Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 75
Filter
Add more filters

Publication year range
1.
Proc Natl Acad Sci U S A ; 118(47)2021 11 23.
Article in English | MEDLINE | ID: mdl-34782459

ABSTRACT

Although there have been many studies of gene variant association with different stages of HIV/AIDS progression in United States and European cohorts, few gene-association studies have assessed genic determinants in sub-Saharan African populations, which have the highest density of HIV infections worldwide. We carried out genome-wide association studies on 766 study participants at risk for HIV-1 subtype C (HIV-1C) infection in Botswana. Three gene associations (AP3B1, PTPRA, and NEO1) were shown to have significant association with HIV-1C acquisition. Each gene association was replicated within Botswana or in the United States-African American or United States-European American AIDS cohorts or in both. Each associated gene has a prior reported influence on HIV/AIDS pathogenesis. Thirteen previously discovered AIDS restriction genes were further replicated in the Botswana cohorts, extending our confidence in these prior AIDS restriction gene reports. This work presents an early step toward the identification of genetic variants associated with and affecting HIV acquisition or AIDS progression in the understudied HIV-1C afflicted Botswana population.


Subject(s)
Genetic Variation , Genome-Wide Association Study , HIV Infections/genetics , Acquired Immunodeficiency Syndrome , Adaptor Protein Complex 3/genetics , Adaptor Protein Complex beta Subunits/genetics , Botswana/epidemiology , Genotype , HIV Infections/epidemiology , Humans , Nerve Tissue Proteins/genetics , Receptor-Like Protein Tyrosine Phosphatases, Class 4/genetics , Receptors, Cell Surface/genetics
2.
Am J Hum Genet ; 105(5): 1023-1029, 2019 11 07.
Article in English | MEDLINE | ID: mdl-31630788

ABSTRACT

We describe unrelated individuals with ichthyosis, failure to thrive, thrombocytopenia, photophobia, and progressive hearing loss. Each have bi-allelic mutations in AP1B1, the gene encoding the ß subunit of heterotetrameric adaptor protein 1 (AP-1) complexes, which mediate endomembrane polarization, sorting, and transport. In affected keratinocytes the AP-1 ß subunit is lost, and the γ subunit is greatly reduced, demonstrating destabilization of the AP-1 complex. Affected cells and tissue contain an abundance of abnormal vesicles and show hyperproliferation, abnormal epidermal differentiation, and derangement of intercellular junction proteins. Transduction of affected cells with wild-type AP1B1 rescues the vesicular phenotype, conclusively establishing that loss of AP1B1 function causes this disorder.


Subject(s)
Adaptor Protein Complex 1/genetics , Adaptor Protein Complex beta Subunits/genetics , Deafness/genetics , Genes, Recessive/genetics , Ichthyosis/genetics , Mutation/genetics , Photophobia/genetics , Cell Differentiation/genetics , Cell Proliferation/genetics , Female , Hearing Loss/genetics , Humans , Male , Phenotype , Protein Subunits/genetics , Protein Transport/genetics , Thrombocytopenia/genetics
3.
Am J Hum Genet ; 105(5): 1016-1022, 2019 11 07.
Article in English | MEDLINE | ID: mdl-31630791

ABSTRACT

MEDNIK syndrome (mental retardation, enteropathy, deafness, peripheral neuropathy, ichthyosis, and keratoderma) is an autosomal-recessive disorder caused by bi-allelic mutations in AP1S1, encoding the small σ subunit of the AP-1 complex. Central to the pathogenesis of MEDNIK syndrome is abnormal AP-1-mediated trafficking of copper transporters; this abnormal trafficking results in a hybrid phenotype combining the copper-deficiency-related characteristics of Menkes disease and the copper-toxicity-related characteristics of Wilson disease. We describe three individuals from two unrelated families in whom a MEDNIK-like phenotype segregates with two homozygous null variants in AP1B1, encoding the large ß subunit of the AP-1 complex. Similar to individuals with MEDNIK syndrome, the affected individuals we report display abnormal copper metabolism, evidenced by low plasma copper and ceruloplasmin, but lack evidence of copper toxicity in the liver. Functional characterization of fibroblasts derived from affected individuals closely resembles the abnormal ATP7A trafficking described in MEDNIK syndrome both at baseline and in response to copper treatment. Taken together, our results expand the list of inborn errors of copper metabolism.


Subject(s)
Adaptor Protein Complex 1/genetics , Adaptor Protein Complex beta Subunits/genetics , Genetic Diseases, Inborn/genetics , Mutation/genetics , Protein Subunits/genetics , Cation Transport Proteins/genetics , Child, Preschool , Copper-Transporting ATPases/genetics , Female , Hepatolenticular Degeneration/genetics , Homozygote , Humans , Infant , Male , Phenotype , Protein Transport/genetics , Syndrome
4.
Muscle Nerve ; 66(2): 216-222, 2022 08.
Article in English | MEDLINE | ID: mdl-35508598

ABSTRACT

INTRODUCTION/AIMS: Lambert-Eaton myasthenic syndrome (LEMS) is an autoimmune neuromuscular junction disorder. Long noncoding RNA (lncRNA) can regulate the expression of mRNA and is involved in the development of autoimmune diseases, but few genetic studies are available. In this study we aimed to explore the lncRNA and mRNA changes of LEMS. METHODS: Plasma lncRNA and mRNA expression profiles of three LEMS patients with small cell lung cancer (SCLC) and three matched healthy controls were analyzed by microarray. Differentially expressed lncRNAs and adjacent mRNAs were jointly analyzed, and candidates were verified by quantitative real-time polymerase chain reaction (qRT-PCR). The identified genes were subsequently evaluated in 9, 8, and 4 patients with paraneoplastic LEMS, nontumor LEMS, and SCLC, respectively. Gene ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) analyses were performed to determine possible functions. RESULTS: A total of 320 lncRNA and 168 mRNAs were differentially expressed in the three LEMS with SCLC and compared with healthy controls. Among these, lncRNA LOC338963 and its neighboring mRNA AP3B2 were upregulated jointly, which was confirmed by qRT-PCR. qRT-PCR revealed significant upregulation of the two genes in patients with paraneoplastic LEMS compared with nontumor LEMS or SCLC. GO analysis of AP3B2 identified the enrichment terms anterograde synaptic vesicle transport and establishment of synaptic vesicle localization. KEEG analysis showed that AP3B2 was enriched in lysosomal pathways. DISCUSSION: LOC338963 and AP3B2 were upregulated in patients with paraneoplastic LEMS, suggesting their involvement in pathogenesis. These genes could be targets for exploring the pathomechanism of paraneoplastic LEMS.


Subject(s)
Adaptor Protein Complex 3 , Adaptor Protein Complex beta Subunits , Lambert-Eaton Myasthenic Syndrome , RNA, Long Noncoding , Adaptor Protein Complex 3/genetics , Adaptor Protein Complex beta Subunits/genetics , Humans , Lung Neoplasms , RNA, Long Noncoding/blood , RNA, Long Noncoding/genetics , RNA, Messenger , Small Cell Lung Carcinoma
5.
J Virol ; 94(2)2020 01 06.
Article in English | MEDLINE | ID: mdl-31694949

ABSTRACT

Influenza A virus (IAV) coopts numerous host factors to complete its replication cycle. Here, we identify free fatty acid receptor 2 (FFAR2) as a cofactor for IAV entry into host cells. We found that downregulation of FFAR2 or Ffar2 expression significantly reduced the replication of IAV in A549 or RAW 264.7 cells. The treatment of A549 cells with small interfering RNA (siRNA) targeting FFAR2 or the FFAR2 pathway agonists 2-(4-chlorophenyl)-3-methyl-N-(thiazol-2-yl)butanamide (4-CMTB) and compound 58 (Cmp58) [(S)-2-(4-chlorophenyl)-3,3-dimethyl-N-(5-phenylthiazol-2-yl)butanamide] dramatically inhibited the nuclear accumulation of viral nucleoprotein (NP) at early time points postinfection, indicating that FFAR2 functions in the early stage of the IAV replication cycle. FFAR2 downregulation had no effect on the expression of sialic acid (SA) receptors on the cell membrane, the attachment of IAV to the SA receptors, or the activity of the viral ribonucleoprotein (vRNP) complex. Rather, the amount of internalized IAVs was significantly reduced in FFAR2-knocked-down or 4-CMTB- or Cmp58-treated A549 cells. Further studies showed that FFAR2 associated with ß-arrestin1 and that ß-arrestin1 interacted with the ß2-subunit of the AP-2 complex (AP2B1), the essential adaptor of the clathrin-mediated endocytosis pathway. Notably, siRNA knockdown of either ß-arrestin1 or AP2B1 dramatically impaired IAV replication, and AP2B1 knockdown or treatment with Barbadin, an inhibitor targeting the ß-arrestin1/AP2B1 complex, remarkably decreased the amount of internalized IAVs. Moreover, we found that FFAR2 interacted with three G protein-coupled receptor (GPCR) kinases (i.e., GRK2, GRK5, and GRK6) whose downregulation inhibited IAV replication. Together, our findings demonstrate that the FFAR2 signaling cascade is important for the efficient endocytosis of IAV into host cells.IMPORTANCE To complete its replication cycle, IAV hijacks the host endocytosis machinery to invade cells. However, the underlying mechanisms of how IAV is internalized into host cells remain poorly understood, emphasizing the need to elucidate the role of host factors in IAV entry into cells. In this study, we identified FFAR2 as an important host factor for the efficient replication of both low-pathogenic and highly pathogenic IAV. We revealed that FFAR2 facilitates the internalization of IAV into target cells during the early stage of infection. Upon further characterization of the role of FFAR2-associated proteins in virus replication, we found that the FFAR2-ß-arrestin1-AP2B1 signaling cascade is important for the efficient endocytosis of IAV. Our findings thus further our understanding of the biological details of IAV entry into host cells and establish FFAR2 as a potential target for antiviral drug development.


Subject(s)
Endocytosis , Influenza A virus/physiology , Receptors, G-Protein-Coupled/metabolism , Signal Transduction , Virus Internalization , A549 Cells , Adaptor Protein Complex beta Subunits/genetics , Adaptor Protein Complex beta Subunits/metabolism , Animals , Dogs , HEK293 Cells , Humans , Madin Darby Canine Kidney Cells , Mice , RAW 264.7 Cells , Receptors, G-Protein-Coupled/genetics , Virus Replication/physiology , beta-Arrestin 1/genetics , beta-Arrestin 1/metabolism
6.
Respir Res ; 22(1): 49, 2021 Feb 08.
Article in English | MEDLINE | ID: mdl-33557836

ABSTRACT

BACKGROUND: Mitochondrial dysfunction has emerged as an important player in the pathogenesis of idiopathic pulmonary fibrosis (IPF), a common cause of idiopathic interstitial lung disease in adults. Hermansky-Pudlak syndrome (HPS) is a rare autosomal recessive disorder that causes a similar type of pulmonary fibrosis in younger adults, although the role of mitochondrial dysfunction in this condition is not understood. METHODS: We performed a detailed characterization of mitochondrial structure and function in lung tissues and alveolar epithelial cells deficient in the adaptor protein complex 3 beta 1 (Ap3b1) subunit, the gene responsible for causing subtype 2 of HPS (HPS-2). RESULTS: We observed widespread changes in mitochondrial homeostasis in HPS-2 cells, including the acquisition of abnormally shaped mitochondria, with reduced number of cristae, and markedly reduced activity of the electron transport chain and the tricarboxylic acid cycle. We also found that mitochondrial redox imbalance and activity of the mitochondrial unfolded protein response were dysregulated in HPS-2 cells and this associated with various other changes that appeared to be compensatory to mitochondrial dysfunction. This included an increase in glycolytic activity, an upregulation in the expression of mitochondrial biogenesis factors and enhanced activation of the energy-conserving enzyme AMP-activated protein kinase. CONCLUSION: In summary, our findings indicate that mitochondrial function is dramatically altered in HPS-2 lung tissues, suggesting dysfunction of this organelle might be a driver of HPS lung disease.


Subject(s)
Adaptor Protein Complex 3/genetics , Adaptor Protein Complex beta Subunits/genetics , Homeostasis/physiology , Lung/physiopathology , Mitochondria/physiology , Pulmonary Alveoli/physiopathology , Respiratory Mucosa/physiopathology , Animals , Hermanski-Pudlak Syndrome/genetics , Hermanski-Pudlak Syndrome/pathology , Hermanski-Pudlak Syndrome/physiopathology , Lung/pathology , Mice , Mice, Inbred C57BL , Pulmonary Alveoli/pathology , Respiratory Mucosa/pathology
7.
Am J Med Genet A ; 185(10): 3092-3098, 2021 10.
Article in English | MEDLINE | ID: mdl-34042275

ABSTRACT

Angelman syndrome is a neurodevelopmental disorder characterized by intellectual disability (ID), a distinctive gait pattern, abnormal behaviors, severe impairment in language development, and characteristic facial features. Most cases are caused by the absence of a maternal contribution to the imprinted region on chromosome 15q11-q13. Here, we present the first reported case of a 3-year-old boy with an atypical phenotype of Angelman syndrome due to uniparental isodisomy with two recessive homozygous pathogenic variants: in HERC2 and AP3B2. Known phenotypes related to HERC2 and AP3B2 include ID and early infantile epileptic encephalopathy, respectively. The patient had severe global developmental delay and profound ID and showed a happy demeanor, stereotypic laughter, and hand-flapping movements, but also irritability. Craniofacial dysmorphic features, including brachycephaly, strabismus, wide ala nasi, short philtrum, wide open mouth, and slight hypopigmentation were seen. Progressive microcephaly was noted. Magnetic resonance imaging of the brain showed delayed myelination and cerebral atrophy. Trio whole exome sequencing and CGH-SNP array analysis revealed paternal uniparental isodisomy of chromosome 15 and two coexisting recessive diseases resulting from homozygous HERC2 and AP3B2 pathogenic variants. The pathogenic variant in HERC2 was inherited from his heterozygous-carrier father, and the variant in AP3B2 was de novo. We suppose that these unusual features were the combination of the effect of three concomitant disorders.


Subject(s)
Adaptor Protein Complex 3/genetics , Adaptor Protein Complex beta Subunits/genetics , Angelman Syndrome/genetics , Intellectual Disability/genetics , Ubiquitin-Protein Ligases/genetics , Angelman Syndrome/diagnosis , Angelman Syndrome/pathology , Child, Preschool , Chromosomes, Human, Pair 15/genetics , Genetic Predisposition to Disease , Homozygote , Humans , Intellectual Disability/pathology , Male , Phenotype , Uniparental Disomy/genetics , Exome Sequencing
8.
BMC Med Genet ; 21(1): 51, 2020 03 14.
Article in English | MEDLINE | ID: mdl-32171285

ABSTRACT

INTRODUCTION: The AP4B1 gene encodes a subunit of adaptor protein complex-4 (AP4), a component of intracellular transportation of proteins which plays important roles in neurons. Bi-allelic mutations in AP4B1 cause autosomal recessive spastic paraplegia-47(SPG47). CASE PRESENTATION: Here we present a Chinese patient with spastic tetraplegia, moderate psychomotor development delay and febrile seizures plus. Brain MRIs showed dilated supratentorial ventricle, thin posterior and splenium part of corpus callosum. The patient had little progress through medical treatments and rehabilitating regimens. Whole exome sequencing identified novel compound heterozygous truncating variants c.1207C > T (p.Gln403*) and c.52_53delAC (p.Cys18Glnfs*7) in AP4B1 gene. Causal mutations in AP4B1 have been reported in 29 individuals from 22 families so far, most of which are homozygous mutations. CONCLUSIONS: Our study enriched the genetic and phenotypic spectrum of SPG47. Early discovery, diagnosis and proper treatment on the conditions generally increase chances of improvement on the quality of life for patients.


Subject(s)
Adaptor Protein Complex 4/genetics , Adaptor Protein Complex beta Subunits/genetics , DNA-Binding Proteins/genetics , Psychomotor Disorders/genetics , Quadriplegia/genetics , RNA-Binding Proteins/genetics , Seizures, Febrile/genetics , Asian People , Child , China , Codon, Nonsense , Heterozygote , Humans , Male , Phenotype , Protein Subunits/genetics , Psychomotor Disorders/complications , Quadriplegia/complications , Seizures, Febrile/complications , Exome Sequencing
9.
PLoS Genet ; 13(4): e1006617, 2017 04.
Article in English | MEDLINE | ID: mdl-28369060

ABSTRACT

The Anaplastic Lymphoma Kinase (Alk) receptor tyrosine kinase (RTK) plays a critical role in the specification of founder cells (FCs) in the Drosophila visceral mesoderm (VM) during embryogenesis. Reporter gene and CRISPR/Cas9 deletion analysis reveals enhancer regions in and upstream of the Alk locus that influence tissue-specific expression in the amnioserosa (AS), the VM and the epidermis. By performing high throughput yeast one-hybrid screens (Y1H) with a library of Drosophila transcription factors (TFs) we identify Odd-paired (Opa), the Drosophila homologue of the vertebrate Zic family of TFs, as a novel regulator of embryonic Alk expression. Further characterization identifies evolutionarily conserved Opa-binding cis-regulatory motifs in one of the Alk associated enhancer elements. Employing Alk reporter lines as well as CRISPR/Cas9-mediated removal of regulatory elements in the Alk locus, we show modulation of Alk expression by Opa in the embryonic AS, epidermis and VM. In addition, we identify enhancer elements that integrate input from additional TFs, such as Binou (Bin) and Bagpipe (Bap), to regulate VM expression of Alk in a combinatorial manner. Taken together, our data show that the Opa zinc finger TF is a novel regulator of embryonic Alk expression.


Subject(s)
Drosophila Proteins/genetics , Drosophila melanogaster/embryology , Gene Expression Regulation, Developmental , Homeodomain Proteins/genetics , Receptor Protein-Tyrosine Kinases/genetics , Transcription Factors/genetics , Adaptor Protein Complex 1/genetics , Adaptor Protein Complex 1/metabolism , Adaptor Protein Complex beta Subunits/genetics , Adaptor Protein Complex beta Subunits/metabolism , Anaplastic Lymphoma Kinase , Animals , Animals, Genetically Modified , Binding Sites , CRISPR-Cas Systems , Drosophila Proteins/metabolism , Drosophila melanogaster/genetics , Embryo, Nonmammalian , Enhancer Elements, Genetic , Forkhead Transcription Factors/genetics , Forkhead Transcription Factors/metabolism , Homeodomain Proteins/metabolism , Promoter Regions, Genetic , Receptor Protein-Tyrosine Kinases/metabolism , Transcription Factors/metabolism
10.
Am J Hum Genet ; 99(6): 1368-1376, 2016 Dec 01.
Article in English | MEDLINE | ID: mdl-27889060

ABSTRACT

Early-onset epileptic encephalopathy (EOEE) represents a heterogeneous group of severe disorders characterized by seizures, interictal epileptiform activity with a disorganized electroencephalography background, developmental regression or retardation, and onset before 1 year of age. Among a cohort of 57 individuals with epileptic encephalopathy, we ascertained two unrelated affected individuals with EOEE associated with developmental impairment and autosomal-recessive variants in AP3B2 by means of whole-exome sequencing. The targeted sequencing of AP3B2 in 86 unrelated individuals with EOEE led to the identification of an additional family. We gathered five additional families with eight affected individuals through the Matchmaker Exchange initiative by matching autosomal-recessive mutations in AP3B2. Reverse phenotyping of 12 affected individuals from eight families revealed a homogeneous EOEE phenotype characterized by severe developmental delay, poor visual contact with optic atrophy, and postnatal microcephaly. No spasticity, albinism, or hematological symptoms were reported. AP3B2 encodes the neuron-specific subunit of the AP-3 complex. Autosomal-recessive variations of AP3B1, the ubiquitous isoform, cause Hermansky-Pudlak syndrome type 2. The only isoform for the δ subunit of the AP-3 complex is encoded by AP3D1. Autosomal-recessive mutations in AP3D1 cause a severe disorder cumulating the symptoms of the AP3B1 and AP3B2 defects.


Subject(s)
Adaptor Protein Complex 3/genetics , Adaptor Protein Complex beta Subunits/genetics , Epilepsy/complications , Epilepsy/genetics , Genes, Recessive/genetics , Mutation , Optic Atrophy/complications , Optic Atrophy/genetics , Age of Onset , Child , Child, Preschool , Developmental Disabilities/genetics , Female , Humans , Infant , Infant, Newborn , Male , Microcephaly/genetics , Pedigree , Syndrome
11.
Haematologica ; 104(10): 2091-2099, 2019 10.
Article in English | MEDLINE | ID: mdl-30630984

ABSTRACT

Weibel-Palade bodies are endothelial secretory organelles that contain von Willebrand factor, P-selectin and CD63. Release of von Willebrand factor from Weibel-Palade bodies is crucial for platelet adhesion during primary hemostasis. Endosomal trafficking of proteins like CD63 to Weibel-Palade bodies during maturation is dependent on the adaptor protein complex 3 complex. Mutations in the AP3B1 gene, which encodes the adaptor protein complex 3 ß1 subunit, result in Hermansky-Pudlak syndrome 2, a rare genetic disorder that leads to neutropenia and a mild bleeding diathesis. This is caused by abnormal granule formation in neutrophils and platelets due to defects in trafficking of cargo to secretory organelles. The impact of these defects on the secretory pathway of the endothelium is largely unknown. In this study, we investigated the role of adaptor protein complex 3-dependent mechanisms in trafficking of proteins during Weibel-Palade body maturation in endothelial cells. An ex vivo patient-derived endothelial model of Hermansky-Pudlak syndrome type 2 was established using blood outgrowth endothelial cells that were isolated from a patient with compound heterozygous mutations in AP3B1 Hermansky-Pudlak syndrome type 2 endothelial cells and CRISPR-Cas9-engineered AP3B1-/- endothelial cells contain Weibel-Palade bodies that are entirely devoid of CD63, indicative of disrupted endosomal trafficking. Hermansky-Pudlak syndrome type 2 endothelial cells have impaired Ca2+-mediated and cAMP-mediated exocytosis. Whole proteome analysis revealed that, apart from adaptor protein complex 3 ß1, also the µ1 subunit and the v-SNARE VAMP8 were depleted. Stimulus-induced von Willebrand factor secretion was impaired in CRISPR-Cas9-engineered VAMP8-/-endothelial cells. Our data show that defects in adaptor protein complex 3-dependent maturation of Weibel-Palade bodies impairs exocytosis by affecting the recruitment of VAMP8.


Subject(s)
Adaptor Protein Complex 3 , Adaptor Protein Complex beta Subunits , Endothelial Cells , Exocytosis , Hermanski-Pudlak Syndrome , R-SNARE Proteins/metabolism , Weibel-Palade Bodies , Adaptor Protein Complex 3/genetics , Adaptor Protein Complex 3/metabolism , Adaptor Protein Complex beta Subunits/genetics , Adaptor Protein Complex beta Subunits/metabolism , Calcium Signaling , Cells, Cultured , Endothelial Cells/metabolism , Endothelial Cells/pathology , Hermanski-Pudlak Syndrome/genetics , Hermanski-Pudlak Syndrome/metabolism , Hermanski-Pudlak Syndrome/pathology , Humans , Mutation , Protein Transport , R-SNARE Proteins/genetics , Weibel-Palade Bodies/genetics , Weibel-Palade Bodies/metabolism , Weibel-Palade Bodies/pathology
12.
Mol Biol Rep ; 46(3): 3477-3485, 2019 Jun.
Article in English | MEDLINE | ID: mdl-30847849

ABSTRACT

C3H10T1/2, a mouse mesenchymal stem cell line, is a well-known in vitro model of chondrogenesis that can be easily employed to recapitulate some of the mechanisms intervening in this process. Moreover, these cells can be used to validate the effect of candidate molecules identified by high throughput screening approaches applied to the development of targeted therapy for human disorders in which chondrogenic differentiation may be involved, as in conditions characterized by heterotopic endochondral bone formation. Chondrogenic differentiation of C3H10T1/2 cells can be monitored by applying quantitative polymerase chain reaction (qPCR), one of the most sensitive methods that allows detection of small dynamic changes in gene expression between samples obtained under different experimental conditions. In this work, we have used qPCR to monitor the expression of specific markers during chondrogenic differentiation of C3H10T1/2 cells in micromass cultures. Then we have applied the geNorm approach to identify the most stable reference genes suitable to get a robust normalization of the obtained expression data. Among 12 candidate reference genes (Ap3d1, Csnk2a2, Cdc40, Fbxw2, Fbxo38, Htatsf1, Mon2, Pak1ip1, Zfp91, 18S, ActB, GAPDH) we identified Mon2 and Ap3d1 as the most stable ones during chondrogenesis. ActB, GAPDH and 18S, the most commonly used in the literature, resulted to have an expression level too high compared to the differentiation markers (Sox9, Collagen type 2a1, Collagen type 10a1 and Collagen type 1a1), therefore are actually less recommended for these experimental conditions. In conclusion, we identified nine reference genes that can be equally used to obtain a robust normalization of the gene expression variation during the C3H10T1/2 chondrogenic differentiation.


Subject(s)
Chondrogenesis/genetics , Mesenchymal Stem Cells/cytology , Real-Time Polymerase Chain Reaction/standards , Actins/genetics , Adaptor Protein Complex 3/genetics , Adaptor Protein Complex beta Subunits/genetics , Animals , Cell Differentiation/genetics , Cell Line , Cells, Cultured , Gene Expression Profiling/methods , Gene Expression Profiling/standards , Glyceraldehyde-3-Phosphate Dehydrogenase (Phosphorylating)/genetics , Mice , Mice, Inbred C3H , Proton-Translocating ATPases/genetics , Real-Time Polymerase Chain Reaction/methods , Reference Standards , Transcriptome
13.
Reprod Fertil Dev ; 31(5): 972-982, 2019 Apr.
Article in English | MEDLINE | ID: mdl-30786955

ABSTRACT

Hermansky-Pudlak syndrome (HPS) is an autosomal recessive disorder in humans and mice. Pale ear (ep) and pearl (pe) mice, bearing mutations in the biogenesis of lysosomal organelles complex 3 subunit 1 (Hps1) and adaptor-related protein complex 3, beta 1 subunit (Ap3b1) genes respectively, are mouse models of human HPS Type 1 (HPS1) and Type 2 (HPS2) respectively. In the present study we investigated and compared the reduced fertilities of ep and pe male mice. Both ep and pe males exhibited lower abilities to impregnate C57BL/6J (B6) females, and B6 females mated with ep males produced smaller litters than those mated with pe males. Delayed testis development, reduced sperm count and lower testosterone concentrations were observed in the pe but not ep male mice. However, the reduction in sperm motility was greater in ep than pe males, likely due to the mitochondrial and fibrous sheath abnormalities observed by electron microscopy in the sperm tails of ep males. Together, the results indicate that the Hps1 and Ap3b1 genes play distinct roles in male reproductive system development and spermatogenesis in mice, even though ep and pe males share common phenotypes, including reduced lysosomes in Sertoli cells and dislocated Zn2+ in sperm heads.


Subject(s)
Adaptor Protein Complex 3/metabolism , Adaptor Protein Complex beta Subunits/metabolism , Fertility/physiology , Lysosomes/metabolism , Membrane Proteins/metabolism , Spermatogenesis/physiology , Adaptor Protein Complex 3/genetics , Adaptor Protein Complex beta Subunits/genetics , Animals , Disease Models, Animal , Female , Litter Size , Male , Membrane Proteins/genetics , Mice , Mitochondria/metabolism , Sertoli Cells/metabolism , Spermatozoa/metabolism , Testis/metabolism , Testosterone/blood , Zinc/metabolism
14.
Hum Mutat ; 38(10): 1402-1411, 2017 10.
Article in English | MEDLINE | ID: mdl-28585318

ABSTRACT

Hermansky-Pudlak syndrome type 2 (HPS2) is a syndrome caused by mutations in the beta-3A subunit of the adaptor protein (AP)-3 complex (AP3B1 gene). We describe five unreported cases with four novel mutations, one of which caused aberrant pre-mRNA splicing. A point mutation c.2702C>G in exon 23 of the AP3B1 gene caused deletion of 112 bp in the mRNA in two siblings. This mutation activates a cryptic donor splice site that overrules the wild-type donor splice site of this exon. Three other novel mutations in AP3B1 were identified, that is, a nonsense mutation c.716G>A (p.Trp239Ter), a 1-bp and a 4-bp deletion c.177delA and c.1839_1842delTAGA, respectively, both causing frameshift and premature termination of translation. Mass spectrometry in four of these HPS2 patients demonstrated the (near) absence of all AP-3 complex subunits. Immunoelectron microscopy on the neutrophils of two of these patients showed abnormal granule formation. We found clear mislocalization of myeloperoxidase in the neutrophils even though the content of this protein but not the activity seemed to be present at normal levels. In sum, HPS2 is the result of the absence of the entire AP-3 complex, which results in severe neutropenia with a defect in granule formation as the major hematological finding.


Subject(s)
Adaptor Protein Complex 3/genetics , Adaptor Protein Complex beta Subunits/genetics , Hermanski-Pudlak Syndrome/genetics , RNA Precursors/genetics , RNA Splicing/genetics , Adolescent , Adult , Child , Child, Preschool , Codon, Nonsense/genetics , Exons/genetics , Female , Hermanski-Pudlak Syndrome/physiopathology , Humans , Infant , Male , Middle Aged , Neutrophils/metabolism , Neutrophils/pathology , Phenotype , Point Mutation , RNA Splice Sites/genetics , Sequence Deletion/genetics
16.
J Cell Sci ; 127(Pt 20): 4457-69, 2014 Oct 15.
Article in English | MEDLINE | ID: mdl-25179596

ABSTRACT

Some native epithelia, for example, retinal pigment epithelium (RPE) and kidney proximal tubule (KPT), constitutively lack the basolateral sorting adaptor AP-1B; this results in many basolateral plasma membrane proteins being repositioned to the apical domain, where they perform essential functions for their host organs. We recently reported the underlying apical polarity reversal mechanism: in the absence of AP-1B-mediated basolateral sorting, basolateral proteins are shuttled to the apical plasma membrane through a transcytotic pathway mediated by the plus-end kinesin KIF16B. Here, we demonstrate that this apical transcytotic pathway requires apical sorting of basolateral proteins, which is mediated by apical signals and galectin-4. Using RPE and KPT cell lines, and AP-1B-knockdown MDCK cells, we show that mutation of the N-glycan linked to N727 in the basolateral marker transferrin receptor (TfR) or knockdown of galectin-4 inhibits TfR transcytosis to apical recycling endosomes and the apical plasma membrane, and promotes TfR lysosomal targeting and subsequent degradation. Our results report a new role of galectins in basolateral to apical epithelial transcytosis.


Subject(s)
Adaptor Protein Complex 1/metabolism , Adaptor Protein Complex beta Subunits/metabolism , Cell Membrane/metabolism , Endosomes/metabolism , Epithelial Cells/physiology , Galectin 4/metabolism , Lysosomes/metabolism , Receptors, Transferrin/metabolism , Adaptor Protein Complex 1/genetics , Adaptor Protein Complex beta Subunits/genetics , Animals , Cell Line , Cell Polarity/genetics , Dogs , Galectin 4/genetics , Gene Knockdown Techniques , Humans , Madin Darby Canine Kidney Cells , Mutation/genetics , Protein Sorting Signals/genetics , Protein Transport/genetics , Receptors, Transferrin/genetics , Transcytosis/genetics
17.
Mol Vis ; 22: 783-96, 2016.
Article in English | MEDLINE | ID: mdl-27440996

ABSTRACT

PURPOSE: Ocular refraction is measured in spherical equivalent as the power of the external lens required to focus images on the retina. Myopia (nearsightedness) and hyperopia (farsightedness) are the most common refractive errors, and the leading causes of visual impairment and blindness in the world. The goal of this study is to identify rare and low-frequency variants that influence spherical equivalent. METHODS: We conducted variant-level and gene-level quantitative trait association analyses for mean spherical equivalent, using data from 1,560 individuals in the Beaver Dam Eye Study. Genotyping was conducted using the Illumina exome array. We analyzed 34,976 single nucleotide variants and 11,571 autosomal genes across the genome, using single-variant tests as well as gene-based tests. RESULTS: Spherical equivalent was significantly associated with five genes in gene-based analysis: PTCHD2 at 1p36.22 (p = 3.6 × 10(-7)), CRISP3 at 6p12.3 (p = 4.3 × 10(-6)), NAP1L4 at 11p15.5 (p = 3.6 × 10(-6)), FSCB at 14q21.2 (p = 1.5 × 10(-7)), and AP3B2 at 15q25.2 (p = 1.6 × 10(-7)). The variant-based tests identified evidence suggestive of association with two novel variants in linkage disequilibrium (pairwise r(2) = 0.80) in the TCTE1 gene region at 6p21.1 (rs2297336, minor allele frequency (MAF) = 14.1%, ß = -0.62 p = 3.7 × 10(-6); rs324146, MAF = 16.9%, ß = -0.55, p = 1.4 × 10(-5)). In addition to these novel findings, we successfully replicated a previously reported association with rs634990 near GJD2 at 15q14 (MAF = 47%, ß = -0.29, p=1.8 × 10(-3)). We also found evidence of association with spherical equivalent on 2q37.1 in PRSS56 at rs1550094 (MAF = 31%, ß = -0.33, p = 1.7 × 10(-3)), a region previously associated with myopia. CONCLUSIONS: We identified several novel candidate genes that may play a role in the control of spherical equivalent. However, further studies are needed to replicate these findings. In addition, our results contribute to the increasing evidence that variation in the GJD2 and PRSS56 genes influence the development of refractive errors. Identifying that variation in these genes is associated with spherical equivalent may provide further insight into the etiology of myopia and consequent vision loss.


Subject(s)
Adaptor Protein Complex 3/genetics , Adaptor Protein Complex beta Subunits/genetics , Calcium-Binding Proteins/genetics , Membrane Proteins/genetics , Myopia/genetics , Nuclear Proteins/genetics , Polymorphism, Single Nucleotide , Salivary Proteins and Peptides/genetics , Seminal Plasma Proteins/genetics , Adult , Aged , Aged, 80 and over , Exome/genetics , Eye Proteins/genetics , Female , Genetic Predisposition to Disease , Genome-Wide Association Study , Genotyping Techniques , Humans , Linkage Disequilibrium , Male , Middle Aged , Phenotype
20.
PLoS Genet ; 9(9): e1003812, 2013.
Article in English | MEDLINE | ID: mdl-24086151

ABSTRACT

The regulated secretion of peptide hormones, neural peptides and many growth factors depends on their sorting into large dense core vesicles (LDCVs) capable of regulated exocytosis. LDCVs form at the trans-Golgi network, but the mechanisms that sort proteins to this regulated secretory pathway and the cytosolic machinery that produces LDCVs remain poorly understood. Recently, we used an RNAi screen to identify a role for heterotetrameric adaptor protein AP-3 in regulated secretion and in particular, LDCV formation. Indeed, mocha mice lacking AP-3 have a severe neurological and behavioral phenotype, but this has been attributed to a role for AP-3 in the endolysosomal rather than biosynthetic pathway. We therefore used mocha mice to determine whether loss of AP-3 also dysregulates peptide release in vivo. We find that adrenal chromaffin cells from mocha animals show increased constitutive exocytosis of both soluble cargo and LDCV membrane proteins, reducing the response to stimulation. We also observe increased basal release of both insulin and glucagon from pancreatic islet cells of mocha mice, suggesting a global disturbance in the release of peptide hormones. AP-3 exists as both ubiquitous and neuronal isoforms, but the analysis of mice lacking each of these isoforms individually and together shows that loss of both is required to reproduce the effect of the mocha mutation on the regulated pathway. In addition, we show that loss of the related adaptor protein AP-1 has a similar effect on regulated secretion but exacerbates the effect of AP-3 RNAi, suggesting distinct roles for the two adaptors in the regulated secretory pathway.


Subject(s)
Adaptor Protein Complex 3/genetics , Adaptor Protein Complex beta Subunits/genetics , Cytosol/metabolism , Exocytosis/genetics , Peptide Hormones/metabolism , Adaptor Protein Complex 3/metabolism , Adaptor Protein Complex beta Subunits/metabolism , Animals , Glucagon/genetics , Glucagon/metabolism , Insulin/genetics , Insulin/metabolism , Insulin Secretion , Islets of Langerhans/cytology , Islets of Langerhans/metabolism , Lysosomes , Metabolic Networks and Pathways , Mice , Neurons/metabolism , RNA Interference , Transcription Factor AP-1/genetics
SELECTION OF CITATIONS
SEARCH DETAIL