Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 4 de 4
Filter
1.
N Engl J Med ; 370(20): 1909-1919, 2014 May 15.
Article in English | MEDLINE | ID: mdl-24827035

ABSTRACT

BACKGROUND: Genetic analysis has been successful in identifying causative mutations for individual cardiovascular risk factors. Success has been more limited in mapping susceptibility genes for clusters of cardiovascular risk traits, such as those in the metabolic syndrome. METHODS: We identified three large families with coinheritance of early-onset coronary artery disease, central obesity, hypertension, and diabetes. We used linkage analysis and whole-exome sequencing to identify the disease-causing gene. RESULTS: A founder mutation was identified in DYRK1B, substituting cysteine for arginine at position 102 in the highly conserved kinase-like domain. The mutation precisely cosegregated with the clinical syndrome in all the affected family members and was absent in unaffected family members and unrelated controls. Functional characterization of the disease gene revealed that nonmutant protein encoded by DYRK1B inhibits the SHH (sonic hedgehog) and Wnt signaling pathways and consequently enhances adipogenesis. Furthermore, DYRK1B promoted the expression of the key gluconeogenic enzyme glucose-6-phosphatase. The R102C allele showed gain-of-function activities by potentiating these effects. A second mutation, substituting proline for histidine 90, was found to cosegregate with a similar clinical syndrome in an ethnically distinct family. CONCLUSIONS: These findings indicate a role for DYRK1B in adipogenesis and glucose homeostasis and associate its altered function with an inherited form of the metabolic syndrome. (Funded by the National Institutes of Health.).


Subject(s)
Genetic Predisposition to Disease , Metabolic Syndrome/genetics , Mutation , Protein Serine-Threonine Kinases/genetics , Protein-Tyrosine Kinases/genetics , Coronary Artery Disease/genetics , Diabetes Mellitus, Type 2/genetics , Exome , Female , Founder Effect , Genetic Linkage , Glucose-6-Phosphatase/metabolism , Humans , Hypertension/genetics , Male , Obesity, Abdominal/genetics , Pedigree , Dyrk Kinases
2.
Am J Pathol ; 183(3): 841-56, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23973270

ABSTRACT

First trimester human decidua is composed of decidual cells, CD56(bright)CD16(-) decidual natural killer (dNK) cells, and macrophages. Decidual cells incubated with NK cell-derived IFN-γ and either macrophage-derived TNF-α or IL-1ß synergistically enhanced mRNA and protein expression of IP-10 and I-TAC. Both chemokines recruit CXCR3-expressing NK cells. This synergy required IFN-γ receptor 1 and 2 mediation via JAK/STAT and NFκB signaling pathways. However, synergy was not observed on neutrophil, monocyte, and NK cell-recruiting chemokines. Immunostaining of first trimester decidua localized IP-10, I-TAC, IFN-γR1, and -R2 to vimentin-positive decidual cells versus cytokeratin-positive interstitial trophoblasts. Flow cytometry identified high CXCR3 levels on dNK cells and minority peripheral CD56(bright)CD16(-) pNK cells and intermediate CXCR3 levels on the majority of CD56(dim)CD16(+) pNK cells. Incubation of pNK cells with either IP-10 or I-TAC elicited concentration-dependent enhanced CXCR3 levels and migration of both pNK cell subsets that peaked at 10 ng/mL, whereas each chemokine at a concentration of 50 ng/mL inhibited CXCR3 expression and pNK cell migration. Deciduae from women with preeclampsia, a leading cause of maternal and fetal morbidity and mortality, displayed significantly lower dNK cell numbers and higher IP-10 and I-TAC levels versus gestational age-matched controls. Significantly elevated IP-10 levels in first trimester sera from women eventually developing preeclampsia compared with controls, identifying IP-10 as a novel, robust early predictor of preeclampsia.


Subject(s)
Chemokines/metabolism , Decidua/pathology , Killer Cells, Natural/pathology , Pre-Eclampsia/etiology , Pre-Eclampsia/pathology , CD56 Antigen/metabolism , Chemokines/genetics , Female , Flow Cytometry , Gene Knockdown Techniques , Humans , Interferon-gamma/pharmacology , Interleukin-1beta/pharmacology , Interleukins/metabolism , Killer Cells, Natural/drug effects , Killer Cells, Natural/metabolism , Models, Biological , Oligonucleotide Array Sequence Analysis , Pre-Eclampsia/blood , Pregnancy , Pregnancy Trimester, First/blood , Pregnancy Trimester, First/drug effects , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Small Interfering/metabolism , Receptors, Chemokine/blood , Receptors, Chemokine/genetics , Receptors, Chemokine/metabolism , Receptors, IgG/metabolism , Signal Transduction/drug effects , Tumor Necrosis Factor-alpha/pharmacology
3.
Hum Mutat ; 34(9): 1221-5, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23703864

ABSTRACT

A rare mutation in LRP6 has been shown to underlie autosomal dominant coronary artery disease (CAD) and metabolic syndrome in an Iranian kindred. The prevalence and spectrum of LRP6 mutations in the disease population of the United States is not known. Two hundred white Americans with early onset familial CAD and metabolic syndrome and 2,000 healthy Northern European controls were screened for nonconservative mutations in LRP6. Three novel mutations were identified, which cosegregated with the metabolic traits in the kindreds of the affected subjects and none in the controls. All three mutations reside in the second propeller domain, which plays a critical role in ligand binding. Two of the mutations substituted highly conserved arginines in the second YWTD domain and the third substituted a conserved glycosylation site. The functional characterization of one of the variants showed that it impairs Wnt signaling and acts as a loss of function mutation.


Subject(s)
Coronary Disease/genetics , Coronary Disease/metabolism , Low Density Lipoprotein Receptor-Related Protein-6/genetics , Low Density Lipoprotein Receptor-Related Protein-6/metabolism , Metabolic Syndrome/genetics , Metabolic Syndrome/metabolism , Adult , Aged , Case-Control Studies , Coronary Disease/complications , Europe , Female , Genetic Predisposition to Disease , Genetic Variation , Glycosylation , Humans , Male , Middle Aged , Mutation , Pedigree , Phylogeny , Sequence Alignment , United States , Wnt Proteins/metabolism , Young Adult
4.
Proc Natl Acad Sci U S A ; 108(5): 1914-8, 2011 Feb 01.
Article in English | MEDLINE | ID: mdl-21245321

ABSTRACT

Vascular smooth muscle cell (VSMC) proliferation is an important event in atherosclerosis and other vasculopathies. PDGF signaling is a key mediator of SMC proliferation, but the mechanisms that control its activity remain unclear. We previously identified a mutation in LDL receptor-related protein 6 (LRP6), LRP6(R611C), that causes early atherosclerosis. Examination of human atherosclerotic coronary arteries showed markedly increased expression of LRP6 and colocalization with PDGF receptor ß (PDGFR-ß). Further investigation showed that wild-type LRP6 inhibits but LRP6(R611C) promotes VSMC proliferation in response to PDGF. We found that wild-type LRP6 forms a complex with PDGFR-ß and enhances its lysosomal degradation, functions that are severely impaired in LRP6(R611C). Further, we observed that wild-type and mutant LRP6 regulate cell-cycle activity by triggering differential effects on PDGF-dependent pathways. These findings implicate LRP6 as a critical modulator of PDGF-dependent regulation of cell cycle in smooth muscle and indicate that loss of this function contributes to development of early atherosclerosis in humans.


Subject(s)
Atherosclerosis/physiopathology , Cell Proliferation , LDL-Receptor Related Proteins/physiology , Muscle, Smooth, Vascular/cytology , Platelet-Derived Growth Factor/physiology , Atherosclerosis/pathology , Cyclin D1/metabolism , Humans , Immunohistochemistry , Low Density Lipoprotein Receptor-Related Protein-6 , RNA, Messenger/genetics , Receptor, Platelet-Derived Growth Factor beta/genetics , Signal Transduction
SELECTION OF CITATIONS
SEARCH DETAIL
...