Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
Add more filters











Publication year range
1.
Int J Mol Sci ; 25(16)2024 Aug 20.
Article in English | MEDLINE | ID: mdl-39201703

ABSTRACT

Preeclampsia is a complex pregnancy-related hypertensive disorder which poses significant risks for both maternal and fetal health. Preeclampsia affects 5-8% of pregnancies in the United States, causing a significant public health and economic burden. Despite extensive research, the etiology and pathogenesis of preeclampsia remain elusive, but have been correlated with maternal conditions such as obesity. In recent decades, the incidence of preeclampsia increased along with the prevalence of obesity among women of reproductive age. Maternal obesity has been shown to negatively affect pregnancy in almost all aspects. However, the precise mechanisms by which obesity influences preeclampsia are unclear. Ankyrin repeat and SOCS Box Containing protein 4 (ASB4) is an E3 ubiquitin ligase that can promote the degradation of a wide range of target proteins. ASB4-null mice display a full spectrum of preeclampsia-like phenotypes during pregnancy including hypertension, proteinuria, and decreased litter size. Furthermore, maternal obesity induced by a high-fat diet aggravates preeclampsia-like phenotypes in pregnant mice lacking ASB4. Variants in the ASB4 gene have been associated with obesity in humans, and a functional connection between the ASB4 gene and obesity has been established in mice. This review discusses the connections between preeclampsia, obesity, and ASB4.


Subject(s)
Obesity , Pre-Eclampsia , Suppressor of Cytokine Signaling Proteins , Animals , Female , Humans , Mice , Pregnancy , Obesity/metabolism , Obesity/genetics , Obesity/etiology , Pre-Eclampsia/etiology , Pre-Eclampsia/metabolism , Pre-Eclampsia/genetics , Suppressor of Cytokine Signaling Proteins/genetics , Suppressor of Cytokine Signaling Proteins/metabolism
2.
Int J Mol Sci ; 24(3)2023 Jan 21.
Article in English | MEDLINE | ID: mdl-36768469

ABSTRACT

Obesity is a risk factor for preeclampsia. We investigated how obesity influences preeclampsia in mice lacking ankyrin-repeat-and-SOCS-box-containing-protein 4 (ASB4), which promotes trophoblast differentiation via degrading the inhibitor of DNA-binding protein 2 (ID2). Asb4-/- mice on normal chow (NC) develop mild preeclampsia-like phenotypes during pregnancy, including hypertension, proteinuria, and reduced litter size. Wild-type (WT) and Asb4-/- females were placed on a high-fat diet (HFD) starting at weaning. At the age of 8-9 weeks, they were mated with WT or Asb4-/- males, and preeclamptic phenotypes were assessed. HFD-WT dams had no obvious adverse outcomes of pregnancy. In contrast, HFD-Asb4-/- dams had significantly more severe preeclampsia-like phenotypes compared to NC-Asb4-/- dams. The HFD increased white fat weights and plasma leptin and insulin levels in Asb4-/- females. In the HFD-Asb4-/- placenta, ID2 amounts doubled without changing the transcript levels, indicating that insulin likely increases ID2 at a level of post-transcription. In human first-trimester trophoblast HTR8/SVneo cells, exposure to insulin, but not to leptin, led to a significant increase in ID2. HFD-induced obesity markedly worsens the preeclampsia-like phenotypes in the absence of ASB4. Our data indicate that hyperinsulinemia perturbs the timely removal of ID2 and interferes with proper trophoblast differentiation, contributing to enhanced preeclampsia.


Subject(s)
Insulin , Pre-Eclampsia , Pregnancy , Male , Female , Humans , Animals , Mice , Infant , Insulin/metabolism , Trophoblasts/metabolism , Pre-Eclampsia/genetics , Pre-Eclampsia/metabolism , Leptin/metabolism , Placenta/metabolism , Insulin, Regular, Human , Obesity/complications , Obesity/genetics , Obesity/metabolism , Inhibitor of Differentiation Protein 2/genetics , Inhibitor of Differentiation Protein 2/metabolism
3.
Sci Rep ; 12(1): 14256, 2022 08 22.
Article in English | MEDLINE | ID: mdl-35995931

