Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 40
Filter
1.
J Invest Dermatol ; 143(12): 2468-2475.e6, 2023 12.
Article in English | MEDLINE | ID: mdl-37414245

ABSTRACT

Generalized pustular psoriasis is a potentially life-threatening skin disease, associated with IL36RN disease alleles. IL36RN encodes the IL-36 receptor antagonist (IL-36Ra), a protein that downregulates the activity of IL-36 cytokines by blocking their receptor (IL-36R). Although generalized pustular psoriasis can be treated with IL-36R inhibitors, the structural underpinnings of the IL-36Ra/IL-36R interaction remain poorly understood. In this study, we sought to address this question by systematically investigating the effects of IL36RN sequence changes. We experimentally characterized the effects of 30 IL36RN variants on protein stability. In parallel, we used a machinelearning tool (Rhapsody) to analyze the IL-36Ra three-dimensional structure and predict the impact of all possible amino acid substitutions. This integrated approach identified 21 amino acids that are essential for IL-36Ra stability. We next investigated the effects of IL36RN changes on IL-36Ra/IL-36R binding and IL-36R signaling. Combining invitro assays and machine learning with a second program (mCSM), we identified 13 amino acids that are critical for IL-36Ra/IL36R engagement. Finally, we experimentally validated three representative predictions, further confirming the reliability of Rhapsody and mCSM. These findings shed light on the structural determinants of IL-36Ra activity, with potential to facilitate the design of new IL-36 inhibitors and aid the interpretation of IL36RN variants in diagnostic settings.


Subject(s)
Exanthema , Psoriasis , Skin Diseases, Vesiculobullous , Humans , Amino Acid Substitution , Amino Acids , Interleukins/metabolism , Psoriasis/genetics , Reproducibility of Results
3.
Commun Biol ; 4(1): 544, 2021 05 10.
Article in English | MEDLINE | ID: mdl-33972689

ABSTRACT

Actin-Related Protein-Testis1 (ARP-T1)/ACTRT1 gene mutations cause the Bazex-Dupré-Christol Syndrome (BDCS) characterized by follicular atrophoderma, hypotrichosis, and basal cell cancer. Here, we report an ARP-T1 interactome (PXD016557) that includes proteins involved in ciliogenesis, endosomal recycling, and septin ring formation. In agreement, ARP-T1 localizes to the midbody during cytokinesis and the basal body of primary cilia in interphase. Tissue samples from ARP-T1-associated BDCS patients have reduced ciliary length. The severity of the shortened cilia significantly correlates with the ARP-T1 levels, which was further validated by ACTRT1 knockdown in culture cells. Thus, we propose that ARP-T1 participates in the regulation of cilia length and that ARP-T1-associated BDCS is a case of skin cancer with ciliopathy characteristics.


Subject(s)
Carcinoma, Basal Cell/pathology , Cilia/pathology , Ciliopathies/pathology , Hypotrichosis/pathology , Keratinocytes/pathology , Microfilament Proteins/metabolism , Neoplasms, Basal Cell/pathology , Skin Neoplasms/pathology , Carcinoma, Basal Cell/genetics , Carcinoma, Basal Cell/metabolism , Cilia/metabolism , Ciliopathies/genetics , Ciliopathies/metabolism , Humans , Hypotrichosis/genetics , Hypotrichosis/metabolism , Keratinocytes/metabolism , Microfilament Proteins/genetics , Mutation , Neoplasms, Basal Cell/genetics , Neoplasms, Basal Cell/metabolism , Skin Neoplasms/genetics , Skin Neoplasms/metabolism
6.
Cytogenet Genome Res ; 157(4): 189-196, 2019.
Article in English | MEDLINE | ID: mdl-30974434

ABSTRACT

Hypohidrotic or anhidrotic ectodermal dysplasia (HED/EDA) is characterized by impaired development of the hair, teeth, or sweat glands. HED/EDA is inherited in an X-linked, autosomal dominant, or autosomal recessive pattern and caused by the pathogenic variants in 4 genes: EDA, EDAR, EDARADD, and WNT10A. The aim of the present study was to perform molecular screening of these 4 genes in a cohort of Turkish individuals diagnosed with HED/EDA. We screened for pathogenic variants of WNT10A, EDA, EDAR, and EDARADD through Sanger sequencing. We further assessed the clinical profiles of the affected individuals in order to establish phenotype-genotype correlation. In 17 (63%) out of 27 families, 17 pathogenic variants, 8 being novel, were detected in the 4 well-known ectodermal dysplasia genes. EDAR and EDA variants were identified in 6 families each, WNT10A variants in 4, and an EDARADD variant in 1, accounting for 35.3, 35.3, 23.5, and 5.9% of mutation-positive families, respectively. The low mutation detection rate of the cohort and the number of the EDAR pathogenic variants being as high as the EDA ones were the most noteworthy findings which could be attributed to the high consanguinity rate.


Subject(s)
Ectodermal Dysplasia/genetics , Ectodysplasins/genetics , Edar Receptor/genetics , Edar-Associated Death Domain Protein/genetics , Mutation , Sequence Analysis, DNA/methods , Wnt Proteins/genetics , Consanguinity , Female , Genetic Association Studies , Genetic Predisposition to Disease , Genotype , Humans , Male , Pedigree , Phenotype , Turkey
7.
Exp Dermatol ; 28(10): 1114-1117, 2019 10.
Article in English | MEDLINE | ID: mdl-28603914

ABSTRACT

The identification of loss-of-function mutations of the IL36RN gene encoding the interleukin-36 receptor antagonist (IL-36Ra) in generalized pustular psoriasis (GPP) emphasized the key role of this pathway in skin innate immunity and systemic inflammation. It has been previously shown in vitro that removal of the N-terminal amino acid IL36Ra (M1) is critical to its biological activity, but the in vivo contribution of this processing remains unknown. We report herein a new homozygous (c4G>T, pV2F) missense IL36RN mutation segregating in a family with three GPP-affected patients. The V2F mutation does not alter IL-36Ra protein expression but was devoid of any antagonist activity. Mass spectrometry showed that the V2F IL-36Ra mutant retains its first N-terminal methionine. These results provide the first in vivo demonstration that removal of N-terminal methionine of native IL-36Ra is a mandatory step to reach optimal antagonist activity and to prevent sustained skin and systemic inflammation in humans.


Subject(s)
Interleukins/deficiency , Loss of Function Mutation , Mutation, Missense , Point Mutation , Skin Diseases, Vesiculobullous/genetics , Amino Acid Substitution , C-Reactive Protein/analysis , Child , Consanguinity , Female , HEK293 Cells , Humans , Infant , Interleukins/genetics , Interleukins/physiology , Male , Pedigree , Phenotype , Skin Diseases, Vesiculobullous/pathology , Syndrome
9.
Nat Med ; 23(10): 1226-1233, 2017 Oct.
Article in English | MEDLINE | ID: mdl-28869610

ABSTRACT

Basal cell carcinoma (BCC), the most common human cancer, results from aberrant activation of the Hedgehog signaling pathway. Although most cases of BCC are sporadic, some forms are inherited, such as Bazex-Dupré-Christol syndrome (BDCS)-a cancer-prone genodermatosis with an X-linked, dominant inheritance pattern. We have identified mutations in the ACTRT1 gene, which encodes actin-related protein T1 (ARP-T1), in two of the six families with BDCS that were examined in this study. High-throughput sequencing in the four remaining families identified germline mutations in noncoding sequences surrounding ACTRT1. These mutations were located in transcribed sequences encoding enhancer RNAs (eRNAs) and were shown to impair enhancer activity and ACTRT1 expression. ARP-T1 was found to directly bind to the GLI1 promoter, thus inhibiting GLI1 expression, and loss of ARP-T1 led to activation of the Hedgehog pathway in individuals with BDCS. Moreover, exogenous expression of ACTRT1 reduced the in vitro and in vivo proliferation rates of cell lines with aberrant activation of the Hedgehog signaling pathway. In summary, our study identifies a disease mechanism in BCC involving mutations in regulatory noncoding elements and uncovers the tumor-suppressor properties of ACTRT1.


Subject(s)
Carcinoma, Basal Cell/genetics , Hypotrichosis/genetics , Microfilament Proteins/genetics , Skin Neoplasms/genetics , Animals , CRISPR-Cas Systems , Chromatin Immunoprecipitation , Enhancer Elements, Genetic/genetics , Female , Gene Expression Profiling , Hedgehog Proteins/metabolism , High-Throughput Nucleotide Sequencing , Humans , Male , Mice , Mice, Nude , Mutation , Neoplasm Transplantation , Polymerase Chain Reaction , Sequence Analysis, DNA , Signal Transduction
10.
Mol Genet Metab ; 122(3): 140-144, 2017 11.
Article in English | MEDLINE | ID: mdl-28711407

ABSTRACT

Incontinentia Pigmenti (IP) is a skin disorder with neurological impairment in 30% of cases. The most common disease causing mutation is a deletion of exons 4-10 of the IKBKG gene, located on chromosome Xq28, with skewed X-chromosome inactivation in females, but few cases of random X-inactivation have been reported. We have correlated brain anomalies with X-chromosome inactivation status determined on leucocytes circulating DNA. We reviewed MRI of 18 girls with genetically proven IP. We found three patterns of MRI, normal MRI (n=5), mild white matter abnormalities with cortical and corpus callosum atrophy (n=6), and severe cortical abnormalities suggesting a vascular disease (n=7). Most patients with severe abnormalities had random X-inactivation (6/7,86%), while 80% (4/5) of patients with normal MRI and 100% (6/6) of patients with mild white matter abnormalities had skewed inactivation. These results suggest that skewed chromosome X-inactivation may protect brain from damage, while in case of random inactivation, expression of the mutated IKBKG gene may lead to severe brain lesions.


Subject(s)
Brain/physiopathology , Incontinentia Pigmenti/diagnosis , Incontinentia Pigmenti/genetics , Leukocytes , Magnetic Resonance Imaging/statistics & numerical data , Neuroimaging/statistics & numerical data , Adolescent , Adult , Aged , Brain/diagnostic imaging , Child , Child, Preschool , DNA/blood , Female , Humans , I-kappa B Kinase/genetics , Incontinentia Pigmenti/complications , Incontinentia Pigmenti/physiopathology , Infant , Infant, Newborn , Mutation , Neuroimaging/methods , Sequence Deletion , X Chromosome Inactivation , Young Adult
11.
J Allergy Clin Immunol ; 140(6): 1671-1682.e2, 2017 Dec.
Article in English | MEDLINE | ID: mdl-28249776

ABSTRACT

BACKGROUND: Incontinentia pigmenti (IP; MIM308300) is a severe, male-lethal, X-linked, dominant genodermatosis resulting from loss-of-function mutations in the IKBKG gene encoding nuclear factor κB (NF-κB) essential modulator (NEMO; the regulatory subunit of the IκB kinase [IKK] complex). In 80% of cases of IP, the deletion of exons 4 to 10 leads to the absence of NEMO and total inhibition of NF-κB signaling. Here we describe a new IKBKG mutation responsible for IP resulting in an inactive truncated form of NEMO. OBJECTIVES: We sought to identify the mechanism or mechanisms by which the truncated NEMO protein inhibits the NF-κB signaling pathway. METHODS: We sequenced the IKBKG gene in patients with IP and performed complementation and transactivation assays in NEMO-deficient cells. We also used immunoprecipitation assays, immunoblotting, and an in situ proximity ligation assay to characterize the truncated NEMO protein interactions with IKK-α, IKK-ß, TNF receptor-associated factor 6, TNF receptor-associated factor 2, receptor-interacting protein 1, Hemo-oxidized iron regulatory protein 2 ligase 1 (HOIL-1), HOIL-1-interacting protein, and SHANK-associated RH domain-interacting protein. Lastly, we assessed NEMO linear ubiquitination using immunoblotting and investigated the formation of NEMO-containing structures (using immunostaining and confocal microscopy) after cell stimulation with IL-1ß. RESULTS: We identified a novel splice mutation in IKBKG (c.518+2T>G, resulting in an in-frame deletion: p.DelQ134_R256). The mutant NEMO lacked part of the CC1 coiled-coil and HLX2 helical domain. The p.DelQ134_R256 mutation caused inhibition of NF-κB signaling, although the truncated NEMO protein interacted with proteins involved in activation of NF-κB signaling. The IL-1ß-induced formation of NEMO-containing structures was impaired in fibroblasts from patients with IP carrying the truncated NEMO form (as also observed in HOIL-1-/- cells). The truncated NEMO interaction with SHANK-associated RH domain-interacting protein was impaired in a male fetus with IP, leading to defective linear ubiquitination. CONCLUSION: We identified a hitherto unreported disease mechanism (defective linear ubiquitination) in patients with IP.


Subject(s)
Fibroblasts/physiology , I-kappa B Kinase/metabolism , Incontinentia Pigmenti/metabolism , Skin/pathology , Ubiquitins/metabolism , Cloning, Molecular , Female , HEK293 Cells , Humans , I-kappa B Kinase/genetics , Incontinentia Pigmenti/genetics , Male , Mutation/genetics , NF-kappa B/metabolism , Pedigree , Protein Binding , Signal Transduction , Transcriptional Activation , Ubiquitination
12.
Nat Commun ; 7: 12629, 2016 09 02.
Article in English | MEDLINE | ID: mdl-27586688

ABSTRACT

The NF-κB pathway has critical roles in cancer, immunity and inflammatory responses. Understanding the mechanism(s) by which mutations in genes involved in the pathway cause disease has provided valuable insight into its regulation, yet many aspects remain unexplained. Several lines of evidence have led to the hypothesis that the regulatory/sensor protein NEMO acts as a biological binary switch. This hypothesis depends on the formation of a higher-order structure, which has yet to be identified using traditional molecular techniques. Here we use super-resolution microscopy to reveal the existence of higher-order NEMO lattice structures dependent on the presence of polyubiquitin chains before NF-κB activation. Such structures may permit proximity-based trans-autophosphorylation, leading to cooperative activation of the signalling cascade. We further show that NF-κB activation results in modification of these structures. Finally, we demonstrate that these structures are abrogated in cells derived from incontinentia pigmenti patients.


Subject(s)
I-kappa B Kinase/ultrastructure , Incontinentia Pigmenti/pathology , Microscopy/methods , NF-kappa B/metabolism , Cell Line, Tumor , Enzyme Activation , Humans , I-kappa B Kinase/metabolism , I-kappa B Kinase/physiology , Protein Binding , Protein Structure, Secondary , Ubiquitin/metabolism
13.
J Invest Dermatol ; 136(9): 1811-1819, 2016 09.
Article in English | MEDLINE | ID: mdl-27220475

ABSTRACT

Homozygous or compound heterozygous IL36RN gene mutations underlie the pathogenesis of psoriasis-related pustular eruptions including generalized pustular psoriasis, palmoplantar pustular psoriasis, acrodermatitis continua of Hallopeau, and acute generalized exanthematous pustular eruption. We identified two unreported IL36RN homozygous mutations (c.41C>A/p.Ser14X and c.420_426del/p.Gly141MetfsX29) in patients with familial generalized pustular psoriasis. We analyzed the impact of a spectrum of IL36RN mutations on IL-36 receptor antagonist protein by using site-directed mutagenesis and expression in HEK293T cells. This enabled us to differentiate null mutations with complete absence of IL-36 receptor antagonist (the two previously unreported mutations, c.80T>C/p.Leu27Pro, c.28C>T/p.Arg10X, c.280G>T/p.Glu94X, c.368C>G/p.Thr123Arg, c.368C>T/p.Thr123Met, and c.227C>T/p.Pro76Leu) from mutations with decreased (c.95A>G/p.His32Arg, c.142C>T/p.Arg48Trp, and c.308C>T/p.Ser113Leu) or unchanged (c.304C>T/p.Arg102Trp and c.104A>G/p.Lys35Arg) protein expression. Functional assays measuring the impact of mutations on the capacity to repress IL-36-dependent activation of the NF-κB pathway showed complete functional impairment for null mutations, whereas partial or no impairment was observed for other mutations considered as hypomorphic. Finally, null mutations were associated with severe clinical phenotypes (generalized pustular psoriasis, acute generalized exanthematous pustular eruption), whereas hypomorphic mutations were identified in both localized (palmoplantar pustular psoriasis, acrodermatitis continua of Hallopeau) and generalized variants. These results provide a preliminary basis for genotype-phenotype correlation in patients with deficiency of the IL-36Ra (DITRA), and suggest the involvement of other factors in the modulation of clinical expression.


Subject(s)
Interleukins/genetics , Mutation/genetics , Psoriasis/genetics , Adult , Child , Child, Preschool , Disease Progression , Female , Gene Expression Regulation/genetics , Genetic Association Studies , Genotype , Humans , Male , Phenotype , Prognosis , Psoriasis/physiopathology , Risk Assessment
14.
Am J Med Genet A ; 170A(1): 249-53, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26440664

ABSTRACT

Hypohidrotic ectodermal dysplasia (HED) is a rare disorder characterized by deficient development of structures derived from the ectoderm including hair, nails, eccrine glands, and teeth. HED forms that are caused by mutations in the genes EDA, EDAR, or EDARADD may show almost identical phenotypes, explained by a common signaling pathway. Proper interaction of the proteins encoded by these three genes is important for the activation of the NF-κB signaling pathway and subsequent transcription of the target genes. Mutations in the gene EDARADD are most rarely implicated in HED. Here we describe a novel missense mutation, c.367G>A (p.Asp123Asn), in this gene which did not appear to influence the interaction between EDAR and EDARADD proteins, but led to an impaired ability to activate NF-κB signaling. Female members of the affected family showed either unilateral or bilateral amazia. In addition, an affected girl developed bilateral ovarian teratomas, possibly associated with her genetic condition.


Subject(s)
Ectodermal Dysplasia 1, Anhidrotic/genetics , Edar Receptor/genetics , Edar-Associated Death Domain Protein/genetics , Mutation, Missense/genetics , Ovarian Neoplasms/genetics , Teratoma/genetics , Adolescent , Breast Diseases/genetics , Edar Receptor/metabolism , Edar-Associated Death Domain Protein/metabolism , Female , Hair/growth & development , Humans , Male , NF-kappa B/metabolism , Ovarian Neoplasms/pathology , Pedigree , Signal Transduction/genetics , Teratoma/pathology
15.
Am J Hum Genet ; 94(5): 790-7, 2014 May 01.
Article in English | MEDLINE | ID: mdl-24791904

ABSTRACT

Adaptor protein complex 1 (AP-1) is an evolutionary conserved heterotetramer that promotes vesicular trafficking between the trans-Golgi network and the endosomes. The knockout of most murine AP-1 complex subunits is embryonically lethal, so the identification of human disease-associated alleles has the unique potential to deliver insights into gene function. Here, we report two founder mutations (c.11T>G [p.Phe4Cys] and c.97C>T [p.Arg33Trp]) in AP1S3, the gene encoding AP-1 complex subunit σ1C, in 15 unrelated individuals with a severe autoinflammatory skin disorder known as pustular psoriasis. Because the variants are predicted to destabilize the 3D structure of the AP-1 complex, we generated AP1S3-knockdown cell lines to investigate the consequences of AP-1 deficiency in skin keratinocytes. We found that AP1S3 silencing disrupted the endosomal translocation of the innate pattern-recognition receptor TLR-3 (Toll-like receptor 3) and resulted in a marked inhibition of downstream signaling. These findings identify pustular psoriasis as an autoinflammatory phenotype caused by defects in vesicular trafficking and demonstrate a requirement of AP-1 for Toll-like receptor homeostasis.


Subject(s)
Adaptor Protein Complex 1/genetics , Psoriasis/genetics , Psoriasis/metabolism , Toll-Like Receptor 3/metabolism , Adaptor Protein Complex 1/chemistry , Amino Acid Sequence , Amino Acid Substitution , Cell Line , Female , Gene Knockdown Techniques , Humans , Male , Molecular Sequence Data , Protein Conformation , Protein Transport/genetics
16.
Hum Mutat ; 35(2): 165-77, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24339369

ABSTRACT

Incontinentia pigmenti (IP) is an X-linked-dominant Mendelian disorder caused by mutation in the IKBKG/NEMO gene, encoding for NEMO/IKKgamma, a regulatory protein of nuclear factor kappaB (NF-kB) signaling. In more than 80% of cases, IP is due to recurrent or nonrecurrent deletions causing loss-of-function (LoF) of NEMO/IKKgamma. We review how the local architecture of the IKBKG/NEMO locus with segmental duplication and a high frequency of repetitive elements favor de novo aberrant recombination through different mechanisms producing genomic microdeletion. We report here a new microindel (c.436_471delinsT, p.Val146X) arising through a DNA-replication-repair fork-stalling-and-template-switching and microhomology-mediated-end-joining mechanism in a sporadic IP case. The LoF mutations of IKBKG/NEMO leading to IP include small insertions/deletions (indel) causing frameshift and premature stop codons, which account for 10% of cases. We here present 21 point mutations previously unreported, which further extend the spectrum of pathologic variants: 14/21 predict LoF because of premature stop codon (6/14) or frameshift (8/14), whereas 7/21 predict a partial loss of NEMO/IKKgamma activity (two splicing and five missense). We review how the analysis of IP-associated IKBKG/NEMO hypomorphic mutants has contributed to the understanding of the pathophysiological mechanism of IP disease and has provided important information on affected NF-kB signaling. We built a locus-specific database listing all IKBKG/NEMO variants, accessible at http://IKBKG.lovd.nl.


Subject(s)
Codon, Nonsense , Frameshift Mutation , I-kappa B Kinase/genetics , I-kappa B Kinase/metabolism , Incontinentia Pigmenti/genetics , NF-kappa B/metabolism , Animals , Base Sequence , Chromosomes, Human, X , Genetic Variation , Genotype , Humans , Incontinentia Pigmenti/pathology , Mutation, Missense , NF-kappa B/genetics , Phenotype , Point Mutation , Sequence Deletion , Signal Transduction
17.
Hum Genet ; 133(3): 299-310, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24142340

ABSTRACT

Congenital tufting enteropathy (CTE) is a rare and severe enteropathy recently ascribed to mutations in the epcam gene. Here we establish SPINT2, previously ascribed to congenital sodium diarrhea, as a second gene associated with CTE and report molecular and immunohistochemistry data in 57 CTE patients. Inclusion criteria were early onset diarrhea and intestinal insufficiency with the typical histological CTE abnormalities. The clinical phenotype was registered, the entire coding regions of epcam and SPINT2 sequenced, and immunostaining of EpCAM and SPINT2 performed on intestinal biopsies. An epcam mutation was involved in 41 patients (73 %) who mainly displayed isolated digestive symptoms. Mutations severely affected gene expression since the EpCAM signal on intestinal tissues was either undetectable or low and irregular. Twelve other patients (21 %) carried mutations in SPINT2, and were phenotypically characterized by systematic association with keratitis (p < 10(-4)) and, for half of them, with choanal atresia (p < 10(-4)). Dependency on parenteral nutrition (PN) was comparable in patients with epcam or SPINT2 mutations, but the frequent epcam mutation c.556-14A>G (abnormal splicing) was significantly associated with a better outcome (p = 0.032) with milder PN dependency to weaning in some cases. Finally, four patients (7 %) with isolated digestive symptoms had no detectable epcam or SPINT2 mutation. Two candidate genes, Elf3 and Claudin7, were excluded from this population. Our study allows us to separate CTE patients into at least three genetic classes, each with specific phenotypes. The genetics approach raises the question of the distinction between two congenital enteropathies. Our findings should help improve the diagnosis of CTE, guide toward strategies of long-term PN management, and limit indications for intestinal transplantation to life-threatening PN complications.


Subject(s)
Antigens, Neoplasm/genetics , Cell Adhesion Molecules/genetics , Diarrhea, Infantile/genetics , Malabsorption Syndromes/genetics , Membrane Glycoproteins/genetics , Adolescent , Antigens, Neoplasm/metabolism , Base Sequence , Case-Control Studies , Cell Adhesion Molecules/metabolism , Child , Child, Preschool , Cohort Studies , Epithelial Cell Adhesion Molecule , Female , Genetic Association Studies , Humans , Immunohistochemistry , Infant , Male , Membrane Glycoproteins/metabolism , Mutation , Parenteral Nutrition , Phenotype , Sequence Analysis, DNA , Treatment Outcome
18.
Pediatrics ; 132(4): e1043-7, 2013 Oct.
Article in English | MEDLINE | ID: mdl-24019411

ABSTRACT

YM is the first son of Tunisian consanguineous parents who developed, at 2 weeks of life, an erythematous and scaly eruption, with subsequent rapid evolution toward generalized pustular psoriasis. Afterward, cutaneous flares of diffuse erythematous rash and pustules involving the whole body appeared, with a once weekly periodicity. Intense irritability was present during flares without fever. Moreover, since 1 month of age the infant presented with diarrhea, dysphagia, and reduced feeding rate, with failure to thrive. Laboratory tests during acute flares showed marked leukocytosis, thrombocytosis, and anemia without C-reactive protein elevation. Skin biopsy and clinical presentation were consistent with pustular psoriasis; nevertheless, the patient did not respond to high-potency topical corticosteroids and retinoid acid. As the patient presented with repeated skin flares early after birth, as well as serious constitutional distress with failure to thrive, an autoinflammatory syndrome like interleukine-1-receptor antagonist deficiency or interleukin-36-receptor antagonist deficiency (DITRA) was considered. The hypothesis was reinforced by parental consanguinity, and absence of skin lesion improvement under standard topical treatment. Genetic analyses showed a homozygous mutation in the IL36RN gene (L27P), which represents the same mutation recently described in DITRA patients. At age 6 months we started treatment with the recombinant interleukin-1 receptor antagonist anakinra with efficacy both on constitutional symptoms and skin involvement. DITRA is a recently described autoinflammatory disease characterized by repeated flares of generalized pustular psoriasis, high fever, asthenia, and systemic inflammation. We report herein the first exhaustive clinical description of an infant with DITRA who was successfully treated with anakinra.


Subject(s)
Antirheumatic Agents/therapeutic use , Interleukin 1 Receptor Antagonist Protein/therapeutic use , Receptors, Interleukin/deficiency , Hereditary Autoinflammatory Diseases/genetics , Hereditary Autoinflammatory Diseases/metabolism , Humans , Infant , Interleukin 1 Receptor Antagonist Protein/deficiency , Interleukin 1 Receptor Antagonist Protein/genetics , Interleukin 1 Receptor Antagonist Protein/metabolism , Male , Receptors, Interleukin/antagonists & inhibitors , Receptors, Interleukin/genetics , Treatment Outcome
19.
Biochim Biophys Acta ; 1832(12): 2103-14, 2013 Dec.
Article in English | MEDLINE | ID: mdl-23928362

ABSTRACT

Lipin-1 deficiency is associated with massive rhabdomyolysis episodes in humans, precipitated by febrile illnesses. Despite well-known roles of lipin-1 in lipid biosynthesis and transcriptional regulation, the pathogenic mechanisms leading to rhabdomyolysis remain unknown. Here we show that primary myoblasts from lipin-1-deficient patients exhibit a dramatic decrease in LPIN1 expression and phosphatidic acid phosphatase 1 activity, and a significant accumulation of lipid droplets (LD). The expression levels of LPIN1-target genes [peroxisome proliferator-activated receptors delta and alpha (PPARδ, PPARα), peroxisome proliferator-activated receptor gamma coactivator 1-alpha (PGC-1α), acyl-coenzyme A dehydrogenase, very long (ACADVL), carnitine palmitoyltransferase IB and 2 (CPT1B and CPT2)] were not affected while lipin-2 protein level, a closely related member of the family, was increased. Microarray analysis of patients' myotubes identified 19 down-regulated and 51 up-regulated genes, indicating pleiotropic effects of lipin-1 deficiency. Special attention was paid to the up-regulated ACACB (acetyl-CoA carboxylase beta), a key enzyme in the fatty acid synthesis/oxidation balance. We demonstrated that overexpression of ACACB was associated with free fatty acid accumulation in patients' myoblasts whereas malonyl-carnitine (as a measure of malonyl-CoA) and CPT1 activity were in the normal range in basal conditions accordingly to the normal daily activity reported by the patients. Remarkably ACACB invalidation in patients' myoblasts decreased LD number and size while LPIN1 invalidation in controls induced LD accumulation. Further, pro-inflammatory treatments tumor necrosis factor alpha+Interleukin-1beta(TNF1α+IL-1ß) designed to mimic febrile illness, resulted in increased malonyl-carnitine levels, reduced CPT1 activity and enhanced LD accumulation, a phenomenon reversed by dexamethasone and TNFα or IL-1ß inhibitors. Our data suggest that the pathogenic mechanism of rhabdomyolysis in lipin-1-deficient patients combines the predisposing constitutive impairment of lipid metabolism and its exacerbation by pro-inflammatory cytokines.


Subject(s)
Cytokines/pharmacology , Inflammation Mediators/pharmacology , Lipid Metabolism Disorders/etiology , Lipids , Muscle Fibers, Skeletal/pathology , Myoblasts/pathology , Phosphatidate Phosphatase/genetics , Biomarkers/metabolism , Blotting, Western , Case-Control Studies , Cell Cycle , Cell Proliferation , Child , Child, Preschool , Endoplasmic Reticulum Stress , Female , Gene Expression Profiling , Humans , Lipid Metabolism Disorders/metabolism , Lipid Metabolism Disorders/pathology , Male , Muscle Fibers, Skeletal/drug effects , Muscle Fibers, Skeletal/metabolism , Muscle, Skeletal/drug effects , Muscle, Skeletal/metabolism , Muscle, Skeletal/pathology , Mutation/genetics , Myoblasts/drug effects , Myoblasts/metabolism , Oligonucleotide Array Sequence Analysis , Pancreatitis-Associated Proteins , Phosphatidate Phosphatase/metabolism , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Rhabdomyolysis/etiology , Rhabdomyolysis/metabolism , Rhabdomyolysis/pathology
20.
Hum Mol Genet ; 21(6): 1260-71, 2012 Mar 15.
Article in English | MEDLINE | ID: mdl-22121116

ABSTRACT

IKBKG/NEMO gene mutations cause an X-linked, dominant neuroectodermal disorder named Incontinentia Pigmenti (IP). Located at Xq28, IKBKG/NEMO has a unique genomic organization, as it is part of a segmental duplication or low copy repeat (LCR1-LCR2, >99% identical) containing the gene and its pseudogene copy (IKBKGP). In the opposite direction and outside LCR1, IKBKG/NEMO partially overlaps G6PD, whose mutations cause a common X-linked human enzymopathy. The two LCRs in the IKBKG/NEMO locus are able to recombine through non-allelic homologous recombination producing either a pathological recurrent exon 4-10 IKBKG/NEMO deletion (IKBKGdel) or benign small copy number variations. We here report that the local high frequency of micro/macro-homologies, tandem repeats and repeat/repetitive sequences make the IKBKG/NEMO locus susceptible to novel pathological IP alterations. Indeed, we describe the first two independent instances of inter-locus gene conversion, occurring between the two LCRs, that copies the IKBKGP pseudogene variants into the functional IKBKG/NEMO, causing the de novo occurrence of p.Glu390ArgfsX61 and the IKBKGdel mutations, respectively. Subsequently, by investigating a group of 20 molecularly unsolved IP subjects using a high-density quantitative polymerase chain reaction assay, we have identified seven unique de novo deletions varying from 4.8 to ∼115 kb in length. Each deletion removes partially or completely both IKBKG/NEMO and the overlapping G6PD, thereby uncovering the first deletions disrupting the G6PD gene which were found in patients with IP. Interestingly, the 4.8 kb deletion removes the conserved bidirectional promoterB, shared by the two overlapping IKBKG/NEMO and G6PD genes, leaving intact the alternative IKBKG/NEMO unidirectional promoterA. This promoter, although active in the keratinocytes of the basal dermal layer, is down-regulated during late differentiation. Genomic analysis at the breakpoint sites indicated that other mutational forces, such as non-homologous end joining, Alu-Alu-mediated recombination and replication-based events, might enhance the vulnerability of the IP locus to produce de novo pathological IP alleles.


Subject(s)
Genomics , Glucosephosphate Dehydrogenase/genetics , I-kappa B Kinase/genetics , Incontinentia Pigmenti/genetics , Incontinentia Pigmenti/pathology , Promoter Regions, Genetic/genetics , Sequence Deletion/genetics , Alleles , Cell Differentiation , Cells, Cultured , Female , Homologous Recombination , Humans , Keratinocytes/cytology , Keratinocytes/metabolism , Male , Microsatellite Repeats , Pseudogenes/genetics , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Repetitive Sequences, Nucleic Acid/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...