabolism , Gene Expression Profiling/methods , Gene Expression Regulation/drug effects , Genetic Association Studies , Genetic Markers , Genetic Predisposition to Disease , Hereditary Autoinflammatory Diseases/diagnosis , Hereditary Autoinflammatory Diseases/immunology , Hereditary Autoinflammatory Diseases/metabolism , Heterozygote , Humans , Inflammation Mediators/immunology , Interleukin 1 Receptor Antagonist Protein/genetics , Interleukin 1 Receptor Antagonist Protein/metabolism , Lipopolysaccharides/pharmacology , Male , Monocytes/drug effects , Monocytes/immunology , Mutation , Oligonucleotide Array Sequence Analysis , Phenotype , Polymerase Chain Reaction , Receptors, Tumor Necrosis Factor, Type I/genetics , Receptors, Tumor Necrosis Factor, Type I/immunology , Reproducibility of Results
4.
J Rheumatol ; 38(7): 1378-84, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21459945

ABSTRACT

OBJECTIVE: Tumor necrosis factor (TNF) receptor-associated periodic syndrome (TRAPS) is an autosomal-dominant multisystemic autoinflammatory condition. Patients display different mutations of the TNF receptor superfamily 1A gene (TNFRSF1A), coding for a nearly ubiquitous TNF receptor (TNFR1). No TNFRSF1A mutation has been identified in a proportion of patients with TRAPS-like phenotype. METHODS: We investigated mechanisms downregulating the TNF-induced inflammatory response such as (1) receptor shedding, producing a secreted form acting as a TNF inhibitor; (2) receptor internalization with subsequent induction of apoptosis; and (3) negative regulation of nuclear factor-κB (NF-κB) transcription. We analyzed the sequence of genes known to play a pivotal role in these pathways, in 5 patients with TRAPS symptoms and showing shedding and/or apoptosis defects, but without mutations of the TNFRSF1A gene. RESULTS: Sequence analysis of 3 genes involved in TNFR1 shedding (ERAP1, NUCB2, RBMX) and 3 genes involved in negative regulation of NF-κB signaling (TNFAIP3, CARP-2) or NF-κB transcription (ZFP36) revealed only a few unreported variants, apparently neutral. CONCLUSION: Our study rules out any involvement in the pathogenesis of TRAPS of some of the genes known to regulate TNFR1 shedding and TNF-induced NF-κB signaling and transcription. Gene(s) responsible for TRAPS-like syndrome remain to be investigated among currently unidentified genes likely involved in these pathways, or by applying the genome-wide function-free sequencing approach.


Subject(s)
Familial Mediterranean Fever/genetics , Familial Mediterranean Fever/physiopathology , Hereditary Autoinflammatory Diseases/genetics , Mutation/genetics , Receptors, Tumor Necrosis Factor, Type I/genetics , Tumor Necrosis Factor-alpha/physiology , Adult , Aminopeptidases/genetics , Apoptosis/physiology , Calcium-Binding Proteins/genetics , Case-Control Studies , DNA-Binding Proteins/genetics , Female , Heterogeneous-Nuclear Ribonucleoproteins/genetics , Humans , Male , Minor Histocompatibility Antigens , NF-kappa B/physiology , Nerve Tissue Proteins/genetics , Nucleobindins , Phenotype , Signal Transduction/physiology
5.
PLoS One ; 5(10)2010 Oct 01.
Article in English | MEDLINE | ID: mdl-20957039

ABSTRACT

BACKGROUND: Neuroblastoma (NB) is a severe pediatric tumor originating from neural crest derivatives and accounting for 15% of childhood cancer mortality. The heterogeneous and complex genetic etiology has been confirmed with the identification of mutations in two genes, encoding for the receptor tyrosine kinase Anaplastic Lymphoma Kinase (ALK) and the transcription factor Paired-like Homeobox 2B (PHOX2B), in a limited proportion of NB patients. Interestingly, these two genes are overexpressed in the great majority of primary NB samples and cell lines. These observations led us to test the hypothesis of a regulatory or functional relationship between ALK and PHOX2B underlying NB pathogenesis. METHODOLOGY/PRINCIPAL FINDINGS: Following this possibility, we first confirmed a striking correlation between the transcription levels of ALK, PHOX2B and its direct target PHOX2A in a panel of NB cell lines. Then, we manipulated their expression in NB cell lines by siRNA-mediated knock-down and forced over-expression of each gene under analysis. Surprisingly, PHOX2B- and PHOX2A-directed siRNAs efficiently downregulated each other as well as ALK gene and, consistently, the enhanced expression of PHOX2B in NB cells yielded an increment of ALK protein. We finally demonstrated that PHOX2B drives ALK gene transcription by directly binding its promoter, which therefore represents a novel PHOX2B target. CONCLUSIONS/SIGNIFICANCE: These findings provide a compelling explanation of the concurrent involvement of these two genes in NB pathogenesis and are going to foster a better understanding of molecular interactions at the base of the disease. Moreover, this work opens new perspectives for NBs refractory to conventional therapies that may benefit from the design of novel therapeutic RNAi-based approaches for multiple gene targets.


Subject(s)
Gene Expression Regulation, Neoplastic/physiology , Homeodomain Proteins/physiology , Neuroblastoma/genetics , Protein-Tyrosine Kinases/genetics , Transcription Factors/physiology , Anaplastic Lymphoma Kinase , Cell Line, Tumor , Gene Silencing , Homeodomain Proteins/genetics , Humans , In Vitro Techniques , Neuroblastoma/pathology , RNA, Small Interfering , Receptor Protein-Tyrosine Kinases , Regulatory Sequences, Nucleic Acid , Transcription Factors/genetics , Transcriptional Activation
6.
BMB Rep ; 42(12): 788-93, 2009 Dec 31.
Article in English | MEDLINE | ID: mdl-20044949

ABSTRACT

TLX2 is an orphan homeodomain transcription factor whose expression is mainly associated with tissues derived from neural crest cells. Recently, we have demonstrated that PHOX2A and PHOX2B are able to enhance the neural cell-type specific expression of human TLX2 by binding distally the 5'-flanking region. In the present work, to deepen into the TLX2 transcription regulation, we have focused on the proximal 5'- flanking region of the gene, mapping the transcription start site and identifying a minimal promoter necessary and sufficient for the basal transcription in cell lines from different origin. Site-directed mutagenesis has allowed to demonstrate that the integrity of this sequence is crucial for gene expression, while electrophoretic mobility shift assays and chromatin immunoprecipitation experiments have revealed that such an activity is dependent on the binding of a PBX factor. Consistent with these findings, such a basal promoter activity has resulted to be enhanced by the previously reported PHOX2-responding sequence.


Subject(s)
Gene Expression Regulation/genetics , Homeodomain Proteins/genetics , Base Sequence , Binding Sites , Cloning, Molecular , Humans , Neural Crest/cytology , Neural Crest/metabolism , Promoter Regions, Genetic/genetics , Transcription, Genetic/genetics , Transcriptional Activation/genetics , Tumor Cells, Cultured
7.
Eur J Hum Genet ; 15(8): 848-55, 2007 Aug.
Article in English | MEDLINE | ID: mdl-17505528

ABSTRACT

TLX2 (also known as HOX11L1, Ncx and Enx) is a transcription factor playing a crucial role in the development of the enteric nervous system, as confirmed by mice models exhibiting intestinal hyperganglionosis and pseudo-obstruction. However, congenital defects of TLX2 have been excluded as a major cause of intestinal motility disorders in patients affected with intestinal neuronal dysplasia (IND) or pseudo-obstruction. After demonstrating the direct regulation of TLX2 expression by the homeoprotein PHOX2B, in the present work, we have focused on its paralogue PHOX2A. By co-transfections, electrophoretic mobility shift assays and chromatin immunoprecipitation, we have demonstrated that PHOX2A, like PHOX2B, is involved in the cascade leading to TLX2 transactivation and presumably in the intestinal neuronal differentiation. Based on the hypothesis that missed activation of the TLX2 gene induces the development of enteric nervous system defects, PHOX2A and PHOX2B have been regarded as novel candidate genes involved in IND and pseudo-obstruction and consequently analyzed for mutations in a specific set of 26 patients. We have identified one still unreported PHOX2A variant; however, absence of any functional effect on TLX2 transactivation suggests that regulators or effectors other than the PHOX2 genes must act in the same pathway, likely playing a non redundant and direct role in the pathogenesis of such enteric disorders.


Subject(s)
Gene Expression Regulation/physiology , Homeodomain Proteins/genetics , Homeodomain Proteins/physiology , Intestines/innervation , Transcription Factors/physiology , Animals , Cell Differentiation/genetics , Cell Differentiation/physiology , Cell Line , Cell Line, Tumor , Homeodomain Proteins/biosynthesis , Humans , Mice , Mice, Knockout , Neurons/cytology , Neurons/metabolism , Transcriptional Activation
8.
Int J Biochem Cell Biol ; 39(2): 327-39, 2007.
Article in English | MEDLINE | ID: mdl-17045833

ABSTRACT

Polyalanine expansions in the PHOX2B gene have been detected in the vast majority of patients affected with congenital central hypoventilation syndrome, a neurocristopathy characterized by absence of adequate control of breathing, especially during sleep, with decreased sensitivity to hypoxia and hypercapnia. The correlation between length of the alanine expanded tracts and severity of congenital central hypoventilation syndrome respiratory phenotype has been confirmed by length-dependent cytoplasmic PHOX2B retention with formation of aggregates. To deepen into the molecular mechanisms mediating the effects of PHOX2B polyalanine expansions, we have set up experiments aimed at assessing the fate of cells characterized by PHOX2B polyalanine aggregates. In particular, we have observed that activation of the heat shock response by the drug geldanamycin is efficient both in preventing formation and in inducing clearance of PHOX2B pre-formed polyalanine aggregates in COS-7 cells expressing PHOX2B-GFP fused proteins, and ultimately also in rescuing the PHOX2B ability to transactivate the Dopamine-beta-Hydroxilase promoter. In addition, we have demonstrated elimination of PHOX2B mutant proteins by the proteasome and autophagy, two cellular mechanisms already been involved in the clearance of proteins containing expanded polyglutamine and polyalanine tracts. Moreover, our data suggest that geldanamycin effects on PHOX2B aggregates may be also mediated by the proteasome pathway. Finally, analysis of cellular toxicity due to polyalanine aggregates has confirmed the occurrence of cell apoptosis consequent to expression of PHOX2B carrying the longest expanded alanine tract and shown that geldanamycin can delay cell progression toward the most advanced apoptotic stages.


Subject(s)
Benzoquinones/pharmacology , Cell Nucleus/metabolism , Homeodomain Proteins/chemistry , Homeodomain Proteins/metabolism , Lactams, Macrocyclic/pharmacology , Peptides/chemistry , Transcription Factors/chemistry , Transcription Factors/metabolism , Active Transport, Cell Nucleus/drug effects , Animals , Autophagy , Benzoquinones/toxicity , COS Cells , Chlorocebus aethiops , Dopamine beta-Hydroxylase/genetics , Dose-Response Relationship, Drug , HeLa Cells , Humans , Lactams, Macrocyclic/toxicity , Peptides/genetics , Proteasome Endopeptidase Complex/metabolism , Protein Folding , Protein Transport/drug effects , Transcriptional Activation/drug effects , Transfection , Trinucleotide Repeat Expansion , Ubiquitin/metabolism
9.
Mol Cancer Res ; 4(9): 635-43, 2006 Sep.
Article in English | MEDLINE | ID: mdl-16966433

ABSTRACT

Based on a knocked-out mouse model and a few expression studies, TLX3 is regarded as a homeobox gene crucial for the development of the autonomic nervous system. This gene can undergo rearrangements or deregulation, giving rise to T-cell acute lymphocytic leukemia. The present report is focused on the identification of elements and factors playing a role in the TLX3 physiologic expression regulation and therefore likely to be involved in cancer development. In particular, after identifying the transcription start points, we have made use of in vitro transfection assays to show that the 5'-untranslated region of the gene is necessary for the basal promoter activity in cell lines from different origin. By site-directed mutagenesis, two tandem CCAAT boxes have been localized as critical elements of this region. In vivo chromatin immunoprecipitation and electrophoretic mobility shift assays have indicated that nuclear factor Y (NFY) recognizes these sites in all the analyzed cell lines. The physiologic role of such an interaction has been confirmed by a dominant-negative version of the NFY transcription factor that has turned out to decrease both in vitro TLX3 promoter activity and endogenous amount of mRNA. Finally, a consistent in vivo TLX3 expression impairment was also achieved after NFY mRNA knockdown. The full characterization of the TLX3 transcription regulation will ultimately provide crucial elements to define the involvement of this gene in T-cell acute lymphocytic leukemia development.


Subject(s)
CCAAT-Binding Factor/genetics , Homeodomain Proteins/genetics , Leukemia, T-Cell/genetics , Oncogene Proteins/genetics , Transcription Factors/genetics , Animals , Base Sequence , Cell Line, Tumor , Homeodomain Proteins/biosynthesis , Humans , Leukemia, T-Cell/metabolism , Mice , Molecular Sequence Data , Oncogene Proteins/biosynthesis , Promoter Regions, Genetic , Transcription Initiation Site , Transcription, Genetic
10.
Biochem J ; 395(2): 355-61, 2006 Apr 15.
Article in English | MEDLINE | ID: mdl-16402914

ABSTRACT

The TLX2 (HOX11L1, Ncx, Enx) and PHOX2B genes encode transcription factors crucial in the development of neural-crest-derived cells, leading to ANS (autonomic nervous system) specific neuronal lineages. Moreover, they share a similar expression pattern and are both involved in downstream steps of BMP (bone morphogenetic protein) signalling. In an attempt to reconstruct the gene network sustaining the correct development of the ANS, we have undertaken an in vitro experimental strategy to identify direct upstream regulators of the TLX2 gene. After characterizing a sequence displaying enhancer property in its 5' flanking region, we confirmed the functional link between the human PHOX2B and TLX2 genes. Transient transfections and electrophoretic-mobility-shift assays suggested that PHOX2B is able to bind the cell-specific element in the 5' regulatory region of the TLX2 gene, determining its transactivation in neuroblastoma cells. Such interaction was also confirmed in vivo by means of chromatin immunoprecipitation assay and, in addition, up-regulation of endogenous TLX2 mRNA level was demonstrated following PHOX2B over-expression, by quantitative real-time PCR. Finally, PHOX2B proteins carrying mutations responsible for CCHS (congenital central hypoventilation syndrome) development showed a severe impairment in activating TLX2 expression, both in vitro and in vivo. Taken together, these results support the PHOX2B-TLX2 promoter interaction, suggesting a physiological role in the transcription-factor cascade underlying the differentiation of neuronal lineages of the ANS during human embryogenesis.


Subject(s)
Genes, Homeobox/genetics , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Neural Crest/cytology , Neural Crest/metabolism , Transcription Factors/metabolism , Transcription, Genetic/genetics , 5' Flanking Region/genetics , Animals , Base Sequence , Humans , Hypoventilation/congenital , Hypoventilation/genetics , Mice , Molecular Sequence Data , Mutation/genetics , Sequence Deletion/genetics , Transcriptional Activation/genetics
11.
Gene Expr ; 12(3): 137-49, 2005.
Article in English | MEDLINE | ID: mdl-16127999

ABSTRACT

Neural crest cells arise from the epithelium of the dorsal neural tube and migrate to various districts giving origin, among others, to sympathetic, parasympathetic, and enteric ganglia. It has been shown that the transcription factors HOX11L1, HOX11L2, MASH1, PHOX2A, and PHOX2B are all necessary, to various extents, to the correct development of the autonomic nervous system. To investigate their possible role in the transcriptional regulation of the RET proto-oncogene, a gene playing a crucial role in correct intestinal innervation, we undertook a specific in vitro experimental strategy. Two neuroblastoma cell lines (SK-N-MC and SK-N-BE) were cotransfected with each transcription factor expressing plasmids and sequential deletion constructs of the 5' c-RET flanking region cloned upstream of the Luciferase reporter gene. Here we show that HOX11L1 enhances the activity of the c-RET promoter in SK-N-MC cell line by stimulating a region between -166 bp and -35 bp. Gel shift assays performed with oligonucleotides spanning this promoter sequence showed a change of the SP1 interaction with its binding sites, consequent to transfection with HOX11L1. While HOX11L2 showed no effect in both the cell lines, we have observed PHOX2A, PHOX2B, and MASH1 triggering a reproducible increase in the Luciferase activity in SK-N-BE cell line. A sequence responsible of the PHOX2A-dependent activation has been identified, while PHOX2B seems to act indirectly, as no physical binding has been demonstrated on c-RET promoter.


Subject(s)
Gene Expression Regulation , Neural Crest/metabolism , Proto-Oncogene Proteins c-ret/biosynthesis , Animals , Base Sequence , Basic Helix-Loop-Helix Transcription Factors/biosynthesis , Blotting, Western , Butyrates/pharmacology , COS Cells , Cell Line , Cell Line, Tumor , Cell Nucleus/metabolism , Cells, Cultured , Chlorocebus aethiops , DNA Primers/chemistry , DNA, Complementary/metabolism , Gene Deletion , Genes, Reporter , Genetic Vectors , Homeodomain Proteins/biosynthesis , Humans , In Vitro Techniques , Isobutyrates , Luciferases/metabolism , Molecular Sequence Data , Mutation , Oligonucleotides/chemistry , Oncogene Proteins/biosynthesis , Plasmids/metabolism , Promoter Regions, Genetic , Protein Binding , Proto-Oncogene Mas , Proto-Oncogene Proteins c-ret/genetics , Transcription Factors/biosynthesis , Transcription, Genetic , Transfection
12.
Hum Mol Genet ; 14(13): 1815-24, 2005 Jul 01.
Article in English | MEDLINE | ID: mdl-15888479

ABSTRACT

Congenital central hypoventilation syndrome (CCHS) is a rare neurocristopathy characterized by absence of adequate autonomic control of respiration with decreased sensitivity to hypoxia and hypercapnia. Frameshift mutations and polyalanine triplet expansions in the coding region of PHOX2B have been identified in the vast majority of CCHS patients and a correlation between length of the expanded region and severity of CCHS has been reported. In this work, we have undertaken in vitro analyses aimed at identifying the pathogenetic mechanisms which underlie the effects of PHOX2B mutations in CCHS. According to the known role of this gene, a transcription factor expressed during autonomic nervous system development, we have tested the transcriptional activity of WT and mutant PHOX2B expression constructs on the regulatory regions of two target genes, DbetaH and PHOX2A. We observed that the two sets of mutations play different roles in the transcriptional regulation of these genes, showing a correlation between the length of polyalanine expansions and the severity of reduced transcriptional activity. In particular, although reduced transactivation due to polyalanine expansions may be caused by retention of the mutated protein in the cytoplasm or in the nuclear aggregates, frameshift mutations did not impair the PHOX2B nuclear income, suggesting a different mechanism through which they would exert the observed effects on target promoters. Moreover, the frameshift due to deletion of a cytosine residue seems to cause sequestration of the corresponding mutant PHOX2B in the nucleolar compartment.


Subject(s)
Frameshift Mutation , Homeodomain Proteins/genetics , Hypoventilation/genetics , Peptides/genetics , Transcription Factors/genetics , Animals , Autonomic Nervous System/embryology , COS Cells , Chlorocebus aethiops , Frameshift Mutation/genetics , Gene Expression Regulation/genetics , Homeodomain Proteins/metabolism , Humans , Hypoventilation/pathology , Hypoventilation/physiopathology , Syndrome
13.
Int J Mol Med ; 10(1): 101-6, 2002 Jul.
Article in English | MEDLINE | ID: mdl-12060859

ABSTRACT

Intestinal Neuronal Dysplasia (IND) is a congenital disorder characterized by intestinal motility defects associated with hyperplasia of enteric ganglia. A phenotype resembling human IND has been observed in mice knocked-out for a member of the Hox11 homeobox gene family, Hox11l1, suggesting that the human homologue of this gene could be responsible for congenital disorders of intestinal innervation. However, previous mutation analysis of the coding sequence of the HOX11L1 gene in patients affected with IND detected neither mutations nor other nucleotide variants. In the present work, a detailed study of the non coding promoter region of this gene was undertaken in patients affected with IND, with Hirschsprung associated IND and with neurogenic chronic intestinal pseudo-obstruction. No alterations potentially impairing expression of HOX11L1, such as nucleotide variants, small deletions or cytogenetic alterations, could be identified thus further excluding the direct involvement of this gene in the pathogenesis of human intestinal motility disorders.


Subject(s)
Enteric Nervous System , Gastrointestinal Motility , Homeodomain Proteins/genetics , Oncogene Proteins/genetics , Promoter Regions, Genetic , Autonomic Nervous System Diseases/genetics , Autonomic Nervous System Diseases/physiopathology , Enteric Nervous System/abnormalities , Hirschsprung Disease/genetics , Hirschsprung Disease/physiopathology , Homeodomain Proteins/metabolism , Humans , Intestinal Diseases/genetics , Intestinal Diseases/physiopathology , Intestinal Pseudo-Obstruction/genetics , Intestinal Pseudo-Obstruction/physiopathology , Oncogene Proteins/metabolism , Proto-Oncogene Proteins , Sequence Analysis, DNA
14.
Eur J Hum Genet ; 10(3): 183-7, 2002 Mar.
Article in English | MEDLINE | ID: mdl-11973622

ABSTRACT

Hirschsprung disease (HSCR) is a complex disorder characterised by aganglia of distal gastrointestinal tracts. The highest proportion of both familial and sporadic cases is due to mutations of the RET proto-oncogene. Five germline mutations in the glial cell-line-derived neurotrophic factor (GDNF) gene, one of the RET ligands, have been detected in HSCR patients. Pedigrees analysis and the observed association between these GDNF alterations and RET variants in the same patients raised the question of whether the GDNF gene plays any causative/predisposing role in HSCR pathogenesis. In the present work, we have studied the ability of GDNF proteins, each bearing one of the reported mutations, to activate RET by performing a functional test in cultured neuroblastoma cells. Consistently with the lack of genotype/phenotype correlation in human subjects, our results indicate absence of detectable alterations of mutant GDNF induced RET activation.


Subject(s)
Drosophila Proteins , Hirschsprung Disease/genetics , Mutation , Nerve Growth Factors , Nerve Tissue Proteins/genetics , Proto-Oncogene Proteins/genetics , Receptor Protein-Tyrosine Kinases/genetics , Animals , COS Cells , Genotype , Glial Cell Line-Derived Neurotrophic Factor , Glial Cell Line-Derived Neurotrophic Factor Receptors , Humans , Ligands , Mutagenesis, Site-Directed , Phenotype , Phosphorylation , Proto-Oncogene Mas , Proto-Oncogene Proteins c-ret , Transfection , Tumor Cells, Cultured
15.
Mutagenesis ; 17(1): 25-30, 2002 Jan.
Article in English | MEDLINE | ID: mdl-11752230

ABSTRACT

Deregulation of control of the apoptotic process in Fanconi anaemia (FA) appears to be one of the main features of this disease at the cellular level. We show here that FA cells are resistant to treatments with rhodamine-1,2,3 and doxycycline, which both interfere with mitochondrial functionality by different mechanisms. In contrast, normal lymphoblastoid cells are severely affected by these treatments, which result in acute ATP depletion and a significant enhancement of the fraction of cells undergoing apoptotic cell death. FA cells are very sensitive to the action of 2-deoxy-D-glucose (2dG) and iodoacetic acid (IAA), two inhibitors of glycolytic metabolism. The ability of FA cells to sustain metabolic insults interfering with energy production and balance may be linked with the pathological manifestations of the disease, including susceptibility to acute myeloid leukemia. These findings suggest that FA genes may be involved in a pathway that mediates a protective response to stress. We suggest that a peculiar metabolic regulation in FA cells could explain both defective apoptosis and susceptibility to oxidative stress.


Subject(s)
Apoptosis/physiology , Cell Cycle Proteins , DNA-Binding Proteins , Energy Metabolism , Fanconi Anemia/pathology , Nuclear Proteins , Adenosine Triphosphate/metabolism , Cell Line, Transformed/drug effects , Cell Line, Transformed/metabolism , Deoxyglucose/pharmacology , Doxycycline/pharmacology , Electron Transport/drug effects , Fanconi Anemia/classification , Fanconi Anemia/genetics , Fanconi Anemia/metabolism , Fanconi Anemia Complementation Group A Protein , Fanconi Anemia Complementation Group Proteins , Genetic Predisposition to Disease , Glycolysis/drug effects , Herpesvirus 4, Human , Humans , Intracellular Membranes/drug effects , Iodoacetic Acid/pharmacology , Leukemia, Myeloid/genetics , Mitochondria/drug effects , Oxidative Stress , Proteins/genetics , Proteins/physiology , Rhodamine 123/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL