Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
1.
Cell ; 2024 Jun 28.
Article in English | MEDLINE | ID: mdl-38959890

ABSTRACT

Hypothalamic neural circuits regulate instinctive behaviors such as food seeking, the fight/flight response, socialization, and maternal care. Here, we identified microdeletions on chromosome Xq23 disrupting the brain-expressed transient receptor potential (TRP) channel 5 (TRPC5). This family of channels detects sensory stimuli and converts them into electrical signals interpretable by the brain. Male TRPC5 deletion carriers exhibited food seeking, obesity, anxiety, and autism, which were recapitulated in knockin male mice harboring a human loss-of-function TRPC5 mutation. Women carrying TRPC5 deletions had severe postpartum depression. As mothers, female knockin mice exhibited anhedonia and depression-like behavior with impaired care of offspring. Deletion of Trpc5 from oxytocin neurons in the hypothalamic paraventricular nucleus caused obesity in both sexes and postpartum depressive behavior in females, while Trpc5 overexpression in oxytocin neurons in knock-in mice reversed these phenotypes. We demonstrate that TRPC5 plays a pivotal role in mediating innate human behaviors fundamental to survival, including food seeking and maternal care.

2.
Cell Rep Med ; 4(8): 101155, 2023 08 15.
Article in English | MEDLINE | ID: mdl-37586323

ABSTRACT

New approaches are needed to treat people whose obesity and type 2 diabetes (T2D) are driven by specific mechanisms. We investigate a deletion on chromosome 16p11.2 (breakpoint 2-3 [BP2-3]) encompassing SH2B1, a mediator of leptin and insulin signaling. Phenome-wide association scans in the UK (N = 502,399) and Estonian (N = 208,360) biobanks show that deletion carriers have increased body mass index (BMI; p = 1.3 × 10-10) and increased rates of T2D. Compared with BMI-matched controls, deletion carriers have an earlier onset of T2D, with poorer glycemic control despite higher medication usage. Cystatin C, a biomarker of kidney function, is significantly elevated in deletion carriers, suggesting increased risk of renal impairment. In a Mendelian randomization study, decreased SH2B1 expression increases T2D risk (p = 8.1 × 10-6). We conclude that people with 16p11.2 BP2-3 deletions have early, complex obesity and T2D and may benefit from therapies that enhance leptin and insulin signaling.


Subject(s)
Diabetes Mellitus, Type 2 , Insulins , Metabolic Diseases , Humans , Leptin , Diabetes Mellitus, Type 2/genetics , Obesity/genetics , Adaptor Proteins, Signal Transducing
3.
Nat Med ; 28(12): 2537-2546, 2022 12.
Article in English | MEDLINE | ID: mdl-36536256

ABSTRACT

Serotonin reuptake inhibitors and receptor agonists are used to treat obesity, anxiety and depression. Here we studied the role of the serotonin 2C receptor (5-HT2CR) in weight regulation and behavior. Using exome sequencing of 2,548 people with severe obesity and 1,117 control individuals without obesity, we identified 13 rare variants in the gene encoding 5-HT2CR (HTR2C) in 19 unrelated people (3 males and 16 females). Eleven variants caused a loss of function in HEK293 cells. All people who carried variants had hyperphagia and some degree of maladaptive behavior. Knock-in male mice harboring a human loss-of-function HTR2C variant developed obesity and reduced social exploratory behavior; female mice heterozygous for the same variant showed similar deficits with reduced severity. Using the 5-HT2CR agonist lorcaserin, we found that depolarization of appetite-suppressing proopiomelanocortin neurons was impaired in knock-in mice. In conclusion, we demonstrate that 5-HT2CR is involved in the regulation of human appetite, weight and behavior. Our findings suggest that melanocortin receptor agonists might be effective in treating severe obesity in individuals carrying HTR2C variants. We suggest that HTR2C should be included in diagnostic gene panels for severe childhood-onset obesity.


Subject(s)
Obesity, Morbid , Receptor, Serotonin, 5-HT2C , Animals , Child , Female , Humans , Male , Mice , HEK293 Cells , Obesity/genetics , Receptor, Serotonin, 5-HT2C/genetics , Serotonin , Serotonin 5-HT2 Receptor Agonists/pharmacology , Adaptation, Psychological
4.
J Clin Endocrinol Metab ; 107(6): e2532-e2544, 2022 05 17.
Article in English | MEDLINE | ID: mdl-35137184

ABSTRACT

CONTEXT: Genetic variants affecting the nuclear hormone receptor coactivator steroid receptor coactivator, SRC-1, have been identified in people with severe obesity and impair melanocortin signaling in cells and mice. As a result, obese patients with SRC-1 deficiency are being treated with a melanocortin 4 receptor agonist in clinical trials. OBJECTIVE: Here, our aim was to comprehensively describe and characterize the clinical phenotype of SRC-1 variant carriers to facilitate diagnosis and clinical management. METHODS: In genetic studies of 2462 people with severe obesity, we identified 23 rare heterozygous variants in SRC-1. We studied 29 adults and 18 children who were SRC-1 variant carriers and performed measurements of metabolic and endocrine function, liver imaging, and adipose tissue biopsies. Findings in adult SRC-1 variant carriers were compared to 30 age- and body mass index (BMI)-matched controls. RESULTS: The clinical spectrum of SRC-1 variant carriers included increased food intake in children, normal basal metabolic rate, multiple fractures with minimal trauma (40%), persistent diarrhea, partial thyroid hormone resistance, and menorrhagia. Compared to age-, sex-, and BMI-matched controls, adult SRC-1 variant carriers had more severe adipose tissue fibrosis (46.2% vs 7.1% respectively, P = .03) and a suggestion of increased liver fibrosis (5/13 cases vs 2/13 in controls, odds ratio = 3.4), although this was not statistically significant. CONCLUSION: SRC-1 variant carriers exhibit hyperphagia in childhood, severe obesity, and clinical features of partial hormone resistance. The presence of adipose tissue fibrosis and hepatic fibrosis in young patients suggests that close monitoring for the early development of obesity-associated metabolic complications is warranted.


Subject(s)
Nuclear Receptor Coactivator 1 , Obesity, Morbid , Female , Fibrosis , Humans , Male , Nuclear Receptor Coactivator 1/genetics , Obesity, Morbid/complications , Obesity, Morbid/genetics
5.
N Engl J Med ; 385(17): 1581-1592, 2021 10 21.
Article in English | MEDLINE | ID: mdl-34614324

ABSTRACT

BACKGROUND: GNAS encodes the Gαs (stimulatory G-protein alpha subunit) protein, which mediates G protein-coupled receptor (GPCR) signaling. GNAS mutations cause developmental delay, short stature, and skeletal abnormalities in a syndrome called Albright's hereditary osteodystrophy. Because of imprinting, mutations on the maternal allele also cause obesity and hormone resistance (pseudohypoparathyroidism). METHODS: We performed exome sequencing and targeted resequencing in 2548 children who presented with severe obesity, and we unexpectedly identified 22 GNAS mutation carriers. We investigated whether the effect of GNAS mutations on melanocortin 4 receptor (MC4R) signaling explains the obesity and whether the variable clinical spectrum in patients might be explained by the results of molecular assays. RESULTS: Almost all GNAS mutations impaired MC4R signaling. A total of 6 of 11 patients who were 12 to 18 years of age had reduced growth. In these patients, mutations disrupted growth hormone-releasing hormone receptor signaling, but growth was unaffected in carriers of mutations that did not affect this signaling pathway (mean standard-deviation score for height, -0.90 vs. 0.75, respectively; P = 0.02). Only 1 of 10 patients who reached final height before or during the study had short stature. GNAS mutations that impaired thyrotropin receptor signaling were associated with developmental delay and with higher thyrotropin levels (mean [±SD], 8.4±4.7 mIU per liter) than those in 340 severely obese children who did not have GNAS mutations (3.9±2.6 mIU per liter; P = 0.004). CONCLUSIONS: Because pathogenic mutations may manifest with obesity alone, screening of children with severe obesity for GNAS deficiency may allow early diagnosis, improving clinical outcomes, and melanocortin agonists may aid in weight loss. GNAS mutations that are identified by means of unbiased genetic testing differentially affect GPCR signaling pathways that contribute to clinical heterogeneity. Monogenic diseases are clinically more variable than their classic descriptions suggest. (Funded by Wellcome and others.).


Subject(s)
GTP-Binding Protein alpha Subunits, Gs/genetics , Mutation , Pediatric Obesity/genetics , Receptor, Melanocortin, Type 4/metabolism , Adolescent , Body Height , Child , Chromogranins/genetics , Female , GTP-Binding Protein alpha Subunits, Gs/deficiency , Humans , Male , Mutation, Missense , Receptors, Thyrotropin/metabolism , Signal Transduction , Exome Sequencing
6.
Cell Rep ; 34(12): 108862, 2021 03 23.
Article in English | MEDLINE | ID: mdl-33761344

ABSTRACT

The Melanocortin-4 Receptor (MC4R) plays a pivotal role in energy homeostasis. We used human MC4R mutations associated with an increased or decreased risk of obesity to dissect mechanisms that regulate MC4R function. Most obesity-associated mutations impair trafficking to the plasma membrane (PM), whereas obesity-protecting mutations either accelerate recycling to the PM or decrease internalization, resulting in enhanced signaling. MC4R mutations that do not affect canonical Gαs protein-mediated signaling, previously considered to be non-pathogenic, nonetheless disrupt agonist-induced internalization, ß-arrestin recruitment, and/or coupling to Gαs, establishing their causal role in severe obesity. Structural mapping reveals ligand-accessible sites by which MC4R couples to effectors and residues involved in the homodimerization of MC4R, which is disrupted by multiple obesity-associated mutations. Human genetic studies reveal that endocytosis, intracellular trafficking, and homodimerization regulate MC4R function to a level that is physiologically relevant, supporting the development of chaperones, agonists, and allosteric modulators of MC4R for weight loss therapy.


Subject(s)
Body Weight/genetics , Endocytosis , Genetic Variation , Protein Multimerization , Receptor, Melanocortin, Type 4/genetics , Animals , COS Cells , Cell Membrane/metabolism , Chlorocebus aethiops , Cyclic AMP/metabolism , Extracellular Signal-Regulated MAP Kinases/metabolism , GTP-Binding Protein alpha Subunits, Gs , HEK293 Cells , Humans , Models, Biological , Mutant Proteins/metabolism , Mutation/genetics , Phosphorylation , Receptor, Melanocortin, Type 4/chemistry , Signal Transduction , beta-Arrestins/metabolism
7.
Cell Metab ; 31(6): 1107-1119.e12, 2020 06 02.
Article in English | MEDLINE | ID: mdl-32492392

ABSTRACT

Obesity is genetically heterogeneous with monogenic and complex polygenic forms. Using exome and targeted sequencing in 2,737 severely obese cases and 6,704 controls, we identified three genes (PHIP, DGKI, and ZMYM4) with an excess burden of very rare predicted deleterious variants in cases. In cells, we found that nuclear PHIP (pleckstrin homology domain interacting protein) directly enhances transcription of pro-opiomelanocortin (POMC), a neuropeptide that suppresses appetite. Obesity-associated PHIP variants repressed POMC transcription. Our demonstration that PHIP is involved in human energy homeostasis through transcriptional regulation of central melanocortin signaling has potential diagnostic and therapeutic implications for patients with obesity and developmental delay. Additionally, we found an excess burden of predicted deleterious variants involving genes nearest to loci from obesity genome-wide association studies. Genes and gene sets influencing obesity with variable penetrance provide compelling evidence for a continuum of causality in the genetic architecture of obesity, and explain some of its missing heritability.


Subject(s)
Intracellular Signaling Peptides and Proteins/genetics , Pediatric Obesity/genetics , Pro-Opiomelanocortin/genetics , Adult , Animals , Cells, Cultured , Child , Chlorocebus aethiops , Exome , Female , Genetic Variation/genetics , Humans , Male , Mice , Mice, Inbred C57BL , Middle Aged
8.
PLoS Genet ; 15(1): e1007603, 2019 01.
Article in English | MEDLINE | ID: mdl-30677029

ABSTRACT

The variation in weight within a shared environment is largely attributable to genetic factors. Whilst many genes/loci confer susceptibility to obesity, little is known about the genetic architecture of healthy thinness. Here, we characterise the heritability of thinness which we found was comparable to that of severe obesity (h2 = 28.07 vs 32.33% respectively), although with incomplete genetic overlap (r = -0.49, 95% CI [-0.17, -0.82], p = 0.003). In a genome-wide association analysis of thinness (n = 1,471) vs severe obesity (n = 1,456), we identified 10 loci previously associated with obesity, and demonstrate enrichment for established BMI-associated loci (pbinomial = 3.05x10-5). Simulation analyses showed that different association results between the extremes were likely in agreement with additive effects across the BMI distribution, suggesting different effects on thinness and obesity could be due to their different degrees of extremeness. In further analyses, we detected a novel obesity and BMI-associated locus at PKHD1 (rs2784243, obese vs. thin p = 5.99x10-6, obese vs. controls p = 2.13x10-6 pBMI = 2.3x10-13), associations at loci recently discovered with much larger sample sizes (e.g. FAM150B and PRDM6-CEP120), and novel variants driving associations at previously established signals (e.g. rs205262 at the SNRPC/C6orf106 locus and rs112446794 at the PRDM6-CEP120 locus). Our ability to replicate loci found with much larger sample sizes demonstrates the value of clinical extremes and suggest that characterisation of the genetics of thinness may provide a more nuanced understanding of the genetic architecture of body weight regulation and may inform the identification of potential anti-obesity targets.


Subject(s)
Muscle Proteins/genetics , Neoplasm Proteins/genetics , Obesity, Morbid/genetics , Receptors, Cell Surface/genetics , Thinness/genetics , Transcription Factors/genetics , Adult , Alleles , Body Mass Index , Female , Genetic Predisposition to Disease , Genome-Wide Association Study , Humans , Male , Middle Aged , Obesity, Morbid/physiopathology , Polymorphism, Single Nucleotide , Thinness/physiopathology
9.
Cell ; 176(4): 729-742.e18, 2019 02 07.
Article in English | MEDLINE | ID: mdl-30661757

ABSTRACT

Hypothalamic melanocortin neurons play a pivotal role in weight regulation. Here, we examined the contribution of Semaphorin 3 (SEMA3) signaling to the development of these circuits. In genetic studies, we found 40 rare variants in SEMA3A-G and their receptors (PLXNA1-4; NRP1-2) in 573 severely obese individuals; variants disrupted secretion and/or signaling through multiple molecular mechanisms. Rare variants in this set of genes were significantly enriched in 982 severely obese cases compared to 4,449 controls. In a zebrafish mutagenesis screen, deletion of 7 genes in this pathway led to increased somatic growth and/or adiposity demonstrating that disruption of Semaphorin 3 signaling perturbs energy homeostasis. In mice, deletion of the Neuropilin-2 receptor in Pro-opiomelanocortin neurons disrupted their projections from the arcuate to the paraventricular nucleus, reduced energy expenditure, and caused weight gain. Cumulatively, these studies demonstrate that SEMA3-mediated signaling drives the development of hypothalamic melanocortin circuits involved in energy homeostasis.


Subject(s)
Energy Metabolism/genetics , Melanocortins/metabolism , Semaphorins/genetics , Adolescent , Adult , Animals , Body Weight , Cell Line , Child , Child, Preschool , Disease Models, Animal , Eating , Female , Genetic Variation/genetics , Homeostasis , Humans , Hypothalamus/metabolism , Leptin/metabolism , Male , Mice , Mice, Inbred C57BL , Middle Aged , Nerve Tissue Proteins/metabolism , Neurons/metabolism , Obesity/genetics , Obesity/metabolism , Receptors, Cell Surface/metabolism , Semaphorins/metabolism , Young Adult , Zebrafish
10.
Mol Metab ; 6(10): 1321-1329, 2017 10.
Article in English | MEDLINE | ID: mdl-29031731

ABSTRACT

OBJECTIVE: Pro-opiomelanocortin (POMC)-derived peptides act on neurons expressing the Melanocortin 4 receptor (MC4R) to reduce body weight. Setmelanotide is a highly potent MC4R agonist that leads to weight loss in diet-induced obese animals and in obese individuals with complete POMC deficiency. While POMC deficiency is very rare, 1-5% of severely obese individuals harbor heterozygous mutations in MC4R. We sought to assess the efficacy of Setmelanotide in human MC4R deficiency. METHODS: We studied the effects of Setmelanotide on mutant MC4Rs in cells and the weight loss response to Setmelanotide administration in rodent studies and a human clinical trial. We annotated the functional status of 369 published MC4R variants. RESULTS: In cells, we showed that Setmelanotide is significantly more potent at MC4R than the endogenous ligand alpha-melanocyte stimulating hormone and can disproportionally rescue signaling by a subset of severely impaired MC4R mutants. Wild-type rodents appear more sensitive to Setmelanotide when compared to MC4R heterozygous deficient mice, while MC4R knockout mice fail to respond. In a 28-day Phase 1b clinical trial, Setmelanotide led to weight loss in obese MC4R variant carriers. Patients with POMC defects upstream of MC4R show significantly more weight loss with Setmelanotide than MC4R deficient patients or obese controls. CONCLUSIONS: Setmelanotide led to weight loss in obese people with MC4R deficiency; however, further studies are justified to establish whether Setmelanotide can elicit clinically meaningful weight loss in a subset of the MC4R deficient obese population.


Subject(s)
Receptor, Melanocortin, Type 4/agonists , Receptor, Melanocortin, Type 4/deficiency , alpha-MSH/analogs & derivatives , Adrenal Insufficiency/drug therapy , Adrenal Insufficiency/metabolism , Adult , Amino Acid Sequence , Animals , Female , HEK293 Cells , Humans , Male , Mice , Mice, Knockout , Middle Aged , Obesity/drug therapy , Obesity/metabolism , Pro-Opiomelanocortin/deficiency , Pro-Opiomelanocortin/metabolism , Receptor, Melanocortin, Type 4/genetics , Receptor, Melanocortin, Type 4/metabolism , alpha-MSH/pharmacology
11.
Sci Rep ; 7(1): 4394, 2017 06 29.
Article in English | MEDLINE | ID: mdl-28663568

ABSTRACT

Obesity is a genetically heterogeneous disorder. Using targeted and whole-exome sequencing, we studied 32 human and 87 rodent obesity genes in 2,548 severely obese children and 1,117 controls. We identified 52 variants contributing to obesity in 2% of cases including multiple novel variants in GNAS, which were sometimes found with accelerated growth rather than short stature as described previously. Nominally significant associations were found for rare functional variants in BBS1, BBS9, GNAS, MKKS, CLOCK and ANGPTL6. The p.S284X variant in ANGPTL6 drives the association signal (rs201622589, MAF~0.1%, odds ratio = 10.13, p-value = 0.042) and results in complete loss of secretion in cells. Further analysis including additional case-control studies and population controls (N = 260,642) did not support association of this variant with obesity (odds ratio = 2.34, p-value = 2.59 × 10-3), highlighting the challenges of testing rare variant associations and the need for very large sample sizes. Further validation in cohorts with severe obesity and engineering the variants in model organisms will be needed to explore whether human variants in ANGPTL6 and other genes that lead to obesity when deleted in mice, do contribute to obesity. Such studies may yield druggable targets for weight loss therapies.


Subject(s)
Genetic Association Studies , Genetic Predisposition to Disease , Genetic Variation , Obesity, Morbid/genetics , Pediatric Obesity/genetics , Animals , Case-Control Studies , Chromogranins/chemistry , Chromogranins/genetics , Chromogranins/metabolism , Female , GTP-Binding Protein alpha Subunits, Gs/chemistry , GTP-Binding Protein alpha Subunits, Gs/genetics , GTP-Binding Protein alpha Subunits, Gs/metabolism , Humans , Male , Mice , Models, Molecular , Mutation , Obesity, Morbid/diagnosis , Odds Ratio , Pediatric Obesity/diagnosis , Pedigree , Protein Conformation , Rodentia
12.
Cell ; 155(4): 765-77, 2013 Nov 07.
Article in English | MEDLINE | ID: mdl-24209692

ABSTRACT

Kinase suppressor of Ras 2 (KSR2) is an intracellular scaffolding protein involved in multiple signaling pathways. Targeted deletion of Ksr2 leads to obesity in mice, suggesting a role in energy homeostasis. We explored the role of KSR2 in humans by sequencing 2,101 individuals with severe early-onset obesity and 1,536 controls. We identified multiple rare variants in KSR2 that disrupt signaling through the Raf-MEKERK pathway and impair cellular fatty acid oxidation and glucose oxidation in transfected cells; effects that can be ameliorated by the commonly prescribed antidiabetic drug, metformin. Mutation carriers exhibit hyperphagia in childhood, low heart rate, reduced basal metabolic rate and severe insulin resistance. These data establish KSR2 as an important regulator of energy intake, energy expenditure, and substrate utilization in humans. Modulation of KSR2-mediated effects may represent a novel therapeutic strategy for obesity and type 2 diabetes.


Subject(s)
Insulin Resistance , Obesity/genetics , Protein Serine-Threonine Kinases/genetics , Age Factors , Age of Onset , Amino Acid Sequence , Animals , Child , Energy Metabolism , Fatty Acids/metabolism , Female , Glucose/metabolism , Humans , Hyperphagia/genetics , Hyperphagia/metabolism , MAP Kinase Signaling System , Male , Mice , Models, Molecular , Molecular Sequence Data , Obesity/epidemiology , Obesity/metabolism , Oxidation-Reduction , Protein Serine-Threonine Kinases/chemistry , Protein Serine-Threonine Kinases/metabolism , Protein Structure, Tertiary , Proto-Oncogene Proteins B-raf/chemistry , Proto-Oncogene Proteins B-raf/metabolism , Sequence Alignment
13.
Mol Cell ; 44(4): 585-96, 2011 Nov 18.
Article in English | MEDLINE | ID: mdl-22099306

ABSTRACT

The 7-methylguanosine cap added to the 5' end of mRNA is required for efficient gene expression in eukaryotes. In mammals, methylation of the guanosine cap is catalyzed by RNMT (RNA guanine-7 methyltransferase), an enzyme previously thought to function as a monomer. We have identified an obligate component of the mammalian cap methyltransferase, RAM (RNMT-Activating Mini protein)/Fam103a1, a previously uncharacterized protein. RAM consists of an N-terminal RNMT-activating domain and a C-terminal RNA-binding domain. As monomers RNMT and RAM have a relatively weak affinity for RNA; however, together their RNA affinity is significantly increased. RAM is required for efficient cap methylation in vitro and in vivo, and is indirectly required to maintain mRNA expression levels, for mRNA translation and for cell viability. Our findings demonstrate that RAM is an essential component of the core gene expression machinery.


Subject(s)
Gene Expression Regulation , Methyltransferases/metabolism , Nuclear Proteins/metabolism , Protein Biosynthesis/genetics , RNA Caps/metabolism , RNA-Binding Proteins/metabolism , Recombinant Proteins/metabolism , Amino Acid Sequence , Binding Sites , Blotting, Western , Cell Survival , Conserved Sequence , Guanosine/analogs & derivatives , Guanosine/genetics , Guanosine/metabolism , HEK293 Cells , Humans , Immunoprecipitation , Methylation , Methyltransferases/genetics , Molecular Sequence Data , Nuclear Proteins/chemistry , Nuclear Proteins/genetics , Protein Structure, Tertiary , RNA Caps/genetics , RNA-Binding Proteins/chemistry , RNA-Binding Proteins/genetics , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , S-Adenosylmethionine/genetics , S-Adenosylmethionine/metabolism , Sequence Alignment
14.
Mov Disord ; 24(1): 104-8, 2009 Jan 15.
Article in English | MEDLINE | ID: mdl-19006224

ABSTRACT

We screened for mutations in the PARKIN, DJ-1, and PINK1 genes in a Taiwanese cohort (68 probands; 58 sporadic and 10 familial) with early-onset parkinsonism (EOP, onset <50 years of age). We identified 9 patients harboring mutations in PARKIN (three compound heterozygous and six single heterozygous carriers), 3 patients with heterozygous PINK1 mutations (including two novel substitutions M341I and P209A), and no DJ-1 mutations. Our frequencies of PARKIN (two allele mutation, 4.4%; single allele, 8.8%) and PINK1 (single heterozygous, 4.4%) mutations in Taiwanese-Chinese are similar to those in Caucasian and other Asian EOP patients. Although the role of heterozygosity of recessive genes in EOP remains to be resolved, molecular analysis and functional imaging will play a decisive role in differential diagnosis and determined therapeutic strategy.


Subject(s)
Asian People/genetics , Intracellular Signaling Peptides and Proteins/genetics , Oncogene Proteins/genetics , Parkinsonian Disorders/ethnology , Protein Kinases/genetics , Ubiquitin-Protein Ligases/genetics , Adolescent , Adult , Age of Onset , Amino Acid Substitution , China/ethnology , Cohort Studies , Exons/genetics , Female , Gene Frequency , Genes, Recessive , Genotype , Humans , Male , Middle Aged , Mutation, Missense , Parkinsonian Disorders/genetics , Phenotype , Polymerase Chain Reaction , Protein Deglycase DJ-1 , RNA, Messenger/genetics , Taiwan/epidemiology , Young Adult
15.
J Natl Cancer Inst ; 97(22): 1645-51, 2005 Nov 16.
Article in English | MEDLINE | ID: mdl-16288117

ABSTRACT

BACKGROUND: We previously reported that all-trans-retinoic acid (RA) treatment can prevent in vitro transformation of immortalized human bronchial epithelial (HBE) cells. METHODS: To determine whether methylation inhibits RARbeta expression in HBE cells, we used sodium bisulfite sequencing to compare RARbeta P2 promoter methylation patterns in RA-sensitive (BEAS-2B) and RA-resistant (BEAS-2B-R1) HBE cells. Immunoblotting was used to assess induction of the RARbeta, placental transforming growth factor beta (PTGF-beta), Fos-related antigen 1 (Fra-1), and transglutaminase II (TGase II) proteins by RA following treatment with azacitidine, a DNA demethylating agent. The expression, transcriptional activity, and growth suppressive activity of RARbeta1', a novel RAR isoform, were evaluated in lung cancer cells transfected with RARbeta1', and expression was also studied in paired normal lung tissues and lung tumors. All statistical tests were two-sided. RESULTS: Hypermethylation was observed in the 3' region of the RARbeta P2 promoter of BEAS-2B-R1 but not BEAS-2B cells. Azacitidine treatment of BEAS-2B-R1 cells restored RA-inducible RARbeta2 and PTGF-beta expression but not that of RARbeta1', Fra-1, or TGase II. RARbeta1' expression was repressed in RA-resistant BEAS-2B-R1 cells and in lung cancers, compared with adjacent normal lung tissues. BEAS-2B-R1 cells transiently transfected with RARbeta1' had increased RA-dependent activation of a retinoic acid receptor element (RARE)-containing reporter plasmid compared with vector control (mean = 3.2, 95% confidence interval [CI] = 3.1 to 3.3 versus mean = 1.4, 95% CI = 1.3 to 1.5; P<.001). In H358 lung cancer cells transiently transfected with RARbeta1', RA treatment restored target gene expression compared with that in vector-transfected cells and suppressed cell growth compared with that in untreated cells (4 microM; treated mean = 0.49 versus untreated mean = 1.0, difference = 0.51, 95% CI = 0.35 to 0.67, P = .003; 8 microM: treated mean = 0.50 versus untreated mean = 1.0, difference = 0.50, 95% CI = 0.26 to 0.74, P = .015). CONCLUSION: Restoration of RARbeta1' expression may overcome retinoid resistance in lung carcinogenesis.


Subject(s)
Antimetabolites, Antineoplastic/pharmacology , Azacitidine/pharmacology , Bronchial Neoplasms/prevention & control , Lung Neoplasms/drug therapy , Receptors, Retinoic Acid/metabolism , Respiratory Mucosa/pathology , Bone Morphogenetic Proteins/metabolism , Cell Line, Tumor , DNA Methylation/drug effects , GTP-Binding Proteins/metabolism , Gene Expression Regulation, Neoplastic/drug effects , Growth Differentiation Factor 15 , Humans , Immunoblotting , Luciferases/analysis , Lung Neoplasms/metabolism , Membrane Proteins/metabolism , Promoter Regions, Genetic/drug effects , Protein Glutamine gamma Glutamyltransferase 2 , Protein Isoforms , Proto-Oncogene Proteins c-fos/metabolism , Receptors, Retinoic Acid/genetics , Respiratory Mucosa/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Transcription, Genetic/drug effects , Transfection , Transglutaminases/metabolism , Tretinoin/pharmacology
16.
Arch Neurol ; 62(1): 82-7, 2005 Jan.
Article in English | MEDLINE | ID: mdl-15642853

ABSTRACT

BACKGROUND: Loss of function of the parkin gene (PRKN) is the predominant genetic cause of juvenile and early-onset parkinsonism in Japan, Europe, and the United States. OBJECTIVES: To evaluate the frequency of PRKN mutations in Taiwanese (ethnic Chinese) patients with early-onset parkinsonism and to explore genotype-phenotype correlations. DESIGN: Clinical assessment included medical, neurologic, and psychiatric evaluation. Genomic DNA sequencing and quantitative polymerase chain reaction were performed to identify PRKN mutations. Gene expression was examined in patient lymphoblastoid cell lines, in which PRKN mutations were identified. PATIENTS: Forty-one Taiwanese patients with early-onset parkinsonism (aged <50 years at onset). RESULTS: Four of 41 probands had PRKN mutations. One proband had compound heterozygous mutations, with a PRKN exon 2 deletion and an exon 7 G284R substitution. The phenotype resembled typical Parkinson disease. Three patients were mutation carriers. One proband had PRKN exon 2 and exon 3 deletions in the same allele. However, this patient's phenotype was that of classic "parkin-proven" autosomal recessive juvenile parkinsonism, characterized by symmetrical foot dystonia at onset, gait disturbance, diurnal change, and very slow progression. The 2 remaining carriers had novel heterozygous exon 11 R396G substitutions. Patients with PRKN mutations were younger at onset than those without mutations, and they required a lower dose of levodopa despite longer disease duration. CONCLUSIONS: Mutations in PRKN are a rare cause of early-onset parkinsonism in Taiwanese individuals. The overall mutation frequency, adjusted for age at onset, was comparable with that reported for white cohorts; however, the point mutations identified seem to be population specific.


Subject(s)
Mutation , Parkinsonian Disorders/genetics , Ubiquitin-Protein Ligases/genetics , Adult , Arginine/genetics , Cohort Studies , DNA Mutational Analysis/methods , Exons , Family Health , Female , Glycine/genetics , Humans , Male , Middle Aged , Neurologic Examination , Pedigree , RNA, Messenger/biosynthesis , Reverse Transcriptase Polymerase Chain Reaction/methods , Taiwan
17.
Mov Disord ; 19(9): 1065-9, 2004 Sep.
Article in English | MEDLINE | ID: mdl-15372597

ABSTRACT

Recently, mutations in DJ-1 (PARK7) were described as a novel cause of early-onset parkinsonism. We analysed the DJ-1 gene in a cohort of patients originating from Taiwan with early-onset Parkinson's disease; 41 subjects were clinically and genetically examined. These patients were evaluated previously for the presence of parkin mutations (PARK2) and were found to be negative. The entire DJ-1 open-reading frame was amplified from cDNA, analysed for size alterations indicative of mutations affecting splice motifs, and sequenced to identify coding variants. In addition, we developed quantitative polymerase chain reaction assays to examine the genomic copy number of DJ-1 exons. No potential splice site mutations, coding sequence alterations, or exon deletion/duplications were detected. Our results and previous studies suggest that alterations to DJ-1 are not a common cause of early-onset Parkinson's disease and other causes, genetic and/or environmental, remain to be identified.


Subject(s)
Oncogene Proteins/genetics , Parkinson Disease/ethnology , Parkinson Disease/genetics , Point Mutation/genetics , Adult , Age of Onset , China/ethnology , Cohort Studies , DNA, Complementary/genetics , Electrophoresis, Agar Gel , Exons/genetics , Female , Gene Deletion , Humans , Intracellular Signaling Peptides and Proteins , Male , Middle Aged , Protein Deglycase DJ-1 , Reverse Transcriptase Polymerase Chain Reaction , Taiwan
18.
Mov Disord ; 18(11): 1306-11, 2003 Nov.
Article in English | MEDLINE | ID: mdl-14639672

ABSTRACT

We report on an evaluation of coding variants within the parkin gene to assess their frequency in a North American clinical series of 313 Parkinson's disease (PD) cases and 192 unrelated controls. We hypothesized that the carrier frequency of parkin coding mutations, exon deletions, or duplications may be greater in PD cases. However, point mutations and exonic deletions/duplications, reported previously as pathogenic in homozygous or compound heterozygous individuals, occurred in both cases and controls with similar frequencies (3.8% in cases, 3.1% in controls). Furthermore, only stratified subanalyses detected any genetic association between the V380L common coding polymorphism and PD. We discuss the implication of parkin mutations for Parkinson's disease from this population perspective.


Subject(s)
Chromosome Mapping/methods , Parkinson Disease/genetics , Ubiquitin-Protein Ligases/genetics , Adult , Aged , Aged, 80 and over , Chromatography, High Pressure Liquid , DNA Mutational Analysis , Exons/genetics , Female , Gene Deletion , Humans , Male , Middle Aged , Point Mutation/genetics , Polymerase Chain Reaction , Polymorphism, Genetic/genetics , United States/epidemiology
SELECTION OF CITATIONS
SEARCH DETAIL
...