Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
1.
J Bone Miner Res ; 38(9): 1350-1363, 2023 09.
Article in English | MEDLINE | ID: mdl-37436066

ABSTRACT

Genome-wide association studies (GWASs) have advanced our understanding of the genetics of osteoporosis; however, the challenge has been converting associations to causal genes. Studies have utilized transcriptomics data to link disease-associated variants to genes, but few population transcriptomics data sets have been generated on bone at the single-cell level. To address this challenge, we profiled the transcriptomes of bone marrow-derived stromal cells (BMSCs) cultured under osteogenic conditions from five diversity outbred (DO) mice using single-cell RNA-seq (scRNA-seq). The goal of the study was to determine if BMSCs could serve as a model to generate cell type-specific transcriptomic profiles of mesenchymal lineage cells from large populations of mice to inform genetic studies. By enriching for mesenchymal lineage cells in vitro, coupled with pooling of multiple samples and downstream genotype deconvolution, we demonstrate the scalability of this model for population-level studies. We demonstrate that dissociation of BMSCs from a heavily mineralized matrix had little effect on viability or their transcriptomic signatures. Furthermore, we show that BMSCs cultured under osteogenic conditions are diverse and consist of cells with characteristics of mesenchymal progenitors, marrow adipogenic lineage precursors (MALPs), osteoblasts, osteocyte-like cells, and immune cells. Importantly, all cells were similar from a transcriptomic perspective to cells isolated in vivo. We employed scRNA-seq analytical tools to confirm the biological identity of profiled cell types. SCENIC was used to reconstruct gene regulatory networks (GRNs), and we observed that cell types show GRNs expected of osteogenic and pre-adipogenic lineage cells. Further, CELLECT analysis showed that osteoblasts, osteocyte-like cells, and MALPs captured a significant component of bone mineral density (BMD) heritability. Together, these data suggest that BMSCs cultured under osteogenic conditions coupled with scRNA-seq can be used as a scalable and biologically informative model to generate cell type-specific transcriptomic profiles of mesenchymal lineage cells in large populations. © 2023 The Authors. Journal of Bone and Mineral Research published by Wiley Periodicals LLC on behalf of American Society for Bone and Mineral Research (ASBMR).


Subject(s)
Collaborative Cross Mice , Mesenchymal Stem Cells , Mice , Animals , Collaborative Cross Mice/genetics , Cell Differentiation/genetics , Transcriptome/genetics , Genome-Wide Association Study , Single-Cell Gene Expression Analysis , Cells, Cultured , Mesenchymal Stem Cells/metabolism , Osteogenesis/genetics , Stromal Cells/metabolism , Bone Marrow Cells
2.
Nat Commun ; 12(1): 3408, 2021 06 07.
Article in English | MEDLINE | ID: mdl-34099702

ABSTRACT

Genome-wide association studies (GWASs) for osteoporotic traits have identified over 1000 associations; however, their impact has been limited by the difficulties of causal gene identification and a strict focus on bone mineral density (BMD). Here, we use Diversity Outbred (DO) mice to directly address these limitations by performing a systems genetics analysis of 55 complex skeletal phenotypes. We apply a network approach to cortical bone RNA-seq data to discover 66 genes likely to be causal for human BMD GWAS associations, including the genes SERTAD4 and GLT8D2. We also perform GWAS in the DO for a wide-range of bone traits and identify Qsox1 as a gene influencing cortical bone accrual and bone strength. In this work, we advance our understanding of the genetics of osteoporosis and highlight the ability of the mouse to inform human genetics.


Subject(s)
Bone Density/genetics , Osteoporosis/genetics , Oxidoreductases Acting on Sulfur Group Donors/genetics , Animals , Cell Differentiation/genetics , Collaborative Cross Mice , Datasets as Topic , Female , Femur/physiology , Fluoresceins/administration & dosage , Fluorescent Dyes/administration & dosage , Genome-Wide Association Study , Glycosyltransferases/genetics , Humans , Male , Mesenchymal Stem Cells , Mice , Mice, Knockout , Osteoblasts , Osteogenesis/genetics , RNA-Seq , Single-Cell Analysis
3.
J Bone Miner Res ; 36(8): 1566-1579, 2021 08.
Article in English | MEDLINE | ID: mdl-33900658

ABSTRACT

Bone metastasis is a complication of prostate cancer in up to 90% of men afflicted with advanced disease. Therapies that reduce androgen exposure remain at the forefront of treatment. However, most prostate cancers transition to a state whereby reducing testicular androgen action becomes ineffective. A common mechanism of this transition is intratumoral production of testosterone (T) using the adrenal androgen precursor dehydroepiandrosterone (DHEA) through enzymatic conversion by 3ß- and 17ß-hydroxysteroid dehydrogenases (3ßHSD and 17ßHSD). Given the ability of prostate cancer to form blastic metastases in bone, we hypothesized that osteoblasts might be a source of androgen synthesis. RNA expression analyses of murine osteoblasts and human bone confirmed that at least one 3ßHSD and 17ßHSD enzyme isoform was expressed, suggesting that osteoblasts are capable of generating androgens from adrenal DHEA. Murine osteoblasts were treated with 100 nM and 1 µM DHEA or vehicle control. Conditioned media from these osteoblasts were assayed for intermediate and active androgens by liquid chromatography-tandem mass spectrometry. As DHEA was consumed, the androgen intermediates androstenediol and androstenedione were generated and subsequently converted to T. Conditioned media of DHEA-treated osteoblasts increased androgen receptor (AR) signaling, prostate-specific antigen (PSA) production, and cell numbers of the androgen-sensitive prostate cancer cell lines C4-2B and LNCaP. DHEA did not induce AR signaling in osteoblasts despite AR expression in this cell type. We describe an unreported function of osteoblasts as a source of T that is especially relevant during androgen-responsive metastatic prostate cancer invasion into bone. © 2021 American Society for Bone and Mineral Research (ASBMR). This article has been contributed to by US Government employees and their work is in the public domain in the USA.


Subject(s)
Androgens , Prostatic Neoplasms , Animals , Cell Line, Tumor , Dehydroepiandrosterone , Humans , Male , Mice , Osteoblasts , Receptors, Androgen , Testosterone
4.
Blood ; 138(8): 662-673, 2021 08 26.
Article in English | MEDLINE | ID: mdl-33786584

ABSTRACT

Chronic natural killer large granular lymphocyte (NK-LGL) leukemia, also referred to as chronic lymphoproliferative disorder of NK cells, is a rare disorder defined by prolonged expansion of clonal NK cells. Similar prevalence of STAT3 mutations in chronic T-LGL and NK-LGL leukemia is suggestive of common pathogenesis. We undertook whole-genome sequencing to identify mutations unique to NK-LGL leukemia. The results were analyzed to develop a resequencing panel that was applied to 58 patients. Phosphatidylinositol 3-kinase pathway gene mutations (PIK3CD/PIK3AP1) and TNFAIP3 mutations were seen in 5% and 10% of patients, respectively. TET2 was exceptional in that mutations were present in 16 (28%) of 58 patient samples, with evidence that TET2 mutations can be dominant and exclusive to the NK compartment. Reduced-representation bisulfite sequencing revealed that methylation patterns were significantly altered in TET2 mutant samples. The promoter of TET2 and that of PTPRD, a negative regulator of STAT3, were found to be methylated in additional cohort samples, largely confined to the TET2 mutant group. Mutations in STAT3 were observed in 19 (33%) of 58 patient samples, 7 of which had concurrent TET2 mutations. Thrombocytopenia and resistance to immunosuppressive agents were uniquely observed in those patients with only TET2 mutation (Games-Howell post hoc test, P = .0074; Fisher's exact test, P = .00466). Patients with STAT3 mutation, inclusive of those with TET2 comutation, had lower hematocrit, hemoglobin, and absolute neutrophil count compared with STAT3 wild-type patients (Welch's t test, P ≤ .015). We present the discovery of TET2 mutations in chronic NK-LGL leukemia and evidence that it identifies a unique molecular subtype.


Subject(s)
DNA-Binding Proteins/genetics , Dioxygenases/genetics , Leukemia, Large Granular Lymphocytic/genetics , Mutation , Neoplasm Proteins/genetics , Registries , Chronic Disease , DNA-Binding Proteins/blood , Dioxygenases/blood , Female , Humans , Leukemia, Large Granular Lymphocytic/blood , Male , Neoplasm Proteins/blood
5.
J Neurosci ; 37(48): 11744-11757, 2017 11 29.
Article in English | MEDLINE | ID: mdl-29066557

ABSTRACT

The retrotrapezoid nucleus (RTN) consists, by definition, of Phox2b-expressing, glutamatergic, non-catecholaminergic, noncholinergic neurons located in the parafacial region of the medulla oblongata. An unknown proportion of RTN neurons are central respiratory chemoreceptors and there is mounting evidence for biochemical diversity among these cells. Here, we used multiplexed in situ hybridization and single-cell RNA-Seq in male and female mice to provide a more comprehensive view of the phenotypic diversity of RTN neurons. We now demonstrate that the RTN of mice can be identified with a single and specific marker, Neuromedin B mRNA (Nmb). Most (∼75%) RTN neurons express low-to-moderate levels of Nmb and display chemoreceptor properties. Namely they are activated by hypercapnia, but not by hypoxia, and express proton sensors, TASK-2 and Gpr4. These Nmb-low RTN neurons also express varying levels of transcripts for Gal, Penk, and Adcyap1, and receptors for substance P, orexin, serotonin, and ATP. A subset of RTN neurons (∼20-25%), typically larger than average, express very high levels of Nmb mRNA. These Nmb-high RTN neurons do not express Fos after hypercapnia and have low-to-undetectable levels of Kcnk5 or Gpr4 transcripts; they also express Adcyap1, but are essentially devoid of Penk and Gal transcripts. In male rats, Nmb is also a marker of the RTN but, unlike in mice, this gene is expressed by other types of nearby neurons located within the ventromedial medulla. In sum, Nmb is a selective marker of the RTN in rodents; Nmb-low neurons, the vast majority, are central respiratory chemoreceptors, whereas Nmb-high neurons likely have other functions.SIGNIFICANCE STATEMENT Central respiratory chemoreceptors regulate arterial PCO2 by adjusting lung ventilation. Such cells have recently been identified within the retrotrapezoid nucleus (RTN), a brainstem nucleus defined by genetic lineage and a cumbersome combination of markers. Using single-cell RNA-Seq and multiplexed in situ hybridization, we show here that a single marker, Neuromedin B mRNA (Nmb), identifies RTN neurons in rodents. We also suggest that >75% of these Nmb neurons are chemoreceptors because they are strongly activated by hypercapnia and express high levels of proton sensors (Kcnk5 and Gpr4). The other RTN neurons express very high levels of Nmb, but low levels of Kcnk5/Gpr4/pre-pro-galanin/pre-pro-enkephalin, and do not respond to hypercapnia. Their function is unknown.


Subject(s)
Medulla Oblongata/metabolism , Neurokinin B/analogs & derivatives , Animals , Female , Gene Expression , Hypoxia/genetics , Hypoxia/metabolism , Male , Medulla Oblongata/chemistry , Mice , Mice, Inbred C57BL , Mice, Transgenic , Neurokinin B/analysis , Neurokinin B/biosynthesis , Neurokinin B/genetics , Neurons/chemistry , Neurons/metabolism , Organ Culture Techniques , Rats , Rats, Sprague-Dawley
6.
Respir Res ; 18(1): 97, 2017 05 18.
Article in English | MEDLINE | ID: mdl-28521775

ABSTRACT

BACKGROUND: We conducted a genome-wide association study (GWAS) of subclinical interstitial lung disease (ILD), defined as high attenuation areas (HAA) on CT, in the population-based Multi-Ethnic Study of Atherosclerosis Study. METHODS: We measured the percentage of high attenuation areas (HAA) in the lung fields on cardiac CT scan defined as voxels with CT attenuation values between -600 and -250 HU. Genetic analyses were performed in MESA combined across race/ethnic groups: non-Hispanic White (n = 2,434), African American (n = 2,470), Hispanic (n = 2,065) and Chinese (n = 702), as well as stratified by race/ethnicity. RESULTS: Among 7,671 participants, regions at genome-wide significance were identified for basilar peel-core ratio of HAA in FLJ35282 downstream of ANRIL (rs7852363, P = 2.1x10-9) and within introns of SNAI3-AS1 (rs140142658, P = 9.6x10-9) and D21S2088E (rs3079677, P = 2.3x10-8). Within race/ethnic groups, 18 additional loci were identified at genome-wide significance, including genes related to development (FOXP4), cell adhesion (ALCAM) and glycosylation (GNPDA2, GYPC, GFPT1 and FUT10). Among these loci, SNP rs6844387 near GNPDA2 demonstrated nominal evidence of replication in analysis of n = 1,959 participants from the Framingham Heart Study (P = 0.029). FOXP4 region SNP rs2894439 demonstrated evidence of validation in analysis of n = 228 White ILD cases from the Columbia ILD Study compared to race/ethnicity-matched controls from MESA (one-sided P = 0.007). In lung tissue from 15 adults with idiopathic pulmonary fibrosis compared to 15 adults without lung disease. ANRIL (P = 0.001), ALCAM (P = 0.03) and FOXP4 (P = 0.046) were differentially expressed. CONCLUSIONS: Our results suggest novel roles for protein glycosylation and cell cycle disinhibition by long non-coding RNA in the pathogenesis of ILD.


Subject(s)
Asian People/genetics , Black or African American/genetics , Genome-Wide Association Study/methods , Hispanic or Latino/genetics , Lung Diseases, Interstitial/genetics , White People/genetics , Adult , Aged , Aged, 80 and over , Female , Humans , Longitudinal Studies , Lung Diseases, Interstitial/diagnosis , Male , Middle Aged , Polymorphism, Single Nucleotide/genetics , Population Surveillance/methods
7.
Ann Am Thorac Soc ; 14(5): 628-635, 2017 May.
Article in English | MEDLINE | ID: mdl-28248552

ABSTRACT

RATIONALE: The receptor for advanced glycation end products (RAGE) is underexpressed in idiopathic pulmonary fibrosis (IPF) lung, but the role of RAGE in human lung fibrosis remains uncertain. OBJECTIVES: To examine (1) the association between IPF risk and variation at rs2070600, a functional missense variant in AGER (the gene that codes for RAGE), and (2) the associations between plasma-soluble RAGE (sRAGE) levels with disease severity and time to death or lung transplant in IPF. METHODS: We genotyped the rs2070600 single-nucleotide polymorphism in 108 adults with IPF and 324 race-/ethnicity-matched control subjects. We measured plasma sRAGE by ELISA in 103 adults with IPF. We used generalized linear and additive models as well as Cox models to control for potential confounders. We repeated our analyses in 168 (genetic analyses) and 177 (sRAGE analyses) adults with other forms of interstitial lung disease (ILD). RESULTS: There was no association between rs2070600 variation among adults with IPF (P = 0.31). Plasma sRAGE levels were lower among adults with IPF and other forms of ILD than in control subjects (P < 0.001). The rs2070600 allele A was associated with a 49% lower sRAGE level (95% confidence interval [CI], 11 to 71%; P = 0.02) among adults with IPF. In adjusted analyses, lower sRAGE levels were associated with greater disease severity (14% sRAGE decrement per 10% FVC decrement; 95% CI, 5 to 22%) and a higher rate of death or lung transplant at 1 year (adjusted hazard ratio, 1.9 per logarithmic unit of sRAGE decrement; 95% CI, 1.2-3.3) in IPF. Similar findings were observed in a heterogeneous group of adults with other forms of ILD. CONCLUSIONS: Lower plasma sRAGE levels may be a biological measure of disease severity in IPF. Variation at the rs2070600 single-nucleotide polymorphism was not associated with IPF risk.


Subject(s)
Idiopathic Pulmonary Fibrosis/blood , Idiopathic Pulmonary Fibrosis/genetics , Receptor for Advanced Glycation End Products/blood , Receptor for Advanced Glycation End Products/genetics , Aged , Aged, 80 and over , Case-Control Studies , Disease Progression , Female , Genotype , Humans , Idiopathic Pulmonary Fibrosis/surgery , Lung Transplantation , Male , Middle Aged , Polymorphism, Single Nucleotide , Proportional Hazards Models , Prospective Studies , Solubility , United States
8.
Cell Metab ; 19(4): 667-81, 2014 Apr 01.
Article in English | MEDLINE | ID: mdl-24703698

ABSTRACT

Early in the pathogenesis of type 2 diabetes mellitus (T2DM), dysregulated glucagon secretion from pancreatic α cells occurs prior to impaired glucose-stimulated insulin secretion (GSIS) from ß cells. However, whether hyperglucagonemia is causally linked to ß cell dysfunction remains unclear. Here we show that glucagon stimulates via cAMP-PKA-CREB signaling hepatic production of the neuropeptide kisspeptin1, which acts on ß cells to suppress GSIS. Synthetic kisspeptin suppresses GSIS in vivo in mice and from isolated islets in a kisspeptin1 receptor-dependent manner. Kisspeptin1 is increased in livers and in serum from humans with T2DM and from mouse models of diabetes mellitus. Importantly, liver Kiss1 knockdown in hyperglucagonemic, glucose-intolerant, high-fat-diet fed, and Lepr(db/db) mice augments GSIS and improves glucose tolerance. These observations indicate a hormonal circuit between the liver and the endocrine pancreas in glycemia regulation and suggest in T2DM a sequential link between hyperglucagonemia via hepatic kisspeptin1 to impaired insulin secretion.


Subject(s)
Gene Expression Regulation/physiology , Glucagon/metabolism , Gluconeogenesis/physiology , Insulin/metabolism , Kisspeptins/metabolism , Liver/metabolism , Animals , Gene Expression Regulation/drug effects , Gene Knockdown Techniques , Glucagon/pharmacology , Gluconeogenesis/drug effects , Immunohistochemistry , Insulin Secretion , Kisspeptins/blood , Kisspeptins/genetics , Luciferases , Mice , Mice, Inbred NOD , Models, Biological
9.
J Transl Med ; 9: 91, 2011 Jun 10.
Article in English | MEDLINE | ID: mdl-21663644

ABSTRACT

BACKGROUND: Blood specimen collection at an early study visit is often included in observational studies or clinical trials for analysis of secondary outcome biomarkers. A common protocol is to store buffy coat specimens for future DNA isolation and these may remain in frozen storage for many years. It is uncertain if the DNA remains suitable for modern genome wide association (GWA) genotyping. METHODS: We isolated DNA from 120 Action to Control Cardiovascular Risk in Diabetes (ACCORD) clinical trial buffy coats sampling a range of storage times up to 9 years and other factors that could influence DNA yield. We performed TaqMan SNP and GWA genotyping to test whether the DNA retained integrity for high quality genetic analysis. RESULTS: We tested two QIAGEN automated protocols for DNA isolation, preferring the Compromised Blood Protocol despite similar yields. We isolated DNA from all 120 specimens (yield range 1.1-312 ug per 8.5 ml ACD tube of whole blood) with only 3/120 samples yielding < 10 ug DNA. Age of participant at blood draw was negatively associated with yield (mean change -2.1 ug/year). DNA quality was very good based on gel electrophoresis QC, TaqMan genotyping of 6 SNPs (genotyping no-call rate 1.1% in 702 genotypes), and excellent quality GWA genotyping data (maximum per sample genotype missing rate 0.64%). CONCLUSIONS: When collected as a long term clinical trial or biobank specimen for DNA, buffy coats can be stored for up to 9 years in a -80°C frozen state and still produce high yields of DNA suitable for GWA analysis and other genetic testing.


Subject(s)
Blood Buffy Coat/metabolism , Blood Specimen Collection/methods , DNA/genetics , Genetic Testing , Genome, Human/genetics , Genome-Wide Association Study , Tissue Preservation/methods , Adult , Aged , Aged, 80 and over , Female , Genotype , Humans , Linear Models , Male , Middle Aged , Polymorphism, Single Nucleotide/genetics , Quality Control , Time Factors
10.
J Biol Chem ; 285(27): 20818-26, 2010 Jul 02.
Article in English | MEDLINE | ID: mdl-20439468

ABSTRACT

Lamin A, a key component of the nuclear lamina, is generated from prelamin A by four post-translational processing steps: farnesylation, endoproteolytic release of the last three amino acids of the protein, methylation of the C-terminal farnesylcysteine, and finally, endoproteolytic release of the last 15 amino acids of the protein (including the farnesylcysteine methyl ester). The last cleavage step, mediated by ZMPSTE24, releases mature lamin A. This processing scheme has been conserved through vertebrate evolution and is widely assumed to be crucial for targeting lamin A to the nuclear envelope. However, its physiologic importance has never been tested. To address this issue, we created mice with a "mature lamin A-only" allele (Lmna(LAO)), which contains a stop codon immediately after the last codon of mature lamin A. Thus, Lmna(LAO/LAO) mice synthesize mature lamin A directly, bypassing prelamin A synthesis and processing. The levels of mature lamin A in Lmna(LAO/LAO) mice were indistinguishable from those in "prelamin A-only" mice (Lmna(PLAO/PLAO)), where all of the lamin A is produced from prelamin A. Lmna(LAO/LAO) exhibited normal body weights and had no detectable disease phenotypes. A higher frequency of nuclear blebs was observed in Lmna(LAO/LAO) embryonic fibroblasts; however, the mature lamin A in the tissues of Lmna(LAO/LAO) mice was positioned normally at the nuclear rim. We conclude that prelamin A processing is dispensable in mice and that direct synthesis of mature lamin A has little if any effect on the targeting of lamin A to the nuclear rim in mouse tissues.


Subject(s)
Cell Nucleus/pathology , Fibroblasts/pathology , Lamin Type A/biosynthesis , Animals , Blotting, Western , Conserved Sequence , Crosses, Genetic , Embryo, Mammalian , Fibroblasts/cytology , Fibroblasts/metabolism , Introns , Lamin Type A/genetics , Methylation , Mice , Mice, Knockout , Microscopy, Fluorescence , Mutagenesis, Site-Directed , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Phenotype , Protein Modification, Translational , Protein Precursors/genetics , Protein Precursors/metabolism , Vertebrates
11.
Proc Natl Acad Sci U S A ; 107(11): 5076-81, 2010 Mar 16.
Article in English | MEDLINE | ID: mdl-20145110

ABSTRACT

Nuclear lamins are components of the nuclear lamina, a structural scaffolding for the cell nucleus. Defects in lamins A and C cause an array of human diseases, including muscular dystrophy, lipodystrophy, and progeria, but no diseases have been linked to the loss of lamins B1 or B2. To explore the functional relevance of lamin B2, we generated lamin B2-deficient mice and found that they have severe brain abnormalities resembling lissencephaly, with abnormal layering of neurons in the cerebral cortex and cerebellum. This neuronal layering abnormality is due to defective neuronal migration, a process that is dependent on the organized movement of the nucleus within the cell. These studies establish an essential function for lamin B2 in neuronal migration and brain development.


Subject(s)
Cerebellum/abnormalities , Cerebellum/embryology , Cerebral Cortex/abnormalities , Cerebral Cortex/embryology , Lamin Type B/deficiency , Animals , Cell Movement , Cerebellum/pathology , Cerebral Cortex/pathology , Gene Silencing , Lamin Type B/metabolism , Mice , Neurons/pathology
12.
Physiol Genomics ; 37(3): 294-302, 2009 May 13.
Article in English | MEDLINE | ID: mdl-19336533

ABSTRACT

HG.CAST-(D9Mit249-D9Mit133) (HG9) congenic mice are homozygous for CAST/EiJ chromosome (Chr) 9 alleles from approximately 9 to 84 Mbp on a C57BL6/J-hg/hg (HG) background. This region contains the carcass fat in high growth mice (Carfhg2) quantitative trait locus (QTL), and while its obesity-promoting effects have been confirmed in HG9 mice, its underlying genetic basis remains elusive. To refine the location of Carfhg2, we preformed a linkage analysis in two congenic F2 intercrosses and progeny-tested a recombinant F2 male. These analyses narrowed Carfhg2 to between 33.0 and 40.8 Mbp on Chr 9. To identify candidate genes we measured the expression of 44 transcripts surrounding the Carfhg2 peak in adipose, brain, liver, and muscle tissues from F2 mice using Biomark 48.48 Dynamic Arrays. In total, 68% (30 of the 44) of genes were regulated by a significant expression QTL (eQTL) in at least one tissue. To prioritize genes with eQTL we used Network Edge Orienting, a causality modeling tool. These analyses advance our goal of identifying the molecular basis of Carfhg2.


Subject(s)
Chromosome Mapping/methods , Chromosomes, Mammalian/genetics , Obesity/genetics , Quantitative Trait Loci/genetics , Adiposity/genetics , Analysis of Variance , Animals , Crosses, Genetic , DNA, Complementary/chemistry , DNA, Complementary/genetics , Female , Gene Expression Profiling , Genetic Predisposition to Disease/genetics , Lod Score , Male , Mice , Mice, Congenic , Mice, Inbred C57BL , Molecular Sequence Data , Reverse Transcriptase Polymerase Chain Reaction , Sequence Analysis, DNA
13.
Hum Mol Genet ; 18(13): 2462-71, 2009 Jul 01.
Article in English | MEDLINE | ID: mdl-19376814

ABSTRACT

Hutchinson-Gilford progeria syndrome (HGPS) is caused by point mutations that increase utilization of an alternate splice donor site in exon 11 of LMNA (the gene encoding lamin C and prelamin A). The alternate splicing reduces transcripts for wild-type prelamin A and increases transcripts for a truncated prelamin A (progerin). Here, we show that antisense oligonucleotides (ASOs) against exon 11 sequences downstream from the exon 11 splice donor site promote alternate splicing in both wild-type and HGPS fibroblasts, increasing the synthesis of progerin. Indeed, wild-type fibroblasts transfected with these ASOs exhibit progerin levels similar to (or greater than) those in fibroblasts from HGPS patients. This progerin was farnesylated, as judged by metabolic labeling studies. The synthesis of progerin in wild-type fibroblasts was accompanied by the same nuclear shape and gene-expression perturbations observed in HGPS fibroblasts. An ASO corresponding to the 5' portion of intron 11 also promoted alternate splicing. In contrast, an ASO against exon 11 sequences 5' to the alternate splice site reduced alternate splicing in HGPS cells and modestly lowered progerin levels. Thus, different ASOs can be used to increase or decrease 'HGPS splicing'. ASOs represent a new and powerful tool for recreating HGPS pathophysiology in wild-type cells.


Subject(s)
Mutation , Nuclear Proteins/biosynthesis , Nuclear Proteins/genetics , Oligonucleotides, Antisense/therapeutic use , Progeria/genetics , Progeria/therapy , Protein Precursors/biosynthesis , Protein Precursors/genetics , Alternative Splicing , Base Sequence , Cell Line , Cells, Cultured , Exons , Fibroblasts/metabolism , Genetic Therapy , Humans , Lamin Type A , Molecular Sequence Data , Oligonucleotides, Antisense/genetics , Oligonucleotides, Antisense/metabolism , Progeria/metabolism
14.
J Biol Chem ; 283(15): 9797-804, 2008 Apr 11.
Article in English | MEDLINE | ID: mdl-18230615

ABSTRACT

HIV protease inhibitors (HIV-PIs) are key components of highly active antiretroviral therapy, but they have been associated with adverse side effects, including partial lipodystrophy and metabolic syndrome. We recently demonstrated that a commonly used HIV-PI, lopinavir, inhibits ZMPSTE24, thereby blocking lamin A biogenesis and leading to an accumulation of prelamin A. ZMPSTE24 deficiency in humans causes an accumulation of prelamin A and leads to lipodystrophy and other disease phenotypes. Thus, an accumulation of prelamin A in the setting of HIV-PIs represents a plausible mechanism for some drug side effects. Here we show, with metabolic labeling studies, that lopinavir leads to the accumulation of the farnesylated form of prelamin A. We also tested whether a new and chemically distinct HIV-PI, darunavir, inhibits ZMPSTE24. We found that darunavir does not inhibit the biochemical activity of ZMPSTE24, nor does it lead to an accumulation of farnesyl-prelamin A in cells. This property of darunavir is potentially attractive. However, all HIV-PIs, including darunavir, are generally administered with ritonavir, an HIV-PI that is used to block the metabolism of other HIV-PIs. Ritonavir, like lopinavir, inhibits ZMPSTE24 and leads to an accumulation of prelamin A.


Subject(s)
HIV Infections/diet therapy , HIV Protease Inhibitors/pharmacology , Membrane Proteins/metabolism , Metalloendopeptidases/metabolism , Nuclear Proteins/metabolism , Protein Precursors/metabolism , Protein Processing, Post-Translational/drug effects , Sulfonamides/pharmacology , Animals , Darunavir , HIV Infections/enzymology , HIV Infections/genetics , HIV Protease Inhibitors/adverse effects , HIV Protease Inhibitors/therapeutic use , Humans , Lamin Type A , Lipodystrophy/chemically induced , Lipodystrophy/enzymology , Lipodystrophy/genetics , Lopinavir , Membrane Proteins/antagonists & inhibitors , Membrane Proteins/genetics , Metabolic Syndrome/chemically induced , Metabolic Syndrome/enzymology , Metabolic Syndrome/genetics , Metalloendopeptidases/antagonists & inhibitors , Metalloendopeptidases/genetics , Mice , Mice, Knockout , Pyrimidinones/adverse effects , Pyrimidinones/pharmacology , Pyrimidinones/therapeutic use , Ritonavir/adverse effects , Ritonavir/pharmacology , Ritonavir/therapeutic use , Sulfonamides/adverse effects , Sulfonamides/therapeutic use
15.
Proc Natl Acad Sci U S A ; 104(33): 13432-7, 2007 Aug 14.
Article in English | MEDLINE | ID: mdl-17652517

ABSTRACT

HIV protease inhibitors (HIV-PIs) target the HIV aspartyl protease, which cleaves the HIV gag-pol polyprotein into shorter proteins required for the production of new virions. HIV-PIs are a cornerstone of treatment for HIV but have been associated with lipodystrophy and other side effects. In both human and mouse fibroblasts, we show that HIV-PIs caused an accumulation of prelamin A. The prelamin A in HIV-PI-treated fibroblasts migrated more rapidly than nonfarnesylated prelamin A, comigrating with the farnesylated form of prelamin A that accumulates in ZMPSTE24-deficient fibroblasts. The accumulation of farnesyl-prelamin A in response to HIV-PI treatment was exaggerated in fibroblasts heterozygous for Zmpste24 deficiency. HIV-PIs inhibited the endoproteolytic processing of a GFP-prelamin A fusion protein. The HIV-PIs did not affect the farnesylation of HDJ-2, nor did they inhibit protein farnesyltransferase in vitro. HIV-PIs also did not inhibit the activities of the isoprenyl-cysteine carboxyl methyltransferase ICMT or the prenylprotein endoprotease RCE1 in vitro, but they did inhibit ZMPSTE24 (IC(50): lopinavir, 18.4 +/- 4.6 microM; tipranavir, 1.2 +/- 0.4 microM). We conclude that the HIV-PIs inhibit ZMPSTE24, leading to an accumulation of farnesyl-prelamin A. The inhibition of ZMPSTE24 by HIV-PIs could play a role in the side effects of these drugs.


Subject(s)
HIV Protease Inhibitors/pharmacology , Membrane Proteins/antagonists & inhibitors , Metalloendopeptidases/antagonists & inhibitors , Nuclear Proteins/metabolism , Protein Precursors/metabolism , Base Sequence , DNA Primers , Humans , Lamin Type A
16.
Hum Mutat ; 28(9): 882-9, 2007 Sep.
Article in English | MEDLINE | ID: mdl-17469202

ABSTRACT

Hutchinson-Gilford progeria syndrome (HGPS) is a rare precocious aging syndrome caused by mutations in LMNA that lead to synthesis of a mutant form of prelamin A, generally called progerin, that cannot be processed to mature lamin A. Most HGPS patients have a recurrent heterozygous de novo mutation in exon 11 of LMNA, c.1824C>T/p.G608G; this synonymous mutation activates a nearby cryptic splice donor site, resulting in synthesis of the mutant prelamin A, progerin, which lacks 50 amino acids within the carboxyl-terminal domain. Abnormal splicing is incomplete, so the mutant allele produces some normally-spliced transcripts. Nevertheless, the synthesis of progerin is sufficient to cause misshapen nuclei in cultured cells and severe disease phenotypes in affected patients. Here we present two patients with extraordinarily severe forms of progeria caused by unusual mutations in LMNA. One had a splice site mutation (c.1968+1G>A; or IVS11+1G>A), and the other had a novel synonymous coding region mutation (c.1821G>A/p.V607V). Both mutations caused very frequent use of the same exon 11 splice donor site that is activated in typical HGPS patients. As a consequence, the ratios of progerin mRNA and protein to wild-type were higher than in typical HGPS patients. Fibroblasts from both patients exhibited nuclear shape abnormalities typical of HGPS, and cells treated with a protein farnesyltransferase inhibitor exhibited fewer misshapen nuclei. Thus, farnesyltransferase inhibitors may prove to be useful even when progerin expression levels are higher than those in typical HGPS patients.


Subject(s)
Lamin Type A/genetics , Mutation , Nuclear Proteins/genetics , Progeria/genetics , Protein Precursors/genetics , Cells, Cultured , Child, Preschool , DNA Mutational Analysis , Gene Expression Regulation , Humans , Infant, Newborn , Male , Up-Regulation
SELECTION OF CITATIONS
SEARCH DETAIL
...