Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 31
Filter
2.
Genet Med ; 19(8): 875-882, 2017 08.
Article in English | MEDLINE | ID: mdl-28125082

ABSTRACT

PURPOSE: Joubert syndrome (JS) is a genetically and clinically heterogeneous ciliopathy characterized by distinct cerebellar and brainstem malformations resulting in the diagnostic "molar tooth sign" on brain imaging. To date, more than 30 JS genes have been identified, but these do not account for all patients. METHODS: In our cohort of 100 patients with JS from 86 families, we prospectively performed extensive clinical evaluation and provided molecular diagnosis using a targeted 27-gene Molecular Inversion Probes panel followed by whole-exome sequencing (WES). RESULTS: We identified the causative gene in 94% of the families; 126 (27 novel) unique potentially pathogenic variants were found in 20 genes, including KIAA0753 and CELSR2, which had not previously been associated with JS. Genotype-phenotype correlation revealed the absence of retinal degeneration in patients with TMEM67, C5orf52, or KIAA0586 variants. Chorioretinal coloboma was associated with a decreased risk for retinal degeneration and increased risk for liver disease. TMEM67 was frequently associated with kidney disease. CONCLUSION: In JS, WES significantly increases the yield for molecular diagnosis, which is essential for reproductive counseling and the option of preimplantation and prenatal diagnosis as well as medical management and prognostic counseling for the age-dependent and progressive organ-specific manifestations, including retinal, liver, and kidney disease.Genet Med advance online publication 26 January 2017.


Subject(s)
Abnormalities, Multiple/diagnosis , Abnormalities, Multiple/genetics , Cerebellum/abnormalities , Eye Abnormalities/diagnosis , Eye Abnormalities/genetics , Kidney Diseases, Cystic/diagnosis , Kidney Diseases, Cystic/genetics , Molecular Diagnostic Techniques , Retina/abnormalities , Abnormalities, Multiple/physiopathology , Adolescent , Adult , Cerebellum/physiopathology , Child , Child, Preschool , Cohort Studies , Coloboma/diagnosis , Coloboma/genetics , Eye Abnormalities/physiopathology , Female , Humans , Infant , Kidney Diseases/diagnosis , Kidney Diseases/genetics , Kidney Diseases, Cystic/physiopathology , Liver Diseases/diagnosis , Liver Diseases/genetics , Male , Molecular Probes , Prospective Studies , Retina/physiopathology , Retinal Degeneration/diagnosis , Retinal Degeneration/genetics , Whole Genome Sequencing , Young Adult
3.
J Med Genet ; 54(8): 521-529, 2017 08.
Article in English | MEDLINE | ID: mdl-28087721

ABSTRACT

BACKGROUND: Joubert syndrome is a clinically and genetically heterogeneous ciliopathy. Neuroimaging findings have not been systematically evaluated in a large cohort of patients with Joubert syndrome in correlation with molecular genetic cause and cognitive function. METHODS: Brain MRI of 110 patients with Joubert syndrome was included in this study. A comprehensive evaluation of brain MRI studies for infratentorial and supratentorial morphological abnormalities was performed. Genetic cause was identified by whole-exome sequencing, and cognitive functions were assessed with age-appropriate neurocognitive tests in a subset of patients. RESULTS: The cerebellar hemispheres were enlarged in 18% of the patients, mimicking macrocerebellum. The posterior fossa was enlarged in 42% of the patients, resembling Dandy-Walker malformation. Abnormalities of the brainstem, such as protuberance at the ventral contour of the midbrain, were present in 66% of the patients. Abnormalities of the supratentorial brain were present in approximately one-third of the patients, most commonly malrotation of the hippocampi. Mild ventriculomegaly, which typically did not require shunting, was present in 23% of the patients. No correlation between neuroimaging findings and molecular genetic cause was apparent. A novel predictor of outcome was identified; the more severe the degree of vermis hypoplasia, the worse the neurodevelopmental outcome was. CONCLUSIONS: The spectrum of neuroimaging findings in Joubert syndrome is wide. Neuroimaging does not predict the genetic cause, but may predict the neurodevelopmental outcome. A high degree of vermis hypoplasia correlates with worse neurodevelopmental outcome. This finding is important for prognostic counselling in Joubert syndrome.


Subject(s)
Abnormalities, Multiple/diagnostic imaging , Abnormalities, Multiple/psychology , Cerebellum/abnormalities , Cerebellum/diagnostic imaging , Cognition , Eye Abnormalities/diagnostic imaging , Eye Abnormalities/psychology , Kidney Diseases, Cystic/diagnostic imaging , Kidney Diseases, Cystic/psychology , Magnetic Resonance Imaging , Retina/abnormalities , Abnormalities, Multiple/genetics , Child , Child, Preschool , Cohort Studies , Eye Abnormalities/genetics , Female , Humans , Kidney Diseases, Cystic/genetics , Male , Neuroimaging , Prognosis , Retina/diagnostic imaging , Exome Sequencing
4.
Am J Med Genet A ; 173(3): 661-666, 2017 Mar.
Article in English | MEDLINE | ID: mdl-28052552

ABSTRACT

Joubert syndrome is a ciliopathy characterized by a specific constellation of central nervous system malformations that result in the pathognomonic "molar tooth sign" on imaging. More than 27 genes are associated with Joubert syndrome, but some patients do not have mutations in any of these genes. Celsr1, Celsr2, and Celsr3 are the mammalian orthologues of the drosophila planar cell polarity protein, flamingo; they play important roles in neural development, including axon guidance, neuronal migration, and cilium polarity. Here, we report bi-allelic mutations in CELSR2 in a Joubert patient with cortical heterotopia, microophthalmia, and growth hormone deficiency. © 2017 Wiley Periodicals, Inc.


Subject(s)
Abnormalities, Multiple/diagnosis , Abnormalities, Multiple/genetics , Cadherins/genetics , Cerebellum/abnormalities , Eye Abnormalities/diagnosis , Eye Abnormalities/genetics , Human Growth Hormone/deficiency , Kidney Diseases, Cystic/diagnosis , Kidney Diseases, Cystic/genetics , Microphthalmos/genetics , Mutation , Retina/abnormalities , Alleles , Cadherins/chemistry , Child , Facies , Female , High-Throughput Nucleotide Sequencing , Humans , Magnetic Resonance Imaging , Models, Molecular , Polymorphism, Single Nucleotide , Protein Conformation
5.
Br J Haematol ; 176(1): 118-123, 2017 Jan.
Article in English | MEDLINE | ID: mdl-27766632

ABSTRACT

Hermansky-Pudlak syndrome (HPS) encompasses disorders with abnormal function of lysosomes and lysosome-related organelles, and some patients who develop immunodeficiency. The basic mechanisms contributing to immune dysfunction in HPS are ill-defined. We analysed natural killer (NK) cells from patients diagnosed with HPS-1, HPS-2, HPS-4, and an unreported HPS subtype. NK cells from an HPS-2 and an unreported HPS subtype share a similar cellular phenotype with defective granule release and cytotoxicity, but differ in cytokine exocytosis. Defining NK cell activity in several types of HPS provides insights into cellular defects of the disorder and understanding of mechanisms contributing to HPS pathogenesis.


Subject(s)
Hermanski-Pudlak Syndrome/pathology , Killer Cells, Natural/pathology , Cells, Cultured , Cytoplasmic Granules/metabolism , Cytotoxicity, Immunologic , Exocytosis , Hermanski-Pudlak Syndrome/classification , Hermanski-Pudlak Syndrome/etiology , Hermanski-Pudlak Syndrome/immunology , Humans , Killer Cells, Natural/immunology , Killer Cells, Natural/metabolism , Phenotype
6.
Mol Genet Metab ; 119(3): 284-287, 2016 11.
Article in English | MEDLINE | ID: mdl-27641950

ABSTRACT

PURPOSE: Hemophilia B, an X-linked disease, manifests with recurrent soft tissue bleeding episodes. Hermansky-Pudlak syndrome, a rare autosomal recessive disorder, is characterized by oculocutaneous albinism and an increased tendency to bleed due to a platelet storage pool defect. We report a novel mutation in HPS6 in a Caucasian man with hemophilia B and oculocutaneous albinism. RESULTS: The patient was diagnosed with hemophilia B at age 4months due to recurrent soft tissue bleeding episodes, and he was also diagnosed with Hermansky-Pudlak syndrome at 32years of age due to unexplained oculocutaneous albinism. His factor IX level was markedly reduced at 13%; whole exome and Sanger sequencing showed the Durham mutation in F9 (NM_000133.3). The diagnosis of Hermansky-Pudlak syndrome subtype 6 was established by demonstrating absence of platelet delta granules on whole mount electron microscopy, an abnormal secondary wave in platelet aggregation studies, and a novel homozygous c.1114 C>T (p.Arg372*) mutation in HPS6 (NM_024747.5) on exome analysis and Sanger sequencing. Clinical phenotyping revealed no evidence of recurrent or unusual infections, interstitial lung disease or pulmonary fibrosis, or neurological disorders. The patient was treated with fresh frozen plasma, recombinant factor IX, and aminocaproic acid. Treatment with desmopressin was added to his regimen after he was diagnosed with Hermansky-Pudlak syndrome. Treatment of bleeding episodes results in effective hemostasis, and the patient has not required platelet or blood product transfusions. CONCLUSIONS: This report highlights the need to consider Hermansky-Pudlak syndrome as an etiology of oculocutaneous albinism even in patients with known hematologic disorders associated with bleeding. Identification of a novel mutation in HPS6 in an individual with hemophilia B shows that, although quite rare, patients may be diagnosed with two independent inherited bleeding disorders. No evidence of lung disease was found in this adult patient with Hermansky-Pudlak syndrome subtype 6.


Subject(s)
Albinism, Oculocutaneous/genetics , Hemophilia B/genetics , Hermanski-Pudlak Syndrome/genetics , Intracellular Signaling Peptides and Proteins/genetics , Adult , Albinism, Oculocutaneous/pathology , Exome , Female , Hemophilia B/pathology , Hermanski-Pudlak Syndrome/pathology , Humans , Male , Mutation , Phenotype
7.
J Med Genet ; 53(5): 318-29, 2016 05.
Article in English | MEDLINE | ID: mdl-27095636

ABSTRACT

BACKGROUND: Laminins are heterotrimeric complexes, consisting of α, ß and γ subunits that form a major component of basement membranes and extracellular matrix. Laminin complexes have different, but often overlapping, distributions and functions. METHODS: Under our clinical protocol, NCT00068224, we have performed extensive clinical and neuropsychiatric phenotyping, neuroimaging and molecular analysis in patients with laminin α1 (LAMA1)-associated lamininopathy. We investigated the consequence of mutations in LAMA1 using patient-derived fibroblasts and neuronal cells derived from neuronal stem cells. RESULTS: In this paper we describe individuals with biallelic mutations in LAMA1, all of whom had the cerebellar dysplasia, myopia and retinal dystrophy, in addition to obsessive compulsive traits, tics and anxiety. Patient-derived fibroblasts have impaired adhesion, reduced migration, abnormal morphology and increased apoptosis due to impaired activation of Cdc42, a member of the Rho family of GTPases that is involved in cytoskeletal dynamics. LAMA1 knockdown in human neuronal cells also showed abnormal morphology and filopodia formation, supporting the importance of LAMA1 in neuronal migration, and marking these cells potentially useful tools for disease modelling and therapeutic target discovery. CONCLUSION: This paper broadens the phenotypes associated with LAMA1 mutations. We demonstrate that LAMA1 deficiency can lead to alteration in cytoskeletal dynamics, which may invariably lead to alteration in dendrite growth and axonal formation. Estimation of disease prevalence based on population studies in LAMA1 reveals a prevalence of 1-20 in 1 000 000. TRIAL REGISTRATION NUMBER: NCT00068224.


Subject(s)
Cerebellar Diseases/metabolism , Laminin/genetics , Mutation , Myopia/metabolism , Obsessive-Compulsive Disorder/metabolism , Adult , Cell Adhesion , Cell Movement , Cerebellar Diseases/genetics , Cerebellar Diseases/physiopathology , Child , Female , Fibroblasts/metabolism , Fibroblasts/physiology , Humans , Male , Myopia/genetics , Myopia/physiopathology , Neurons/metabolism , Neurons/physiology , Obsessive-Compulsive Disorder/genetics , Obsessive-Compulsive Disorder/physiopathology , Pedigree , Retinal Dystrophies/genetics , Retinal Dystrophies/metabolism , Retinal Dystrophies/physiopathology , Syndrome , Tic Disorders/genetics , Tic Disorders/metabolism , Tic Disorders/physiopathology , Young Adult , cdc42 GTP-Binding Protein
8.
Neurology ; 86(14): 1320-1328, 2016 04 05.
Article in English | MEDLINE | ID: mdl-26944273

ABSTRACT

OBJECTIVE: To delineate the developmental and progressive neurodegenerative features in 9 young adults with the atypical form of Chediak-Higashi disease (CHD) enrolled in a natural history study. METHODS: Patients with atypical clinical features, but diagnostically confirmed CHD by standard evaluation of blood smears and molecular genotyping, underwent complete neurologic evaluation, MRI of the brain, electrophysiologic examination, and neuropsychological testing. Fibroblasts were collected to investigate the cellular phenotype and correlation with the clinical presentation. RESULTS: In 9 mildly affected patients with CHD, we documented learning and behavioral difficulties along with developmental structural abnormalities of the cerebellum and posterior fossa, which are apparent early in childhood. A range of progressive neurologic problems emerge in early adulthood, including cerebellar deficits, polyneuropathies, spasticity, cognitive decline, and parkinsonism. CONCLUSIONS: Patients with undiagnosed atypical CHD manifesting some of these wide-ranging yet nonspecific neurologic complaints may reside in general and specialty neurology clinics. The absence of the typical bleeding or infectious diathesis in mildly affected patients with CHD renders them difficult to diagnose. Identification of these individuals is important not only for close surveillance of potential CHD-related systemic complications but also for a full understanding of the natural history of CHD and the potential role of the disease-causing protein, LYST, to the pathophysiology of other neurodevelopmental and neurodegenerative disorders.


Subject(s)
Cerebellum/pathology , Chediak-Higashi Syndrome , Learning Disabilities , Neurodegenerative Diseases , Adolescent , Adult , Chediak-Higashi Syndrome/complications , Chediak-Higashi Syndrome/diagnosis , Chediak-Higashi Syndrome/genetics , Cranial Fossa, Posterior/pathology , Electromyography , Female , Follow-Up Studies , Humans , Learning Disabilities/etiology , Learning Disabilities/pathology , Learning Disabilities/physiopathology , Magnetic Resonance Imaging , Male , Neurodegenerative Diseases/etiology , Neurodegenerative Diseases/pathology , Neurodegenerative Diseases/physiopathology , Severity of Illness Index , Young Adult
9.
J Allergy Clin Immunol ; 137(4): 1165-1177, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26478006

ABSTRACT

BACKGROUND: Mutations in lysosomal trafficking regulator (LYST) cause Chediak-Higashi syndrome (CHS), a rare immunodeficiency with impaired cytotoxic lymphocyte function, mainly that of natural killer (NK) cells. Our understanding of NK cell function deficiency in patients with CHS and how LYST regulates lytic granule exocytosis is very limited. OBJECTIVE: We sought to delineate cellular defects associated with LYST mutations responsible for the impaired NK cell function seen in patients with CHS. METHODS: We analyzed NK cells from patients with CHS with missense mutations in the LYST ARM/HEAT (armadillo/huntingtin, elongation factor 3, protein phosphatase 2A, and the yeast kinase TOR1) or BEACH (beige and Chediak-Higashi) domains. RESULTS: NK cells from patients with CHS displayed severely reduced cytotoxicity. Mutations in the ARM/HEAT domain led to a reduced number of perforin-containing granules, which were significantly increased in size but able to polarize to the immunologic synapse; however, they were unable to properly fuse with the plasma membrane. Mutations in the BEACH domain resulted in formation of normal or slightly enlarged granules that had markedly impaired polarization to the IS but could be exocytosed on reaching the immunologic synapse. Perforin-containing granules in NK cells from patients with CHS did not acquire certain lysosomal markers (lysosome-associated membrane protein 1/2) but were positive for markers of transport vesicles (cation-independent mannose 6-phosphate receptor), late endosomes (Ras-associated binding protein 27a), and, to some extent, early endosomes (early endosome antigen 1), indicating a lack of integrity in the endolysosomal compartments. NK cells from patients with CHS had normal cytokine compartments and cytokine secretion. CONCLUSION: LYST is involved in regulation of multiple aspects of NK cell lytic activity, ranging from governance of lytic granule size to control of their polarization and exocytosis, as well as regulation of endolysosomal compartment identity. LYST functions in the regulated exocytosis but not in the constitutive secretion pathway.


Subject(s)
Chediak-Higashi Syndrome/physiopathology , Cytokines/metabolism , Exocytosis/physiology , Killer Cells, Natural/metabolism , Lysosomes/physiology , Vesicular Transport Proteins/genetics , Adult , Chediak-Higashi Syndrome/genetics , Female , Genetic Markers , Humans , Male , Mutation, Missense , Vesicular Transport Proteins/physiology
10.
Int J Dermatol ; 55(3): 317-21, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26499269

ABSTRACT

Chediak-Higashi syndrome (CHS; OMIM no. 214500) is an inherited multisystem disorder presenting with hypopigmentation and a propensity to infections due to immunological dysfunction. CHS generally presents in infancy with a fatal outcome, but less severe cases can present in adulthood. Treatment with bone marrow transplantation can be life-saving, so establishing a correct diagnosis is critical. The presence of large granules on examination of peripheral blood smears is suggestive of the diagnosis of CHS in most centers. However, sequencing of the lysosomal trafficking, LYST, gene confirms the diagnosis and can provide a prognosis regarding disease severity. In the case presented here, we performed molecular testing to identify the causative mutation and tabulated published mutation data from 2009 to 2014. We found a novel frameshift mutation in our case and concluded that frameshift and nonsense are the most common types of mutation in CHS, but this may be biased due to underdiagnosis of the milder and atypical forms of the disease.


Subject(s)
Chediak-Higashi Syndrome/genetics , Vesicular Transport Proteins/genetics , Chediak-Higashi Syndrome/diagnosis , Female , Frameshift Mutation , Homozygote , Humans , India , Infant
11.
Mol Genet Metab ; 114(3): 474-82, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25577287

ABSTRACT

Store-operated Ca(2+) entry is the major route of replenishment of intracellular Ca(2+) in animal cells in response to the depletion of Ca(2+) stores in the endoplasmic reticulum. It is primarily mediated by the Ca(2+)-selective release-activated Ca(2+) (CRAC) channel, which consists of the pore-forming subunits ORAI1-3 and the Ca(2+) sensors, STIM1 and STIM2. Recessive loss-of-function mutations in STIM1 or ORAI1 result in immune deficiency and nonprogressive myopathy. Heterozygous gain-of-function mutations in STIM1 cause non-syndromic myopathies as well as syndromic forms of miosis and myopathy with tubular aggregates and Stormorken syndrome; some of these syndromic forms are associated with thrombocytopenia. Increased concentration of Ca(2+) as a result of store-operated Ca(2+) entry is essential for platelet activation. The York Platelet syndrome (YPS) is characterized by thrombocytopenia, striking ultrastructural platelet abnormalities including giant electron-opaque organelles and massive, multilayered target bodies and deficiency of platelet Ca(2+) storage in delta granules. We present clinical and molecular findings in 7 YPS patients from 4 families, demonstrating that YPS patients have a chronic myopathy associated with rimmed vacuoles and heterozygous gain-of-function STIM1 mutations. These findings expand the phenotypic spectrum of STIM1-related human disorders and define the molecular basis of YPS.


Subject(s)
Blood Platelets/pathology , Channelopathies/genetics , Membrane Proteins/genetics , Muscular Diseases/genetics , Neoplasm Proteins/genetics , Adult , Blood Platelet Disorders/genetics , Blood Platelet Disorders/metabolism , Blood Platelets/physiology , Blood Platelets/ultrastructure , Calcium/metabolism , Child , Child, Preschool , Dyslexia/genetics , Dyslexia/metabolism , Erythrocytes, Abnormal/metabolism , Exome/genetics , Female , Heterozygote , Humans , Ichthyosis/genetics , Ichthyosis/metabolism , Infant , Male , Middle Aged , Migraine Disorders/genetics , Migraine Disorders/metabolism , Miosis/genetics , Miosis/metabolism , Muscle Fatigue/genetics , Muscular Diseases/metabolism , Mutation , Pedigree , Sequence Analysis, DNA , Spleen/abnormalities , Spleen/metabolism , Stromal Interaction Molecule 1 , Thrombocytopenia
12.
Mol Genet Metab ; 114(1): 62-5, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25468649

ABSTRACT

Hermansky-Pudlak syndrome (HPS) is a genetic disorder characterized by oculocutaneous albinism, bleeding tendency and susceptibility to pulmonary fibrosis. No curative therapy is available. Genetic correction directed to the lungs, bone marrow and/or gastro-intestinal tract might provide alternative forms of treatment for the diseases multi-systemic complications. We demonstrate that lentiviral-mediated gene transfer corrects the expression and function of the HPS1 gene in patient dermal melanocytes, which opens the way to development of gene therapy for HPS.


Subject(s)
Genetic Therapy , Hermanski-Pudlak Syndrome/therapy , Melanocytes/metabolism , Membrane Proteins/genetics , Cells, Cultured , Gene Expression , Genetic Vectors , Guanine Nucleotide Exchange Factors , Hermanski-Pudlak Syndrome/metabolism , Humans , Lentivirus/genetics , Melanocytes/ultrastructure , Membrane Glycoproteins/metabolism , Membrane Proteins/metabolism , Mutation , Oxidoreductases/metabolism , Proteins/genetics , Proteins/metabolism , Transduction, Genetic
13.
Orphanet J Rare Dis ; 9: 212, 2014 Dec 21.
Article in English | MEDLINE | ID: mdl-25528552

ABSTRACT

BACKGROUND: Chediak-Higashi Syndrome (CHS) is a rare autosomal recessive disease characterized by immunodeficiency, oculocutaneous albinism, neurological dysfunction, and early death. Individuals with CHS present with increased susceptibility to infections of the skin, upper-respiratory tract, gastrointestinal tract, and oral tissues. Classical CHS is caused by mutations in the gene encoding lysosomal trafficking regulator (LYST). Although defects in cytotoxic T cell lytic secretory granule secretion and neutrophil phagocytosis are suggested to contribute to the immunodeficiency in CHS, the underlying molecular mechanisms are unknown. We hypothesized that skin fibroblasts from CHS subjects exhibit impaired immune response due to defective trafficking of inflammatory factors. METHODS AND RESULTS: Primary skin fibroblasts from CHS subjects or healthy controls were assessed for genes encoding inflammatory response factors using PCR array. At baseline, we found CD14, IL1R1 and TLR-1 were down-regulated significantly (≥2 fold change) and the genes encoding TLR-3, IL-1ß and IL-6 were up-regulated in CHS cells compared to control cells. When challenged with E. coli lipopolysaccharide (LPS), CHS cells were less responsive than control cells, with only 8 genes significantly up-regulated (3-68 fold change) compared to baseline values, whereas 28 genes in control cells were significantly up-regulated at a much higher magnitude (3-4,629 fold change). In addition, 50% of the genes significantly up-regulated in LPS-treated control cells were significantly lower in LPS-treated CHS cells. IL-6, a fibroblast-derived proinflammatory cytokine essential for fighting infections was significantly lower in culture media of CHS cells with or without LPS. Furthermore, Western blot and immunofluorescent staining revealed that TLR-2 and TLR-4 were diminished on cell membranes of CHS cells and dissociated from Rab11a. CONCLUSIONS: For the first time, results from our study indicate defective trafficking of TLR-2 and TLR-4 contributes to the hyposensitive response of CHS skin fibroblasts to immunogenic challenge, providing a potential therapeutic target for clinical intervention in CHS.


Subject(s)
Chediak-Higashi Syndrome/diagnosis , Chediak-Higashi Syndrome/immunology , Fibroblasts/immunology , Immunogenetic Phenomena/immunology , Skin/immunology , Cells, Cultured , Chediak-Higashi Syndrome/genetics , Female , Fibroblasts/pathology , Gene Expression Profiling/methods , Humans , Immunogenetic Phenomena/genetics , Immunologic Deficiency Syndromes/diagnosis , Immunologic Deficiency Syndromes/genetics , Immunologic Deficiency Syndromes/immunology , Male , Skin/pathology
14.
Am J Respir Cell Mol Biol ; 50(3): 605-13, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24134621

ABSTRACT

The etiology of Hermansky-Pudlak syndrome (HPS) pulmonary fibrosis (HPSPF), a progressive interstitial lung disease with high mortality, is unknown. Galectin-3 is a ß-galactoside-binding lectin with profibrotic effects. The objective of this study was to investigate the involvement of galectin-3 in HPSPF. Galectin-3 was measured by ELISA, immunohistochemistry, and immunoblotting in human specimens from subjects with HPS and control subjects. Mechanisms of galectin-3 accumulation were studied by quantitative RT-PCR, Northern blot analysis, membrane biotinylation assays, and rescue of HPS1-deficient cells by transfection. Bronchoalveolar lavage galectin-3 concentrations were significantly higher in HPSPF compared with idiopathic pulmonary fibrosis or that from normal volunteers, and correlated with disease severity. Galectin-3 immunostaining was increased in HPSPF compared with idiopathic pulmonary fibrosis or normal lung tissue. Fibroblasts from subjects with HPS subtypes associated with pulmonary fibrosis had increased galectin-3 protein expression compared with cells from nonfibrotic HPS subtypes. Galectin-3 protein accumulation was associated with reduced Galectin-3 mRNA, normal Mucin 1 levels, and up-regulated microRNA-322 in HPSPF cells. Membrane biotinylation assays showed reduced galectin-3 and normal Mucin 1 expression at the plasma membrane in HPSPF cells compared with control cells, which suggests that galectin-3 is mistrafficked in these cells. Reconstitution of HPS1 cDNA into HPS1-deficient cells normalized galectin-3 protein and mRNA levels, as well as corrected galectin-3 trafficking to the membrane. Intracellular galectin-3 levels are regulated by HPS1 protein. Abnormal accumulation of galectin-3 may contribute to the pathogenesis of HPSPF.


Subject(s)
Galectin 3/metabolism , Hermanski-Pudlak Syndrome/complications , Lung/metabolism , Pulmonary Fibrosis/etiology , Alveolar Epithelial Cells/metabolism , Blood Proteins , Bronchoalveolar Lavage Fluid/chemistry , Case-Control Studies , Cells, Cultured , Fibroblasts/metabolism , Galectin 3/genetics , Galectins , Gene Expression Regulation , Hermanski-Pudlak Syndrome/genetics , Hermanski-Pudlak Syndrome/metabolism , Humans , Idiopathic Pulmonary Fibrosis/metabolism , Lung/pathology , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mucin-1/metabolism , Protein Transport , Pulmonary Fibrosis/diagnosis , Pulmonary Fibrosis/genetics , Pulmonary Fibrosis/metabolism , RNA, Messenger/metabolism , Severity of Illness Index , Transfection
15.
Hum Genet ; 132(12): 1383-93, 2013 Dec.
Article in English | MEDLINE | ID: mdl-23925499

ABSTRACT

Two syndromic cognitive impairment disorders have very similar craniofacial dysmorphisms. One is caused by mutations of SATB2, a transcription regulator and the other by heterozygous mutations leading to premature stop codons in UPF3B, encoding a member of the nonsense-mediated mRNA decay complex. Here we demonstrate that the products of these two causative genes function in the same pathway. We show that the SATB2 nonsense mutation in our patient leads to a truncated protein that localizes to the nucleus, forms a dimer with wild-type SATB2 and interferes with its normal activity. This suggests that the SATB2 nonsense mutation has a dominant negative effect. The patient's leukocytes had significantly decreased UPF3B mRNA compared to controls. This effect was replicated both in vitro, where siRNA knockdown of SATB2 in HEK293 cells resulted in decreased UPF3B expression, and in vivo, where embryonic tissue of Satb2 knockout mice showed significantly decreased Upf3b expression. Furthermore, chromatin immunoprecipitation demonstrates that SATB2 binds to the UPF3B promoter, and a luciferase reporter assay confirmed that SATB2 expression significantly activates gene transcription using the UPF3B promoter. These findings indicate that SATB2 activates UPF3B expression through binding to its promoter. This study emphasizes the value of recognizing disorders with similar clinical phenotypes to explore underlying mechanisms of genetic interaction.


Subject(s)
Cognition Disorders/genetics , Craniofacial Abnormalities/genetics , Matrix Attachment Region Binding Proteins/metabolism , RNA-Binding Proteins/genetics , Transcription Factors/metabolism , Transcriptional Activation , Animals , COS Cells , Cells, Cultured , Chlorocebus aethiops , HEK293 Cells , Humans , Matrix Attachment Region Binding Proteins/genetics , Mice , Mice, Knockout , Phenotype , Promoter Regions, Genetic , Syndrome , Transcription Factors/genetics
16.
Exp Cell Res ; 319(15): 2360-7, 2013 Sep 10.
Article in English | MEDLINE | ID: mdl-23810987

ABSTRACT

A fully functional immune system is essential to protect the body against pathogens and other diseases, including cancer. Vesicular trafficking provides the correct localization of proteins within all cell types, but this process is most exquisitely controlled and coordinated in immune cells because of their specialized organelles and their requirement to respond to selected stimuli. More than 60 Rab GTPases play important roles in protein trafficking, but only five Rab-encoding genes have been associated with inherited human disorders, and only one of these (Rab27a) causes an immune defect. Mutations in RAB27A cause Griscelli Syndrome type 2 (GS2), an autosomal recessive disorder of pigmentation and severe immune deficiency. In lymphocytes, Munc13-4 is an effector of Rab27a, and mutations in the gene encoding this protein (UNC13D) cause Familial Hemophagocytic Lymphohistiocytosis Type 3 (FHL3). The immunological features of GS2 and FHL3 include neutropenia, thrombocytopenia, and immunodeficiency due to impaired function of cytotoxic lymphocytes. The small number of disorders caused by mutations in genes encoding Rabs could be due to their essential functions, where defects in these genes could be lethal. However, with the increasing use of next generation sequencing technologies, more mutations in genes encoding Rabs may be identified in the near future.


Subject(s)
Chediak-Higashi Syndrome/metabolism , Crohn Disease/metabolism , Hermanski-Pudlak Syndrome/metabolism , Immunologic Deficiency Syndromes/metabolism , Lymphohistiocytosis, Hemophagocytic/metabolism , Membrane Proteins/metabolism , Piebaldism/metabolism , rab GTP-Binding Proteins/metabolism , Chediak-Higashi Syndrome/genetics , Chediak-Higashi Syndrome/pathology , Crohn Disease/genetics , Crohn Disease/pathology , Gene Expression Regulation , Hermanski-Pudlak Syndrome/genetics , Hermanski-Pudlak Syndrome/pathology , Humans , Immunologic Deficiency Syndromes/genetics , Immunologic Deficiency Syndromes/pathology , Lymphohistiocytosis, Hemophagocytic/genetics , Lymphohistiocytosis, Hemophagocytic/pathology , Membrane Proteins/genetics , Mutation , Piebaldism/genetics , Piebaldism/pathology , Primary Immunodeficiency Diseases , Protein Transport , Signal Transduction , T-Lymphocytes, Cytotoxic/metabolism , T-Lymphocytes, Cytotoxic/pathology , Transport Vesicles/metabolism , Transport Vesicles/pathology , rab GTP-Binding Proteins/genetics , rab27 GTP-Binding Proteins
17.
N Engl J Med ; 369(1): 54-65, 2013 Jul 04.
Article in English | MEDLINE | ID: mdl-23738510

ABSTRACT

BACKGROUND: Neutrophils are the predominant phagocytes that provide protection against bacterial and fungal infections. Genetically determined neutrophil disorders confer a predisposition to severe infections and reveal novel mechanisms that control vesicular trafficking, hematopoiesis, and innate immunity. METHODS: We clinically evaluated seven children from five families who had neutropenia, neutrophil dysfunction, bone marrow fibrosis, and nephromegaly. To identify the causative gene, we performed homozygosity mapping using single-nucleotide polymorphism arrays, whole-exome sequencing, immunoblotting, immunofluorescence, electron microscopy, a real-time quantitative polymerase-chain-reaction assay, immunohistochemistry, flow cytometry, fibroblast motility assays, measurements of apoptosis, and zebrafish models. Correction experiments were performed by transfecting mutant fibroblasts with the nonmutated gene. RESULTS: All seven affected children had homozygous mutations (Thr224Asn or Glu238Lys, depending on the child's ethnic origin) in VPS45, which encodes a protein that regulates membrane trafficking through the endosomal system. The level of VPS45 protein was reduced, as were the VPS45 binding partners rabenosyn-5 and syntaxin-16. The level of ß1 integrin was reduced on the surface of VPS45-deficient neutrophils and fibroblasts. VPS45-deficient fibroblasts were characterized by impaired motility and increased apoptosis. A zebrafish model of vps45 deficiency showed a marked paucity of myeloperoxidase-positive cells (i.e., neutrophils). Transfection of patient cells with nonmutated VPS45 corrected the migration defect and decreased apoptosis. CONCLUSIONS: Defective endosomal intracellular protein trafficking due to biallelic mutations in VPS45 underlies a new immunodeficiency syndrome involving impaired neutrophil function. (Funded by the National Human Genome Research Institute and others.).


Subject(s)
Immunologic Deficiency Syndromes/genetics , Neutropenia/congenital , Vesicular Transport Proteins/genetics , Animals , Child , Endosomes/metabolism , Homozygote , Humans , Immunologic Deficiency Syndromes/congenital , Immunologic Deficiency Syndromes/immunology , Mutation , Neutropenia/genetics , Neutrophils/physiology , Phenotype , Protein Transport , Vesicular Transport Proteins/metabolism , Zebrafish
18.
Traffic ; 14(7): 749-66, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23521701

ABSTRACT

BEACH (named after 'Beige and Chediak-Higashi') is a conserved ∼280 residue domain, present in nine human BEACH domain containing proteins (BDCPs). Most BDCPs are large, containing a PH-like domain for membrane association preceding their BEACH domain, and containing WD40 and other domains for ligand binding. Recent studies found that mutations in individual BDCPs cause several human diseases. BDCP alterations affect lysosome size (LYST and NSMAF), apoptosis (NSMAF), autophagy (LYST, WDFY3, LRBA), granule size (LYST, NBEAL2, NBEA) or synapse formation (NBEA). However, the roles of each BDCP in these membrane events remain controversial. After reviewing studies on individual BDCPs, we propose a unifying hypothesis that BDCPs act as scaffolding proteins that facilitate membrane events, including both fission and fusion, determined by their binding partners. BDCPs may also bind each other, enabling fusion or fission of vesicles that are not necessarily of the same type. Such mechanisms explain why different BDCPs may have roles in autophagy; each BDCP is specific for the cell type or the cargo, but not necessarily specific for attaching to the autophagosome. Further elucidation of these mechanisms, preferably carrying out the same experiment on multiple BDCPs, and possibly using patients' cells, may identify potential targets for therapy.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Adaptor Proteins, Vesicular Transport/metabolism , Disease/genetics , Membrane Proteins/metabolism , Protein Structure, Tertiary , Adaptor Proteins, Signal Transducing/chemistry , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Vesicular Transport/chemistry , Adaptor Proteins, Vesicular Transport/genetics , Amino Acid Sequence , Animals , Chediak-Higashi Syndrome/genetics , Humans , Lysosomes/metabolism , Membrane Proteins/chemistry , Membrane Proteins/genetics , Molecular Sequence Data , Polymorphism, Single Nucleotide , Protein Structure, Tertiary/genetics
20.
Hum Mutat ; 33(12): 1656-64, 2012 Dec.
Article in English | MEDLINE | ID: mdl-22753090

ABSTRACT

Arthrogryposis-renal dysfunction-cholestasis (ARC) syndrome is a rare autosomal recessive multisystem disorder caused by mutations in vacuolar protein sorting 33 homologue B (VPS33B) and VPS33B interacting protein, apical-basolateral polarity regulator (VIPAR). Cardinal features of ARC include congenital joint contractures, renal tubular dysfunction, cholestasis, severe failure to thrive, ichthyosis, and a defect in platelet alpha-granule biogenesis. Most patients with ARC do not survive past the first year of life. We report two patients presenting with a mild ARC phenotype, now 5.5 and 3.5 years old. Both patients were compound heterozygotes with the novel VPS33B donor splice-site mutation c.1225+5G>C in common. Immunoblotting and complementary DNA analysis suggest expression of a shorter VPS33B transcript, and cell-based assays show that c.1225+5G>C VPS33B mutant retains some ability to interact with VIPAR (and thus partial wild-type function). This study provides the first evidence of genotype-phenotype correlation in ARC and suggests that VPS33B c.1225+5G>C mutation predicts a mild ARC phenotype. We have established an interactive online database for ARC (https://grenada.lumc.nl/LOVD2/ARC) comprising all known variants in VPS33B and VIPAR. Also included in the database are 15 novel pathogenic variants in VPS33B and five in VIPAR.


Subject(s)
Arthrogryposis/diagnosis , Arthrogryposis/genetics , Carrier Proteins/genetics , Cholestasis/diagnosis , Cholestasis/genetics , Genetic Association Studies , Renal Insufficiency/diagnosis , Renal Insufficiency/genetics , Vesicular Transport Proteins/genetics , Child, Preschool , Female , HEK293 Cells , Heterozygote , Humans , Male , Models, Molecular , Molecular Diagnostic Techniques , Protein Transport , RNA Splice Sites , Sequence Analysis, DNA
SELECTION OF CITATIONS
SEARCH DETAIL
...