ABSTRACT

Lysyl hydroxylase 2 (LH2) is a member of LH family that catalyzes the hydroxylation of lysine (Lys) residues on collagen, and this particular isozyme has been implicated in various diseases. While its function as a telopeptidyl LH is generally accepted, several fundamental questions remain unanswered: 1. Does LH2 catalyze the hydroxylation of all telopeptidyl Lys residues of collagen? 2. Is LH2 involved in the helical Lys hydroxylation? 3. What are the functional consequences when LH2 is completely absent? To answer these questions, we generated LH2-null MC3T3 cells (LH2KO), and extensively characterized the type I collagen phenotypes in comparison with controls. Cross-link analysis demonstrated that the hydroxylysine-aldehyde (Hylald)-derived cross-links were completely absent from LH2KO collagen with concomitant increases in the Lysald-derived cross-links. Mass spectrometric analysis revealed that, in LH2KO type I collagen, telopeptidyl Lys hydroxylation was completely abolished at all sites while helical Lys hydroxylation was slightly diminished in a site-specific manner. Moreover, di-glycosylated Hyl was diminished at the expense of mono-glycosylated Hyl. LH2KO collagen was highly soluble and digestible, fibril diameters were diminished, and mineralization impaired when compared to controls. Together, these data underscore the critical role of LH2-catalyzed collagen modifications in collagen stability, organization and mineralization in MC3T3 cells.


Subject(s)
Collagen Type I , Procollagen-Lysine, 2-Oxoglutarate 5-Dioxygenase , Collagen/metabolism , Collagen Type I/metabolism , Hydroxylation , Lysine/metabolism , Procollagen-Lysine, 2-Oxoglutarate 5-Dioxygenase/genetics , Procollagen-Lysine, 2-Oxoglutarate 5-Dioxygenase/metabolism , Protein Processing, Post-Translational
4.
Adv Redox Res ; 42022 Apr.
Article in English | MEDLINE | ID: mdl-35515703

ABSTRACT

Vitamin B12 (B12) is an essential co-factor for two enzymes in mammalian metabolism and can also act as a mimetic of superoxide dismutase (SOD) converting superoxide (O2 •‒) to hydrogen peroxide (H2O2). High oral dose B12 decreases renal O2 •‒ and post-ischemia/reperfusion injury in mice and protects against damage induced by hypoxia/reperfusion in mouse kidney proximal tubular cells (BU.MPT). O2 •‒ is unstable and rapidly converted to H2O2. H2O2 mediates oxidative stress associated with O2 •‒. Whether B12 protects against damage induced by H2O2 is unknown. Both BU.MPT cells and mouse brain endothelial cells (bEdn.3) were applied to test the effects of B12 on H2O2-induced cytotoxicity. Both types of cells were treated with different doses of H2O2 with or without different doses of B12. Cell viability was analyzed 24 h later. H2O2 caused cell death only at a very high dose, and high pharmacological dose of B12 did not prevent this detrimental effect in either cell type. In bEnd.3 cells, transcriptional levels of heme oxygenase-1 (HO-1) increased, while nuclear factor erythroid 2-related factor 2 (Nrf2) decreased by H2O2. The levels of transcripts were not affected by the B12 treatment. We conclude that the cytotoxic effects of exogenous H2O2 in BU.MPT and bEdn.3 cells are not prevented by B12.

5.
Front Cardiovasc Med ; 9: 818662, 2022.
Article in English | MEDLINE | ID: mdl-35360009

ABSTRACT

We have previously identified a novel atherosclerosis quantitative trait locus (QTL), Arch atherosclerosis 5 (Aath5), on mouse chromosome 10 by three-way QTL analyses between Apoe -/- mice on a DBA/2J, 129S6 and C57BL/6J background. The DBA/2J haplotype at the Aath5 locus was associated with smaller plaque size. One of the candidate genes underlying Aath5 was Stabilin-2 (Stab2), which encodes a clearance receptor for hyaluronan (HA) predominantly expressed in liver sinusoidal endothelial cells (LSECs). However, the role of Stab2 in atherosclerosis is unknown. A congenic line of Apoe -/- mice carrying Aath5 covering the Stab2 DBA allele on a background of 129S6 confirmed the small reductions of atherosclerotic plaque development. To further determine whether Stab2 is an underlying gene for Aath5, we generated Stab2 -/- Apoe -/- mice on a C57BL/6J background. When fed with a Western diet for 8 weeks, Stab2 -/- Apoe -/- males developed approximately 30% smaller plaques than Stab2 +/+ Apoe -/- mice. HA was accumulated in circulation but not in major organs in the Stab2 deficient mice. STAB2-binding molecules that are involved in atherosclerosis, including acLDL, apoptotic cells, heparin and vWF were not likely the direct cause of the protection in the Stab2 -/- Apoe -/- males. These data indicate that reduction of Stab2 is protective against atherosclerotic plaque development, and that Stab2 is a contributing gene underlying Aath5, although its effect is small. To test whether non-synonymous amino acid changes unique to DBA/2J affect the function of STAB2 protein, we made HEK293 cell lines expressing STAB2129 or STAB2DBA proteins, as well as STAB2129 proteins carrying each of five DBA-unique replacements that have been predicted to be deleterious. These mutant cells were capable of internalizing 125I -HA and DiI-acLDL similarly to the control cells. These results indicate that the amino acid changes unique to DBA/2J are not affecting the function of STAB2 protein, and support our previous observation that the reduced transcription of Stab2 in the liver sinusoid as a consequence of the insertion of a viral-derived sequence, intracisternal A particle, is the primary contributor to the athero-protection conferred by the DBA/2J allele.

6.
Genes (Basel) ; 11(12)2020 11 28.
Article in English | MEDLINE | ID: mdl-33260687

ABSTRACT

Atherosclerosis in different vascular locations leads to distinct clinical consequences, such as ischemic stroke and myocardial infarction. Genome-wide association studies in humans revealed that genetic loci responsible for carotid plaque and coronary artery disease were not overlapping, suggesting that distinct genetic pathways might be involved for each location. While elevated plasma cholesterol is a common risk factor, plaque development in different vascular beds is influenced by hemodynamics and intrinsic vascular integrity. Despite the limitation of species differences, mouse models provide platforms for unbiased genetic approaches. Mouse strain differences also indicate that susceptibility to atherosclerosis varies, depending on vascular locations, and that the location specificity is genetically controlled. Quantitative trait loci analyses in mice suggested candidate genes, including Mertk and Stab2, although how each gene affects the location-specific atherosclerosis needs further elucidation. Another unbiased approach of single-cell transcriptome analyses revealed the presence of a small subpopulation of vascular smooth muscle cells (VSMCs), which are "hyper-responsive" to inflammatory stimuli. These cells are likely the previously-reported Sca1+ progenitor cells, which can differentiate into multiple lineages in plaques. Further spatiotemporal analyses of the progenitor cells are necessary, since their distribution pattern might be associated with the location-dependent plaque development.


Subject(s)
Atherosclerosis , Muscle, Smooth, Vascular/metabolism , Myocytes, Smooth Muscle/metabolism , Plaque, Atherosclerotic , Animals , Atherosclerosis/genetics , Atherosclerosis/metabolism , Cell Adhesion/genetics , Genome-Wide Association Study , Mice , Plaque, Atherosclerotic/genetics , Plaque, Atherosclerotic/metabolism
7.
Mamm Genome ; 31(1-2): 2-16, 2020 02.
Article in English | MEDLINE | ID: mdl-31912264

ABSTRACT

Stabilin2 (Stab2) encodes a large transmembrane protein which is predominantly expressed in the liver sinusoidal endothelial cells (LSECs) and functions as a scavenger receptor for various macromolecules including hyaluronans (HA). In DBA/2J mice, plasma HA concentration is ten times higher than in 129S6 or C57BL/6J mice, and this phenotype is genetically linked to the Stab2 locus. Stab2 mRNA in the LSECs was significantly lower in DBA/2J than in 129S6, leading to reduced STAB2 proteins in the DBA/2J LSECs. We found a retrovirus-derived transposable element, intracisternal A particle (IAP), in the promoter region of Stab2DBA which likely interferes with normal expression in the LSECs. In contrast, in other tissues of DBA/2J mice, the IAP drives high ectopic Stab2DBA transcription starting within the 5' long terminal repeat of IAP in a reverse orientation and continuing through the downstream Stab2DBA. Ectopic transcription requires the Stab2-IAP element but is dominantly suppressed by the presence of loci on 59.7-73.0 Mb of chromosome (Chr) 13 from C57BL/6J, while the same region in 129S6 requires additional loci for complete suppression. Chr13:59.9-73 Mb contains a large number of genes encoding Krüppel-associated box-domain zinc-finger proteins that target transposable elements-derived sequences and repress their expression. Despite the high amount of ectopic Stab2DBA transcript in tissues other than liver, STAB2 protein was undetectable and unlikely to contribute to the plasma HA levels of DBA/2J mice. Nevertheless, the IAP insertion and its effects on the transcription of the downstream Stab2DBA exemplify that stochastic evolutional events could significantly influence susceptibility to complex but common diseases.


