Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 34
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Am J Hum Genet ; 110(3): 499-515, 2023 03 02.
Article in English | MEDLINE | ID: mdl-36724785

ABSTRACT

Telomere maintenance 2 (TELO2), Tel2 interacting protein 2 (TTI2), and Tel2 interacting protein 1 (TTI1) are the three components of the conserved Triple T (TTT) complex that modulates activity of phosphatidylinositol 3-kinase-related protein kinases (PIKKs), including mTOR, ATM, and ATR, by regulating the assembly of mTOR complex 1 (mTORC1). The TTT complex is essential for the expression, maturation, and stability of ATM and ATR in response to DNA damage. TELO2- and TTI2-related bi-allelic autosomal-recessive (AR) encephalopathies have been described in individuals with moderate to severe intellectual disability (ID), short stature, postnatal microcephaly, and a movement disorder (in the case of variants within TELO2). We present clinical, genomic, and functional data from 11 individuals in 9 unrelated families with bi-allelic variants in TTI1. All present with ID, and most with microcephaly, short stature, and a movement disorder. Functional studies performed in HEK293T cell lines and fibroblasts and lymphoblastoid cells derived from 4 unrelated individuals showed impairment of the TTT complex and of mTOR pathway activity which is improved by treatment with Rapamycin. Our data delineate a TTI1-related neurodevelopmental disorder and expand the group of disorders related to the TTT complex.


Subject(s)
Microcephaly , Movement Disorders , Neurodevelopmental Disorders , Humans , Intracellular Signaling Peptides and Proteins , HEK293 Cells , TOR Serine-Threonine Kinases
2.
Proc Natl Acad Sci U S A ; 120(21): e2214936120, 2023 05 23.
Article in English | MEDLINE | ID: mdl-37192162

ABSTRACT

Influenza A virus (IAV) enters host cells mostly through clathrin-dependent receptor-mediated endocytosis. A single bona fide entry receptor protein supporting this entry mechanism remains elusive. Here we performed proximity ligation of biotin to host cell surface proteins in the vicinity of attached trimeric hemagglutinin-HRP and characterized biotinylated targets using mass spectrometry. This approach identified transferrin receptor 1 (TfR1) as a candidate entry protein. Genetic gain-of-function and loss-of-function experiments, as well as in vitro and in vivo chemical inhibition, confirmed the functional involvement of TfR1 in IAV entry. Recycling deficient mutants of TfR1 do not support entry, indicating that TfR1 recycling is essential for this function. The binding of virions to TfR1 via sialic acids confirmed its role as a directly acting entry factor, but unexpectedly even headless TfR1 promoted IAV particle uptake in trans. TIRF microscopy localized the entering virus-like particles in the vicinity of TfR1. Our data identify TfR1 recycling as a revolving door mechanism exploited by IAV to enter host cells.


Subject(s)
Influenza A virus , Transferrin , Influenza A virus/physiology , Virus Internalization , Endocytosis/physiology , Receptors, Transferrin/genetics , Receptors, Transferrin/metabolism
3.
J Cell Sci ; 135(13)2022 07 01.
Article in English | MEDLINE | ID: mdl-35673994

ABSTRACT

In formin-family proteins, actin filament nucleation and elongation activities reside in the formin homology 1 (FH1) and FH2 domains, with reaction rates that vary by at least 20-fold between formins. Each cell expresses distinct formins that assemble one or several actin structures, raising the question of what confers each formin its specificity. Here, using the formin Fus1 in Schizosaccharomyces pombe, we systematically probed the importance of formin nucleation and elongation rates in vivo. Fus1 assembles the actin fusion focus, necessary for gamete fusion to form the zygote during sexual reproduction. By constructing chimeric formins with combinations of FH1 and FH2 domains previously characterized in vitro, we establish that changes in formin nucleation and elongation rates have direct consequences on fusion focus architecture, and that Fus1 native high nucleation and low elongation rates are optimal for fusion focus assembly. We further describe a point mutant in Fus1 FH2 that preserves native nucleation and elongation rates in vitro but alters function in vivo, indicating an additional FH2 domain property. Thus, rates of actin assembly are tailored for assembly of specific actin structures.


Subject(s)
Schizosaccharomyces pombe Proteins , Schizosaccharomyces , Actin Cytoskeleton/metabolism , Actins/metabolism , Formins , Microfilament Proteins/metabolism , Schizosaccharomyces/genetics , Schizosaccharomyces/metabolism , Schizosaccharomyces pombe Proteins/genetics , Schizosaccharomyces pombe Proteins/metabolism
4.
Proc Natl Acad Sci U S A ; 118(36)2021 09 07.
Article in English | MEDLINE | ID: mdl-34479996

ABSTRACT

Excessive production of viral glycoproteins during infections poses a tremendous stress potential on the endoplasmic reticulum (ER) protein folding machinery of the host cell. The host cell balances this by providing more ER resident chaperones and reducing translation. For viruses, this unfolded protein response (UPR) offers the potential to fold more glycoproteins. We postulated that viruses could have developed means to limit the inevitable ER stress to a beneficial level for viral replication. Using a relevant human pathogen, influenza A virus (IAV), we first established the determinant for ER stress and UPR induction during infection. In contrast to a panel of previous reports, we identified neuraminidase to be the determinant for ER stress induction, and not hemagglutinin. IAV relieves ER stress by expression of its nonstructural protein 1 (NS1). NS1 interferes with the host messenger RNA processing factor CPSF30 and suppresses ER stress response factors, such as XBP1. In vivo viral replication is increased when NS1 antagonizes ER stress induction. Our results reveal how IAV optimizes glycoprotein expression by balancing folding capacity.


Subject(s)
Endoplasmic Reticulum Stress/physiology , Influenza A virus/genetics , Neuraminidase/metabolism , A549 Cells , Endoplasmic Reticulum/metabolism , HEK293 Cells , Host-Pathogen Interactions/physiology , Humans , Influenza A virus/metabolism , Influenza A virus/pathogenicity , Unfolded Protein Response/genetics , Unfolded Protein Response/physiology , Viral Nonstructural Proteins/genetics , Virus Replication/genetics
5.
Hum Mol Genet ; 31(1): 1-9, 2021 12 17.
Article in English | MEDLINE | ID: mdl-33693784

ABSTRACT

Knobloch syndrome is an autosomal recessive phenotype mainly characterized by retinal detachment and encephalocele caused by biallelic pathogenic variants in the COL18A1 gene. However, there are patients clinically diagnosed as Knobloch syndrome with unknown molecular etiology not linked to COL18A1. We studied an historical pedigree (published in 1998) designated as KNO2 (Knobloch type 2 syndrome with intellectual disability, autistic behavior, retinal degeneration, encephalocele). Whole exome sequencing of the two affected siblings and the normal parents resulted in the identification of a PAK2 non-synonymous substitution p.(Glu435Lys) as a causative variant. The variant was monoallelic and apparently de novo in both siblings indicating a likely germ-line mosaicism in one of the parents; the mosaicism, however, could not be observed after deep sequencing of blood parental DNA. PAK2 encodes a member of a small group of serine/threonine kinases; these P21-activating kinases (PAKs) are essential in signal transduction and cellular regulation (cytoskeletal dynamics, cell motility, death and survival signaling and cell cycle progression). Structural analysis of the PAK2 p.(Glu435Lys) variant that is located in the kinase domain of the protein predicts a possible compromise in the kinase activity. Functional analysis of the p.(Glu435Lys) PAK2 variant in transfected HEK293T cells results in a partial loss of the kinase activity. PAK2 has been previously suggested as an autism-related gene. Our results show that PAK2-induced phenotypic spectrum is broad and not fully understood. We conclude that the KNO2 syndrome in the studied family is dominant and caused by a deleterious variant in the PAK2 gene.


Subject(s)
Retinal Degeneration , Retinal Detachment , Encephalocele/diagnosis , Encephalocele/genetics , Encephalocele/pathology , HEK293 Cells , Humans , Mutation , Retinal Degeneration/genetics , Retinal Degeneration/pathology , Retinal Detachment/congenital , Retinal Detachment/genetics , p21-Activated Kinases/genetics
6.
Am J Hum Genet ; 107(2): 311-324, 2020 08 06.
Article in English | MEDLINE | ID: mdl-32738225

ABSTRACT

Aminoacyl-tRNA synthetases (ARSs) are ubiquitous, ancient enzymes that charge amino acids to cognate tRNA molecules, the essential first step of protein translation. Here, we describe 32 individuals from 21 families, presenting with microcephaly, neurodevelopmental delay, seizures, peripheral neuropathy, and ataxia, with de novo heterozygous and bi-allelic mutations in asparaginyl-tRNA synthetase (NARS1). We demonstrate a reduction in NARS1 mRNA expression as well as in NARS1 enzyme levels and activity in both individual fibroblasts and induced neural progenitor cells (iNPCs). Molecular modeling of the recessive c.1633C>T (p.Arg545Cys) variant shows weaker spatial positioning and tRNA selectivity. We conclude that de novo and bi-allelic mutations in NARS1 are a significant cause of neurodevelopmental disease, where the mechanism for de novo variants could be toxic gain-of-function and for recessive variants, partial loss-of-function.


Subject(s)
Aspartate-tRNA Ligase/genetics , Gain of Function Mutation/genetics , Loss of Function Mutation/genetics , Neurodevelopmental Disorders/genetics , RNA, Transfer, Amino Acyl/genetics , Alleles , Amino Acyl-tRNA Synthetases/genetics , Cell Line , Female , Genetic Predisposition to Disease/genetics , Humans , Male , Pedigree , RNA, Transfer/genetics , Stem Cells/physiology
7.
Clin Genet ; 104(3): 324-333, 2023 09.
Article in English | MEDLINE | ID: mdl-37317634

ABSTRACT

Intellectual developmental disorder with paroxysmal dyskinesia or seizures (IDDPADS, OMIM#619150) is an ultra-rare childhood-onset autosomal recessive movement disorder manifesting paroxysmal dyskinesia, global developmental delay, impaired cognition, progressive psychomotor deterioration and/or drug-refractory seizures. We investigated three consanguineous Pakistani families with six affected individuals presenting overlapping phenotypes partially consistent with the reported characteristics of IDDPADS. Whole exome sequencing identified a novel missense variant in Phosphodiesterase 2A (PDE2A): NM_002599.4: c.1514T > C p.(Phe505Ser) that segregated with the disease status of individuals in these families. Retrospectively, we performed haplotype analysis that revealed a 3.16 Mb shared haplotype at 11q13.4 among three families suggesting a founder effect in this region. Moreover, we also observed abnormal mitochondrial morphology in patient fibroblasts compared to controls. Belonging to diverse age groups (13 years-60 years), patients presented paroxysmal dyskinesia, developmental delay, cognitive abnormalities, speech impairment, and drug-refractory seizures with variable onset of disease (as early as 3 months of age to 7 years). Together with the previous reports, we observed that intellectual disability, progressive psychomotor deterioration, and drug-refractory seizures are consistent outcomes of the disease. However, permanent choreodystonia showed variability. We also noticed that the later onset of paroxysmal dyskinesia manifests severe attacks in terms of duration. Being the first report from Pakistan, we add to the clinical and mutation spectrum of PDE2A-related recessive disease raising the total number of patients from six to 12 and variants from five to six. Together, with our findings, the role of PDE2A is strengthened in critical physio-neurological processes.


Subject(s)
Chorea , Intellectual Disability , Humans , Intellectual Disability/genetics , Cyclic Nucleotide Phosphodiesterases, Type 2/genetics , Chorea/genetics , Retrospective Studies , Pedigree , Mutation/genetics , Consanguinity , Seizures
8.
Proc Natl Acad Sci U S A ; 117(6): 3093-3102, 2020 02 11.
Article in English | MEDLINE | ID: mdl-31980531

ABSTRACT

The catalytic activity of the protease MALT1 is required for adaptive immune responses and regulatory T (Treg)-cell development, while dysregulated MALT1 activity can lead to lymphoma. MALT1 activation requires its monoubiquitination on lysine 644 (K644) within the Ig3 domain, localized adjacent to the protease domain. The molecular requirements for MALT1 monoubiquitination and the mechanism by which monoubiquitination activates MALT1 had remained elusive. Here, we show that the Ig3 domain interacts directly with ubiquitin and that an intact Ig3-ubiquitin interaction surface is required for the conjugation of ubiquitin to K644. Moreover, by generating constitutively active MALT1 mutants that overcome the need for monoubiquitination, we reveal an allosteric communication between the ubiquitination site K644, the Ig3-protease interaction surface, and the active site of the protease domain. Finally, we show that MALT1 mutants that alter the Ig3-ubiquitin interface impact the biological response of T cells. Thus, ubiquitin binding by the Ig3 domain promotes MALT1 activation by an allosteric mechanism that is essential for its biological function.


Subject(s)
Mucosa-Associated Lymphoid Tissue Lymphoma Translocation 1 Protein , Ubiquitin , Ubiquitination , Allosteric Regulation , HEK293 Cells , Humans , Mucosa-Associated Lymphoid Tissue Lymphoma Translocation 1 Protein/chemistry , Mucosa-Associated Lymphoid Tissue Lymphoma Translocation 1 Protein/genetics , Mucosa-Associated Lymphoid Tissue Lymphoma Translocation 1 Protein/metabolism , Mutation , Protein Binding , Protein Domains , Ubiquitin/chemistry , Ubiquitin/metabolism , Ubiquitination/genetics , Ubiquitination/physiology
9.
Hum Mol Genet ; 29(7): 1132-1143, 2020 05 08.
Article in English | MEDLINE | ID: mdl-32129449

ABSTRACT

The molecular cause of the majority of rare autosomal recessive disorders remains unknown. Consanguinity due to extensive homozygosity unravels many recessive phenotypes and facilitates the detection of novel gene-disease links. Here, we report two siblings with phenotypic signs, including intellectual disability (ID), developmental delay and microcephaly from a Pakistani consanguineous family in which we have identified homozygosity for p(Tyr103His) in the PSMB1 gene (Genbank NM_002793) that segregated with the disease phenotype. PSMB1 encodes a ß-type proteasome subunit (i.e. ß6). Modeling of the p(Tyr103His) variant indicates that this variant weakens the interactions between PSMB1/ß6 and PSMA5/α5 proteasome subunits and thus destabilizes the 20S proteasome complex. Biochemical experiments in human SHSY5Y cells revealed that the p(Tyr103His) variant affects both the processing of PSMB1/ß6 and its incorporation into proteasome, thus impairing proteasome activity. CRISPR/Cas9 mutagenesis or morpholino knock-down of the single psmb1 zebrafish orthologue resulted in microcephaly, microphthalmia and reduced brain size. Genetic evidence in the family and functional experiments in human cells and zebrafish indicates that PSMB1/ß6 pathogenic variants are the cause of a recessive disease with ID, microcephaly and developmental delay due to abnormal proteasome assembly.


Subject(s)
Dwarfism/genetics , Microcephaly/genetics , Proteasome Endopeptidase Complex/genetics , Alleles , Animals , Child , Consanguinity , Developmental Disabilities/complications , Developmental Disabilities/genetics , Developmental Disabilities/pathology , Dwarfism/complications , Female , Homozygote , Humans , Intellectual Disability/complications , Intellectual Disability/genetics , Intellectual Disability/pathology , Male , Microcephaly/complications , Microcephaly/pathology , Models, Molecular , Pedigree , Phenotype , Zebrafish/genetics
10.
Hum Mol Genet ; 29(4): 618-623, 2020 03 13.
Article in English | MEDLINE | ID: mdl-31903486

ABSTRACT

In a consanguineous Pakistani family with two affected individuals, a homozygous variant Gly399Val in the eighth transmembrane domain of the taurine transporter SLC6A6 was identified resulting in a hypomorph transporting capacity of ~15% compared with normal. Three-dimensional modeling of this variant has indicated that it likely causes displacement of the Tyr138 (TM3) side chain, important for transport of taurine. The affected individuals presented with rapidly progressive childhood retinal degeneration, cardiomyopathy and almost undetectable plasma taurine levels. Oral taurine supplementation of 100 mg/kg/day resulted in maintenance of normal blood taurine levels. Following approval by the ethics committee, a long-term supplementation treatment was introduced. Remarkably, after 24-months, the cardiomyopathy was corrected in both affected siblings, and in the 6-years-old, the retinal degeneration was arrested, and the vision was clinically improved. Similar therapeutic approaches could be employed in Mendelian phenotypes caused by the dysfunction of the hundreds of other molecular transporters.


Subject(s)
Cardiomyopathies/drug therapy , Membrane Glycoproteins/deficiency , Membrane Transport Proteins/deficiency , Retinal Degeneration/drug therapy , Taurine/therapeutic use , Adolescent , Biological Transport , Cardiomyopathies/metabolism , Cardiomyopathies/pathology , Child , Female , Humans , Male , Pedigree , Retinal Degeneration/metabolism , Retinal Degeneration/pathology
11.
Am J Hum Genet ; 104(6): 1073-1087, 2019 06 06.
Article in English | MEDLINE | ID: mdl-31079899

ABSTRACT

Cargo transport along the cytoplasmic microtubular network is essential for neuronal function, and cytoplasmic dynein-1 is an established molecular motor that is critical for neurogenesis and homeostasis. We performed whole-exome sequencing, homozygosity mapping, and chromosomal microarray studies in five individuals from three independent pedigrees and identified likely-pathogenic variants in DYNC1I2 (Dynein Cytoplasmic 1 Intermediate Chain 2), encoding a component of the cytoplasmic dynein 1 complex. In a consanguineous Pakistani family with three affected individuals presenting with microcephaly, severe intellectual disability, simplification of cerebral gyration, corpus callosum hypoplasia, and dysmorphic facial features, we identified a homozygous splice donor site variant (GenBank: NM_001378.2:c.607+1G>A). We report two additional individuals who have similar neurodevelopmental deficits and craniofacial features and harbor deleterious variants; one individual bears a c.740A>G (p.Tyr247Cys) change in trans with a 374 kb deletion encompassing DYNC1I2, and an unrelated individual harbors the compound-heterozygous variants c.868C>T (p.Gln290∗) and c.740A>G (p.Tyr247Cys). Zebrafish larvae subjected to CRISPR-Cas9 gene disruption or transient suppression of dync1i2a displayed significantly altered craniofacial patterning with concomitant reduction in head size. We monitored cell death and cell cycle progression in dync1i2a zebrafish models and observed significantly increased apoptosis, likely due to prolonged mitosis caused by abnormal spindle morphology, and this finding offers initial insights into the cellular basis of microcephaly. Additionally, complementation studies in zebrafish demonstrate that p.Tyr247Cys attenuates gene function, consistent with protein structural analysis. Our genetic and functional data indicate that DYNC1I2 dysfunction probably causes an autosomal-recessive microcephaly syndrome and highlight further the critical roles of the dynein-1 complex in neurodevelopment.


Subject(s)
Craniofacial Abnormalities/etiology , Dyneins/genetics , Intellectual Disability/etiology , Intracranial Arteriovenous Malformations/etiology , Microcephaly/etiology , Mutation , Zebrafish/growth & development , Adult , Alleles , Amino Acid Sequence , Animals , Child, Preschool , Craniofacial Abnormalities/pathology , Dyneins/chemistry , Dyneins/metabolism , Exome , Female , Homozygote , Humans , Infant , Intellectual Disability/pathology , Intracranial Arteriovenous Malformations/pathology , Male , Microcephaly/pathology , Pedigree , Phenotype , Protein Conformation , Sequence Homology , Exome Sequencing , Young Adult , Zebrafish/genetics , Zebrafish/metabolism
12.
Genet Med ; 24(7): 1583-1591, 2022 07.
Article in English | MEDLINE | ID: mdl-35499524

ABSTRACT

PURPOSE: CTR9 is a subunit of the PAF1 complex (PAF1C) that plays a crucial role in transcription regulation by binding CTR9 to RNA polymerase II. It is involved in transcription-coupled histone modification through promoting H3K4 and H3K36 methylation. We describe the clinical and molecular studies in 13 probands, harboring likely pathogenic CTR9 missense variants, collected through GeneMatcher. METHODS: Exome sequencing was performed in all individuals. CTR9 variants were assessed through 3-dimensional modeling of the activated human transcription complex Pol II-DSIF-PAF-SPT6 and the PAF1/CTR9 complex. H3K4/H3K36 methylation analysis, mitophagy assessment based on tetramethylrhodamine ethyl ester perchlorate immunofluorescence, and RNA-sequencing in skin fibroblasts from 4 patients was performed. RESULTS: Common clinical findings were variable degrees of intellectual disability, hypotonia, joint hyperlaxity, speech delay, coordination problems, tremor, and autism spectrum disorder. Mild dysmorphism and cardiac anomalies were less frequent. For 11 CTR9 variants, de novo occurrence was shown. Three-dimensional modeling predicted a likely disruptive effect of the variants on local CTR9 structure and protein interaction. Additional studies in fibroblasts did not unveil the downstream functional consequences of the identified variants. CONCLUSION: We describe a neurodevelopmental disorder caused by (mainly) de novo variants in CTR9, likely affecting PAF1C function.


Subject(s)
Autism Spectrum Disorder , Intellectual Disability , Neurodevelopmental Disorders , Phosphoproteins , Transcription Factors , Gene Expression Regulation , Heterozygote , Humans , Intellectual Disability/genetics , Neurodevelopmental Disorders/genetics , Phosphoproteins/genetics , Transcription Factors/genetics
13.
Hum Mol Genet ; 28(6): 972-979, 2019 03 15.
Article in English | MEDLINE | ID: mdl-30481285

ABSTRACT

FBXL3 (F-Box and Leucine Rich Repeat Protein 3) encodes a protein that contains an F-box and several tandem leucine-rich repeats (LRR) domains. FBXL3 is part of the SCF (Skp1-Cullin-F box protein) ubiquitin ligase complex that binds and leads to phosphorylation-dependent degradation of the central clock protein cryptochromes (CRY1 and CRY2) by the proteasome and its absence causes circadian phenotypes in mice and behavioral problems. No FBXL3-related phenotypes have been described in humans. By a combination of exome sequencing and homozygosity mapping, we analyzed two consanguineous families with intellectual disability and identified homozygous loss-of-function (LoF) variants in FBXL3. In the first family, from Pakistan, an FBXL3 frameshift variant [NM_012158.2:c.885delT:p.(Leu295Phefs*25)] was the onlysegregating variant in five affected individuals in two family loops (LOD score: 3.12). In the second family, from Lebanon, we identified a nonsense variant [NM_012158.2:c.445C>T:p.(Arg149*)]. In a third patient from Italy, a likely deleterious non-synonymous variant [NM_012158.2:c.1072T>C:p.(Cys358Arg)] was identified in homozygosity. Protein 3D modeling predicted that the Cys358Arg change influences the binding with CRY2 by destabilizing the structure of the FBXL3, suggesting that this variant is also likely to be LoF. The eight affected individuals from the three families presented with a similar phenotype that included intellectual disability, developmental delay, short stature and mild facial dysmorphism, mainly large nose with a bulbous tip. The phenotypic similarity and the segregation analysis suggest that FBXL3 biallelic, LoF variants link this gene with syndromic autosomal recessive developmental delay/intellectual disability.


Subject(s)
Alleles , Developmental Disabilities/genetics , Dwarfism/genetics , F-Box Proteins/genetics , Genetic Variation , Intellectual Disability/genetics , Adult , Consanguinity , DNA Mutational Analysis , Developmental Disabilities/diagnosis , Dwarfism/diagnosis , F-Box Proteins/chemistry , Facies , Female , Homozygote , Humans , Intellectual Disability/diagnosis , Male , Middle Aged , Models, Molecular , Pedigree , Phenotype , Protein Conformation , Structure-Activity Relationship , Young Adult
14.
Int J Mol Sci ; 21(2)2020 Jan 18.
Article in English | MEDLINE | ID: mdl-31963646

ABSTRACT

Immune checkpoints are crucial in the maintenance of antitumor immune responses. The activation or blockade of immune checkpoints is dependent on the interactions between receptors and ligands; such interactions can provide inhibitory or stimulatory signals, including the enhancement or suppression of T-cell proliferation, differentiation, and/or cytokine secretion. B-and T-lymphocyte attenuator (BTLA) is a lymphoid-specific cell surface receptor which is present on T-cells and interacts with herpes virus entry mediator (HVEM), which is present on tumor cells. The binding of HVEM to BTLA triggers an inhibitory signal which attenuates the immune response. This feature is interesting for studying the molecular interactions between HVEM and BTLA, as they may be targeted for novel immunotherapies. This work was based on the crystal structure of the BTLA/HVEM complex showing that BTLA binds the N-terminal cysteine-rich domain of HVEM. We investigated the amino acid sequence of HVEM and used molecular modeling methods to develop inhibitors of the BTLA/HVEM interaction. We synthesized novel compounds and determined their ability to interact with the BTLA protein and inhibit the formation of the BTLA/HVEM complex. Our results suggest that the HVEM (14-39) peptide is a potent inhibitor of the formation of the BTLA/HVEM protein complex.


Subject(s)
Disulfides/chemistry , Peptides/pharmacology , Receptors, Immunologic/metabolism , Receptors, Tumor Necrosis Factor, Member 14/metabolism , Binding Sites/drug effects , Crystallography, X-Ray , Humans , Models, Molecular , Molecular Docking Simulation , Peptides/chemical synthesis , Peptides/chemistry , Protein Binding/drug effects , Protein Conformation , Receptors, Immunologic/chemistry , Receptors, Tumor Necrosis Factor, Member 14/chemistry
15.
Genet Med ; 20(7): 778-784, 2018 07.
Article in English | MEDLINE | ID: mdl-28837161

ABSTRACT

PURPOSE: To elucidate the novel molecular cause in two unrelated consanguineous families with autosomal recessive intellectual disability. METHODS: A combination of homozygosity mapping and exome sequencing was used to locate the plausible genetic defect in family F162, while only exome sequencing was followed in the family PKMR65. The protein 3D structure was visualized with the University of California-San Francisco Chimera software. RESULTS: All five patients from both families presented with severe intellectual disability, aggressive behavior, and speech and motor delay. Four of the five patients had microcephaly. We identified homozygous missense variants in LINGO1, p.(Arg290His) in family F162 and p.(Tyr288Cys) in family PKMR65. Both variants were predicted to be pathogenic, and segregated with the phenotype in the respective families. Molecular modeling of LINGO1 suggests that both variants interfere with the glycosylation of the protein. CONCLUSION: LINGO1 is a transmembrane receptor, predominantly found in the central nervous system. Published loss-of-function studies in mouse and zebrafish have established a crucial role of LINGO1 in normal neuronal development and central nervous system myelination by negatively regulating oligodendrocyte differentiation and neuronal survival. Taken together, our results indicate that biallelic LINGO1 missense variants cause autosomal recessive intellectual disability in humans.


Subject(s)
Intellectual Disability/genetics , Membrane Proteins/genetics , Nerve Tissue Proteins/genetics , Alleles , Chromosome Mapping/methods , Family , Female , Gene Frequency/genetics , Genotype , Homozygote , Humans , Language Development Disorders/genetics , Male , Membrane Proteins/physiology , Microcephaly/genetics , Motor Activity/genetics , Mutation, Missense/genetics , Nerve Tissue Proteins/physiology , Pakistan , Pedigree , Phenotype , Sequence Analysis, Protein , Exome Sequencing
16.
Blood ; 128(11): 1490-502, 2016 09 15.
Article in English | MEDLINE | ID: mdl-27369867

ABSTRACT

Angioimmunoblastic T-cell lymphoma (AITL) and other lymphomas derived from follicular T-helper cells (TFH) represent a large proportion of peripheral T-cell lymphomas (PTCLs) with poorly understood pathogenesis and unfavorable treatment results. We investigated a series of 85 patients with AITL (n = 72) or other TFH-derived PTCL (n = 13) by targeted deep sequencing of a gene panel enriched in T-cell receptor (TCR) signaling elements. RHOA mutations were identified in 51 of 85 cases (60%) consisting of the highly recurrent dominant negative G17V variant in most cases and a novel K18N in 3 cases, the latter showing activating properties in in vitro assays. Moreover, half of the patients carried virtually mutually exclusive mutations in other TCR-related genes, most frequently in PLCG1 (14.1%), CD28 (9.4%, exclusively in AITL), PI3K elements (7%), CTNNB1 (6%), and GTF2I (6%). Using in vitro assays in transfected cells, we demonstrated that 9 of 10 PLCG1 and 3 of 3 CARD11 variants induced MALT1 protease activity and increased transcription from NFAT or NF-κB response element reporters, respectively. Collectively, the vast majority of variants in TCR-related genes could be classified as gain-of-function. Accordingly, the samples with mutations in TCR-related genes other than RHOA had transcriptomic profiles enriched in signatures reflecting higher T-cell activation. Although no correlation with presenting clinical features nor significant impact on survival was observed, the presence of TCR-related mutations correlated with early disease progression. Thus, targeting of TCR-related events may hold promise for the treatment of TFH-derived lymphomas.


Subject(s)
Genes, T-Cell Receptor/genetics , Immunoblastic Lymphadenopathy/genetics , Lymphoma, Follicular/genetics , Lymphoma, T-Cell, Peripheral/genetics , Mutation/genetics , Signal Transduction , T-Lymphocytes, Helper-Inducer/immunology , rhoA GTP-Binding Protein/genetics , Biomarkers, Tumor/genetics , Cohort Studies , Follow-Up Studies , High-Throughput Nucleotide Sequencing/methods , Humans , Immunoblastic Lymphadenopathy/immunology , Immunoblastic Lymphadenopathy/pathology , Lymphoma, Follicular/immunology , Lymphoma, Follicular/pathology , Lymphoma, T-Cell, Peripheral/immunology , Lymphoma, T-Cell, Peripheral/pathology , Neoplasm Staging , Prognosis
17.
J Immunol ; 196(7): 3199-211, 2016 Apr 01.
Article in English | MEDLINE | ID: mdl-26921308

ABSTRACT

We have designed and validated a novel generic platform for production of tetravalent IgG1-like chimeric bispecific Abs. The VH-CH1-hinge domains of mAb2 are fused through a peptidic linker to the N terminus of mAb1 H chain, and paired mutations at the CH1-CL interface mAb1 are introduced that force the correct pairing of the two different free L chains. Two different sets of these CH1-CL interface mutations, called CR3 and MUT4, were designed and tested, and prototypic bispecific Abs directed against CD5 and HLA-DR were produced (CD5xDR). Two different hinge sequences between mAb1 and mAb2 were also tested in the CD5xDR-CR3 or -MUT4 background, leading to bispecific Ab (BsAbs) with a more rigid or flexible structure. All four Abs produced bound with good specificity and affinity to CD5 and HLA-DR present either on the same target or on different cells. Indeed, the BsAbs were able to efficiently redirect killing of HLA-DR(+) leukemic cells by human CD5(+) cytokine-induced killer T cells. Finally, all BsAbs had a functional Fc, as shown by their capacity to activate human complement and NK cells and to mediate phagocytosis. CD5xDR-CR3 was chosen as the best format because it had overall the highest functional activity and was very stable in vitro in both neutral buffer and in serum. In vivo, CD5xDR-CR3 was shown to have significant therapeutic activity in a xenograft model of human leukemia.


Subject(s)
Antibodies, Bispecific/biosynthesis , Antibodies, Bispecific/genetics , Immunoglobulin G/biosynthesis , Immunoglobulin G/genetics , Recombinant Fusion Proteins/biosynthesis , Recombinant Fusion Proteins/genetics , Amino Acid Sequence , Animals , Antibodies, Bispecific/chemistry , Antibodies, Bispecific/immunology , Antibodies, Bispecific/isolation & purification , Antigens/immunology , Baculoviridae/genetics , Cell Line , Drug Design , Gene Expression , Genetic Vectors/genetics , Humans , Immunoglobulin G/chemistry , Immunoglobulin G/immunology , Immunoglobulin G/isolation & purification , Models, Molecular , Molecular Sequence Data , Mutation , Protein Binding/immunology , Protein Conformation , Protein Stability , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/immunology , Recombinant Fusion Proteins/isolation & purification , Sequence Alignment , Surface Plasmon Resonance
18.
EMBO J ; 30(20): 4126-41, 2011 Sep 20.
Article in English | MEDLINE | ID: mdl-21934648

ABSTRACT

The V-ATPase V(0) sector associates with the peripheral V(1) sector to form a proton pump. V(0) alone has an additional function, facilitating membrane fusion in the endocytic and late exocytic pathways. V(0) contains a hexameric proteolipid cylinder, which might support fusion as proposed in proteinaceous pore models. To test this, we randomly mutagenized proteolipids. We recovered alleles that preserve proton translocation, normal SNARE activation and trans-SNARE pairing but that impair lipid and content mixing. Critical residues were found in all subunits of the proteolipid ring. They concentrate within the bilayer, close to the ring subunit interfaces. The fusion-impairing proteolipid substitutions stabilize the interaction of V(0) with V(1). Deletion of the vacuolar v-SNARE Nyv1 has the same effect, suggesting that both types of mutations similarly alter the conformation of V(0). Also covalent linkage of subunits in the proteolipid cylinder blocks vacuole fusion. We propose that a SNARE-dependent conformational change in V(0) proteolipids might stimulate fusion by creating a hydrophobic crevice that promotes lipid reorientation and formation of a lipidic fusion pore.


Subject(s)
Lipid Metabolism , Membrane Fusion , Proteolipids/metabolism , SNARE Proteins/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae/metabolism , Vacuoles/metabolism , Vacuolar Proton-Translocating ATPases/metabolism
19.
FASEB J ; 28(11): 4792-805, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25138159

ABSTRACT

Serine proteases, serine protease inhibitors, and protease-activated receptors (PARs) are responsible for several human skin disorders characterized by impaired epidermal permeability barrier function, desquamation, and inflammation. In this study, we addressed the consequences of a catalytically dead serine protease on epidermal homeostasis, the activation of PAR2 and the inhibition by the serine protease inhibitor nexin-1. The catalytically inactive serine protease CAP1/Prss8, when ectopically expressed in the mouse, retained the ability to induce skin disorders as well as its catalytically active counterpart (75%, n=81). Moreover, this phenotype was completely normalized in a PAR2-null background, indicating that the effects mediated by the catalytically inactive CAP1/Prss8 depend on PAR2 (95%, n=131). Finally, nexin-1 displayed analogous inhibitory capacity on both wild-type and inactive mutant CAP1/Prss8 in vitro and in vivo (64% n=151 vs. 89% n=109, respectively), indicating that the catalytic site of CAP1/Prss8 is dispensable for nexin-1 inhibition. Our results demonstrate a novel inhibitory interaction between CAP1/Prss8 and nexin-1, opening the search for specific CAP1/Prss8 antagonists that are independent of its catalytic activity.


Subject(s)
Receptor, PAR-2/metabolism , Serine Endopeptidases/metabolism , Serpin E2/metabolism , Skin/metabolism , Animals , Catalysis , Homeostasis/physiology , Mice , Phenotype , Protease Inhibitors/metabolism
20.
Am J Pathol ; 181(2): 605-15, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22705055

ABSTRACT

CAP1/Prss8 is a membrane-bound serine protease involved in the regulation of several different effectors, such as the epithelial sodium channel ENaC, the protease-activated receptor PAR2, the tight junction proteins, and the profilaggrin polypeptide. Recently, the V170D and the G54-P57 deletion mutations within the CAP1/Prss8 gene, identified in mouse frizzy (fr) and rat hairless (fr(CR)) animals, respectively, have been proposed to be responsible for their skin phenotypes. In the present study, we analyzed those mutations, revealing a change in the protein structure, a modification of the glycosylation state, and an overall reduction in the activation of ENaC of the two mutant proteins. In vivo analyses demonstrated that both fr and fr(CR) mutant animals present analogous reduction of embryonic viability, similar histologic aberrations at the level of the skin, and a significant decrease in the activity of ENaC in the distal colon compared with their control littermates. Hairless rats additionally had dehydration defects in skin and intestine and significant reduction in the body weight. In conclusion, we provided molecular and functional evidence that CAP1/Prss8 mutations are accountable for the defects in fr and fr(CR) animals, and we furthermore demonstrate a decreased function of the CAP1/Prss8 mutant proteins. Therefore, fr and fr(CR) animals are suitable models to investigate the consequences of CAP1/Prss8 action on its target proteins in the whole organism.


Subject(s)
Embryo, Mammalian/metabolism , Embryo, Mammalian/pathology , Epithelial Sodium Channels/metabolism , Mutation/genetics , Serine Endopeptidases/genetics , Skin/embryology , Skin/pathology , Amino Acid Sequence , Animals , Body Weight , Dehydration/metabolism , Dehydration/pathology , Gene Expression Regulation , HEK293 Cells , Hair/pathology , Humans , Inheritance Patterns/genetics , Ion Channel Gating , Mice , Models, Animal , Models, Molecular , Molecular Sequence Data , Mutant Proteins/metabolism , Organ Specificity , Phenotype , Protein Structure, Tertiary , Rats , Serine Endopeptidases/chemistry , Serine Endopeptidases/metabolism , Skin/metabolism , Skin/physiopathology , Structural Homology, Protein , Xenopus
SELECTION OF CITATIONS
SEARCH DETAIL