Subject(s)
Cell Adhesion Molecules, Neuronal/genetics , Ectopic Gene Expression , Genes, Intracisternal A-Particle/genetics , Alleles , Animals , Cell Adhesion Molecules, Neuronal/metabolism , Chromosomes, Mammalian/genetics , Chromosomes, Mammalian/metabolism , DNA Methylation , Endothelial Cells , Genetic Variation , HEK293 Cells , Humans , Hyaluronic Acid/blood , Liver/cytology , Liver/metabolism , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Inbred DBA , Promoter Regions, Genetic
8.
Biochemistry ; 58(50): 5040-5051, 2019 12 17.
Article in English | MEDLINE | ID: mdl-31726007

ABSTRACT

Glycosylation in type I collagen occurs as O-linked galactosyl- (G-) lesser and glucosylgalactosyl-hydroxylysine (GG-Hyl); however, its biological significance is still not well understood. To investigate the function of this modification in bone, we have generated preosteoblast MC3T3-E1 (MC)-derived clones, short hairpin (Sh) clones, in which Glt25d1 gene expression was stably suppressed. In Sh clones, the GLT25D1 protein levels were markedly diminished in comparison to controls (MC and those transfected with the empty vector). In Sh collagen, levels of both G- and GG-Hyl were significantly diminished with a concomitant increase in the level of free-Hyl. In addition, the level of immature divalent cross-links significantly diminished while the level of the mature trivalent cross-link increased. As determined by mass spectrometric analysis, seven glycosylation sites were identified in type I collagen and the most predominant site was at the helical cross-linking site, α1-87. At all of the glycosylation sites, the relative levels of G- and GG-Hyl were markedly diminished, i.e., by ∼50-75%, in Sh collagen, and at five of these sites, the level of Lys hydroxylation was significantly increased. The collagen fibrils in Sh clones were larger, and mineralization was impaired. These results indicate that GLT25D1 catalyzes galactosylation of Hyl throughout the type I collagen molecule and that this modification may regulate maturation of collagen cross-linking, fibrillogenesis, and mineralization.


Subject(s)
Collagen Type I/metabolism , Galactosyltransferases/metabolism , Phenotype , 3T3 Cells , Animals , Biocatalysis , Collagen Type I/chemistry , Glycosylation , Lysine/metabolism , Mice
9.
PLoS Genet ; 15(6): e1008196, 2019 06.
Article in English | MEDLINE | ID: mdl-31173582

ABSTRACT

Covalent intermolecular cross-linking of collagen is essential for tissue stability. Recent studies have demonstrated that cyclophilin B (CypB), an endoplasmic reticulum (ER)-resident peptidyl-prolyl cis-trans isomerase, modulates lysine (Lys) hydroxylation of type I collagen impacting cross-linking chemistry. However, the extent of modulation, the molecular mechanism and the functional outcome in tissues are not well understood. Here, we report that, in CypB null (KO) mouse skin, two unusual collagen cross-links lacking Lys hydroxylation are formed while neither was detected in wild type (WT) or heterozygous (Het) mice. Mass spectrometric analysis of type I collagen showed that none of the telopeptidyl Lys was hydroxylated in KO or WT/Het mice. Hydroxylation of the helical cross-linking Lys residues was almost complete in WT/Het but was markedly diminished in KO. Lys hydroxylation at other sites was also lower in KO but to a lesser extent. A key glycosylation site, α1(I) Lys-87, was underglycosylated while other sites were mostly overglycosylated in KO. Despite these findings, lysyl hydroxylases and glycosyltransferase 25 domain 1 levels were significantly higher in KO than WT/Het. However, the components of ER chaperone complex that positively or negatively regulates lysyl hydroxylase activities were severely reduced or slightly increased, respectively, in KO. The atomic force microscopy-based nanoindentation modulus were significantly lower in KO skin than WT. These data demonstrate that CypB deficiency profoundly affects Lys post-translational modifications of collagen likely by modulating LH chaperone complexes. Together, our study underscores the critical role of CypB in Lys modifications of collagen, cross-linking and mechanical properties of skin.


Subject(s)
Cyclophilins/chemistry , Lysine/chemistry , Procollagen-Lysine, 2-Oxoglutarate 5-Dioxygenase/chemistry , Skin/enzymology , Animals , Collagen Type I/biosynthesis , Collagen Type I/genetics , Cyclophilins/genetics , Cyclophilins/ultrastructure , Endoplasmic Reticulum/chemistry , Endoplasmic Reticulum/enzymology , Glycosylation , Heterozygote , Hydroxylation , Lysine/genetics , Mass Spectrometry , Mice , Mice, Knockout , Microscopy, Atomic Force , Procollagen-Lysine, 2-Oxoglutarate 5-Dioxygenase/genetics , Protein Processing, Post-Translational/genetics , Skin/chemistry
10.
JCI Insight ; 4(12)2019 06 20.
Article in English | MEDLINE | ID: mdl-31217360

ABSTRACT

Engulfment and cell motility protein 1 (ELMO1) is part of a guanine nucleotide exchange factor for Ras-related C3 botulinum toxin substrate (Rac), and ELMO1 polymorphisms were identified to be associated with diabetic nephropathy in genome-wide association studies. We generated a set of Akita Ins2C96Y diabetic mice having 5 graded cardiac mRNA levels of ELMO1 from 30% to 200% of normal and found that severe dilated cardiomyopathy develops in ELMO1-hypermorphic mice independent of renal function at age 16 weeks, whereas ELMO1-hypomorphic mice were completely protected. As ELMO1 expression increased, reactive oxygen species indicators, dissociation of the intercalated disc, mitochondrial fragmentation/dysfunction, cleaved caspase-3 levels, and actin polymerization increased in hearts from Akita mice. Cardiomyocyte-specific overexpression in otherwise ELMO1-hypomorphic Akita mice was sufficient to promote cardiomyopathy. Cardiac Rac1 activity was positively correlated with the ELMO1 levels, and oral administration of a pan-Rac inhibitor, EHT1864, partially mitigated cardiomyopathy of the ELMO1 hypermorphs. Disrupting Nox4, a Rac-independent NADPH oxidase, also partially mitigated it. In contrast, a pan-NADPH oxidase inhibitor, VAS3947, markedly prevented cardiomyopathy. Our data demonstrate that in diabetes mellitus ELMO1 is the "rate-limiting" factor of reactive oxygen species production via both Rac-dependent and Rac-independent NADPH oxidases, which in turn trigger cellular signaling cascades toward cardiomyopathy.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Diabetic Cardiomyopathies/etiology , Reactive Oxygen Species/metabolism , rac GTP-Binding Proteins/metabolism , Adaptor Proteins, Signal Transducing/genetics , Animals , Cell Movement , Connexin 43/metabolism , Diabetic Cardiomyopathies/genetics , Diabetic Cardiomyopathies/metabolism , Diabetic Cardiomyopathies/pathology , Female , Heart/physiopathology , Male , Mice , Mitochondria/metabolism , Mitochondria/pathology , Myocardium/metabolism , Myocytes, Cardiac/metabolism , NADPH Oxidase 4/metabolism
11.
PLoS One ; 12(8): e0182882, 2017.
Article in English | MEDLINE | ID: mdl-28837567

ABSTRACT

Quantitative trait locus (QTL) analyses of intercross populations between widely used mouse inbred strains provide a powerful approach for uncovering genetic factors that influence susceptibility to atherosclerosis. Epistatic interactions are common in complex phenotypes and depend on genetic backgrounds. To dissect genetic architecture of atherosclerosis, we analyzed F2 progeny from a cross between apolipoprotein E-null mice on DBA/2J (DBA-apoE) and C57BL/6J (B6-apoE) genetic backgrounds and compared the results with those from two previous F2 crosses of apolipoprotein E-null mice on 129S6/SvEvTac (129-apoE) and DBA-apoE backgrounds, and B6-apoE and 129-apoE backgrounds. In these round-robin crosses, in which each parental strain was crossed with two others, large-effect QTLs are expected to be detectable at least in two crosses. On the other hand, observation of QTLs in one cross only may indicate epistasis and/or absence of statistical power. For atherosclerosis at the aortic arch, Aath4 on chromosome (Chr)2:66 cM follows the first pattern, with significant QTL peaks in (DBAx129)F2 and (B6xDBA)F2 mice but not in (B6x129)F2 mice. We conclude that genetic variants unique to DBA/2J at Aath4 confer susceptibility to atherosclerosis at the aortic arch. A similar pattern was observed for Aath5 on chr10:35 cM, verifying that the variants unique to DBA/2J at this locus protect against arch plaque development. However, multiple loci, including Aath1 (Chr1:49 cM), and Aath2 (Chr1:70 cM) follow the second type of pattern, showing significant peaks in only one of the three crosses (B6-apoE x 129-apoE). As for atherosclerosis at aortic root, the majority of QTLs, including Ath29 (Chr9:33 cM), Ath44 (Chr1:68 cM) and Ath45 (Chr2:83 cM), was also inconsistent, being significant in only one of the three crosses. Only the QTL on Chr7:37 cM was consistently suggestive in two of the three crosses. Thus QTL analysis of round-robin crosses revealed the genetic architecture of atherosclerosis.


Subject(s)
Apolipoproteins E/genetics , Atherosclerosis/genetics , Quantitative Trait Loci , Animals , Aorta/pathology , Atherosclerosis/pathology , Chromosome Mapping , Crosses, Genetic , Lipids/blood , Mice , Mice, Inbred C57BL , Mice, Inbred DBA , Mice, Transgenic , Plaque, Atherosclerotic/pathology
12.
Arterioscler Thromb Vasc Biol ; 37(7): e82-e91, 2017 07.
Article in English | MEDLINE | ID: mdl-28473436

ABSTRACT

OBJECTIVE: Arch atherosclerosis 4 (Aath4) is a quantitative trait locus for atherosclerotic plaque formation in the inner curve of the aortic arch previously identified in an F2 cross of Apoe-/- mice on DBA/2J and 129S6 backgrounds. C-mer proto-oncogene tyrosine kinase (Mertk), coding for a ligand-activated transmembrane tyrosine kinase, is a candidate gene within the same chromosomal region. Our objective was to determine whether strain differences in Mertk influence plaque formation. APPROACH AND RESULTS: To dissect the strain effects of Mertk on atherosclerosis, we first established a congenic mouse line (Aath4aDBA/DBA ) in which a 5' region of Aath4 of DBA/2J, including Mertk, was backcrossed onto a 129S6-Apoe-/- background. The resulting Aath4aDBA/DBA male mice developed significantly larger plaques compared with control mice (Aath4a129/129 ), proving that the DBA/2J allele of Aath4a is proatherogenic. Thioglycollate-elicited peritoneal macrophages from Aath4aDBA/DBA mice express less than 50% of Mertk mRNA and cell-surface MERTK protein compared with those from the control mice. Moreover, both large and small peritoneal Aath4aDBA/DBA macrophages showed reduced phagocytosis of apoptotic cells. When Mertk cDNAs from 129S6 and DBA/2J mice were overexpressed in HEK293T (human embryonic kidney 293T) cells, phagocytosis of apoptotic cells was equally enhanced in direct proportion to Mertk levels, indicating that phagocytosis is modulated by the amount of MERTK, but that it is not affected by MERTK amino acid differences between 129S6 and DBA/2J. CONCLUSIONS: Reduced transcription of Mertk, rather than differences in MERTK protein structure, determines the reduced efficiency of apoptotic cell clearance in the Aath4aDBA/DBA mice, which, in turn, contributes to their increased susceptibility to atherosclerosis.


Subject(s)
Aorta, Thoracic/enzymology , Aortic Diseases/genetics , Atherosclerosis/genetics , Chromosomes, Mammalian , Haplotypes , Macrophages, Peritoneal/enzymology , Phagocytosis , Plaque, Atherosclerotic , Proto-Oncogene Proteins/metabolism , Receptor Protein-Tyrosine Kinases/metabolism , Animals , Aorta, Thoracic/pathology , Aortic Diseases/enzymology , Aortic Diseases/pathology , Apolipoproteins E/deficiency , Apolipoproteins E/genetics , Apoptosis , Atherosclerosis/enzymology , Atherosclerosis/pathology , Disease Models, Animal , Down-Regulation , Gene Expression Regulation, Enzymologic , Genetic Predisposition to Disease , HEK293 Cells , Humans , Jurkat Cells , Macrophages, Peritoneal/pathology , Mice, 129 Strain , Mice, Congenic , Mice, Inbred DBA , Mice, Knockout , Phenotype , Promoter Regions, Genetic , Proto-Oncogene Mas , Proto-Oncogene Proteins/deficiency , Proto-Oncogene Proteins/genetics , Receptor Protein-Tyrosine Kinases/deficiency , Receptor Protein-Tyrosine Kinases/genetics , Risk Factors , Species Specificity , Transcription, Genetic , Transfection , Vascular Calcification/enzymology , Vascular Calcification/genetics , Vascular Calcification/pathology , c-Mer Tyrosine Kinase
13.
PLoS One ; 10(2): e0117478, 2015.
Article in English | MEDLINE | ID: mdl-25689165

ABSTRACT

The genetic background of apolipoprotein E (apoE) deficient mice influences atherosclerotic plaque development. We previously reported three quantitative trait loci (QTL), Aath1-Aath3, that affect aortic arch atherosclerosis independently of those in the aortic root in a cross between C57BL6 apoEKO mice (B6-apoE) and 129S6 apoEKO mice (129-apoE). To gain further insight into genetic factors that influence atherosclerosis at different vascular locations, we analyzed 335 F2 mice from an intercross between 129-apoE and apoEKO mice on a DBA/2J genetic background (DBA-apoE). The extent of atherosclerosis in the aortic arch was very similar in the two parental strains. Nevertheless, a genome-wide scan identified two significant QTL for plaque size in the aortic arch: Aath4 on Chromosome (Chr) 2 at 137 Mb and Aath5 on Chr 10 at 51 Mb. The DBA alleles of Aath4 and Aath5 respectively confer susceptibility and resistance to aortic arch atherosclerosis over 129 alleles. Both QTL are also independent of those affecting plaque size at the aortic root. Genome analysis suggests that athero-susceptibility of Aath4 in DBA may be contributed by multiple genes, including Mertk and Cd93, that play roles in phagocytosis of apoptotic cells and modulate inflammation. A candidate gene for Aath5 is Stab2, the DBA allele of which is associated with 10 times higher plasma hyaluronan than the 129 allele. Overall, our identification of two new QTL that affect atherosclerosis in an aortic arch-specific manner further supports the involvement of distinct pathological processes at different vascular locations.


Subject(s)
Aorta, Thoracic/pathology , Aortic Diseases/genetics , Apolipoproteins E/genetics , Atherosclerosis/genetics , Quantitative Trait Loci , Alleles , Animals , Aortic Diseases/pathology , Atherosclerosis/pathology , Crosses, Genetic , Genetic Predisposition to Disease , Haplotypes , Male , Mice , Mice, Inbred C57BL , Mice, Inbred DBA , Mice, Knockout , Phenotype
14.
PLoS One ; 9(2): e88274, 2014.
Article in English | MEDLINE | ID: mdl-24586312

ABSTRACT

Apolipoprotein E-null mice on a DBA/2J genetic background (DBA-apoE) are highly susceptible to atherosclerosis in the aortic root area compared with those on a 129S6 background (129-apoE). To explore atherosclerosis-responsible genetic regions, we performed a quantitative trait locus (QTL) analysis using 172 male and 137 female F2 derived from an intercross between DBA-apoE and 129-apoE mice. A genome-wide scan identified two significant QTL for the size of lesions at the root: one is Ath44 on Chromosome (Chr) 1 at 158 Mb, and the other Ath45 on Chr 2 at 162 Mb. Ath44 co-localizes with but appears to be independent of a previously reported QTL, Ath1, while Ath45 is a novel QTL. DBA alleles of both Ath44 and Ath45 confer atherosclerosis-susceptibility. In addition, a QTL on Chr 14 at 73 Mb was found significant only in males, and 129 allele conferring susceptibility. Further analysis detected female-specific interactions between a second QTL on Chr 1 at 73 Mb and a QTL on Chr 3 at 21 Mb, and between Chr 7 at 84 Mb and Chr 12 at 77 Mb. These loci for the root atherosclerosis were independent of QTLs for plasma total cholesterol and QTLs for triglycerides, but a QTL for HDL (Chr 1 at 126 Mb) overlapped with the Ath44. Notably, haplotype analysis among 129S6, DBA/2J and C57BL/6 genomes and their gene expression data narrowed the candidate regions for Ath44 and Ath45 to less than 5 Mb intervals where multiple genome wide associations with cardiovascular phenotypes have also been reported in humans. SNPs in or near Fmo3, Sele and Selp for Ath44, and Lbp and Pkig for Ath45 were suggested for further investigation as potential candidates underlying the atherosclerosis susceptibility.


Subject(s)
Aorta/pathology , Apolipoproteins E/genetics , Atherosclerosis/genetics , Genetic Predisposition to Disease/genetics , Quantitative Trait Loci/genetics , Analysis of Variance , Animals , Atherosclerosis/pathology , Chromosome Mapping , Crosses, Genetic , Female , Haplotypes/genetics , Male , Mice , Mice, Inbred Strains , Microarray Analysis , Microsatellite Repeats/genetics , Sex Factors
15.
Proc Natl Acad Sci U S A ; 110(14): 5600-5, 2013 Apr 02.
Article in English | MEDLINE | ID: mdl-23503843

ABSTRACT

To uncover the potential cardiovascular effects of human polymorphisms influencing transforming growth factor ß1 (TGFß1) expression, we generated mice with Tgfb1 mRNA expression graded in five steps from 10% to 300% normal. Adrenal expression of the genes for mineralocorticoid-producing enzymes ranged from 50% normal in the hypermorphs at age 12 wk to 400% normal in the hypomorphs accompanied with proportionate changes in plasma aldosterone levels, whereas plasma volumes ranged from 50% to 150% normal accompanied by marked compensatory changes in plasma angiotensin II and renin levels. The aldosterone/renin ratio ranged from 0.3 times normal in the 300% hypermorphs to six times in the 10% hypomorphs, which have elevated blood pressure. Urinary output of water and electrolytes are markedly decreased in the 10% hypomorphs without significant change in the glomerular filtration rate. Renal activities for the Na(+), K(+)-ATPase, and epithelial sodium channel are markedly increased in the 10% hypomorphs. The hypertension in the 10% hypomorphs is corrected by spironolactone or amiloride at doses that do not change blood pressure in wild-type mice. Thus, changes in Tgfb1 expression cause marked progressive changes in multiple systems that regulate blood pressure and fluid homeostasis, with the major effects being mediated by changes in adrenocortical function.


Subject(s)
Aldosterone/blood , Gene Expression Regulation/physiology , Hyperaldosteronism/etiology , Natriuresis/physiology , Transforming Growth Factor beta1/metabolism , Amiloride/pharmacology , Angiotensin II/blood , Animals , Blood Pressure/drug effects , DNA Primers/genetics , Gene Expression Regulation/genetics , Glomerular Filtration Rate/physiology , Hyperaldosteronism/metabolism , Mice , Mice, Inbred C57BL , Polymerase Chain Reaction , Renin/blood , Sodium-Potassium-Exchanging ATPase/metabolism , Spironolactone/pharmacology , Transforming Growth Factor beta1/genetics , Urinalysis
16.
Curr Opin Nephrol Hypertens ; 21(1): 92-6, 2012 Jan.
Article in English | MEDLINE | ID: mdl-22048723

ABSTRACT

PURPOSE OF REVIEW: The kallikrein-kinin system (KKS) constitutes a complex multienzyme cascade that produces several bioactive kinin peptides and their derivatives including bradykinin. In addition to the classical notion of the KKS as a potent vasodilator and a mediator of inflammatory responses, recent studies suggest a link between the KKS and oxidative stress. A number of established mouse models with altered levels of KKS components opened the way to evaluate precise functions of the KKS. Here we review recent findings on the role of the KKS in cardiovascular diseases and chronic kidney diseases, and discuss potential benefits of KKS activation in these diseases. RECENT FINDINGS: Deletion of both B1R and B2R in a diabetic mouse model exacerbates its renal phenotypes, suggesting that the KKS exerts protective effects on diabetic nephropathy by suppressing oxidative stress, presumably via nitric oxide and prostaglandins. SUMMARY: Accumulating evidence has highlighted the importance of the KKS as a protective system against oxidative stress and organ damage in the heart and kidney. The activation of the KKS by angiotensin I-converting enzyme inhibitors and vasopeptidase inhibitors is likely to be beneficial in senescence-associated cardiovascular diseases and chronic kidney diseases.


Subject(s)
Cardiovascular Diseases/enzymology , Kallikrein-Kinin System , Kallikreins/metabolism , Kidney Diseases/enzymology , Kinins/metabolism , Oxidative Stress , Angiotensin-Converting Enzyme Inhibitors/therapeutic use , Animals , Cardiovascular Diseases/drug therapy , Enzyme Activation , Humans , Kallikrein-Kinin System/drug effects , Kidney Diseases/drug therapy , Nitric Oxide/metabolism , Oxidative Stress/drug effects , Peptidyl-Dipeptidase A/metabolism , Prostaglandins/metabolism , Receptors, Bradykinin/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL