Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 165
Filter
1.
Front Neurosci ; 17: 1149603, 2023.
Article in English | MEDLINE | ID: mdl-37456993

ABSTRACT

Adult neural stem cells (NSCs) in the mouse subventricular zone (SVZ) serve as a lifelong reservoir for newborn olfactory bulb neurons. Recent studies have identified a slowly dividing subpopulation of embryonic neural stem-progenitor cells (NPCs) as the embryonic origin of adult NSCs. Yet, little is known about how these slowly dividing embryonic NPCs are maintained until adulthood while other NPCs are extinguished by the completion of brain development. The extracellular matrix (ECM) is an essential component of stem cell niches and thus a key determinant of stem cell fate. Here we investigated tissue inhibitors of metalloproteinases (TIMPs)-regulators of ECM remodeling-for their potential roles in the establishment of adult NSCs. We found that Timp2, Timp3, and Timp4 were expressed at high levels in slowly dividing NPCs compared to rapidly dividing NPCs. Deletion of TIMP3 reduced the number of adult NSCs and neuroblasts in the lateral SVZ. In addition, overexpression of TIMP3 in the embryonic NPCs suppressed neuronal differentiation and upregulated the expression levels of Notch signaling relating genes. These results thus suggest that TIMP3 keeps the undifferentiated state of embryonic NPCs, leading to the establishment and maintenance of adult NSCs.

2.
Int J Mol Sci ; 24(4)2023 Feb 15.
Article in English | MEDLINE | ID: mdl-36835312

ABSTRACT

Peroxisome proliferator-activated receptor γ (PPARγ) gene mutations in humans and mice lead to whole-body insulin resistance and partial lipodystrophy. It is unclear whether preserved fat depots in partial lipodystrophy are beneficial for whole-body metabolic homeostasis. We analyzed the insulin response and expression of metabolic genes in the preserved fat depots of PpargC/- mice, a familial partial lipodystrophy type 3 (FPLD3) mouse model resulting from a 75% decrease in Pparg transcripts. Perigonadal fat of PpargC/- mice in the basal state showed dramatic decreases in adipose tissue mass and insulin sensitivity, whereas inguinal fat showed compensatory increases. Preservation of inguinal fat metabolic ability and flexibility was reflected by the normal expression of metabolic genes in the basal or fasting/refeeding states. The high nutrient load further increased insulin sensitivity in inguinal fat, but the expression of metabolic genes became dysregulated. Inguinal fat removal resulted in further impairment of whole-body insulin sensitivity in PpargC/- mice. Conversely, the compensatory increase in insulin sensitivity of the inguinal fat in PpargC/- mice diminished as activation of PPARγ by its agonists restored insulin sensitivity and metabolic ability of perigonadal fat. Together, we demonstrated that inguinal fat of PpargC/- mice plays a compensatory role in combating perigonadal fat abnormalities.


Subject(s)
Insulin Resistance , Lipodystrophy, Familial Partial , PPAR gamma , Animals , Humans , Mice , Insulin/metabolism , Insulin/pharmacology , Insulin Resistance/genetics , Lipodystrophy, Familial Partial/genetics , Mutation , PPAR gamma/genetics , PPAR gamma/metabolism
4.
Front Neurosci ; 16: 848654, 2022.
Article in English | MEDLINE | ID: mdl-35784847

ABSTRACT

Spatial navigation and orientation are emerging as promising markers for altered cognition in prodromal Alzheimer's disease, and even in cognitively normal individuals at risk for Alzheimer's disease. The different APOE gene alleles confer various degrees of risk. The APOE2 allele is considered protective, APOE3 is seen as control, while APOE4 carriage is the major known genetic risk for Alzheimer's disease. We have used mouse models carrying the three humanized APOE alleles and tested them in a spatial memory task in the Morris water maze. We introduce a new metric, the absolute winding number, to characterize the spatial search strategy, through the shape of the swim path. We show that this metric is robust to noise, and works for small group samples. Moreover, the absolute winding number better differentiated APOE3 carriers, through their straighter swim paths relative to both APOE2 and APOE4 genotypes. Finally, this novel metric supported increased vulnerability in APOE4 females. We hypothesized differences in spatial memory and navigation strategies are linked to differences in brain networks, and showed that different genotypes have different reliance on the hippocampal and caudate putamen circuits, pointing to a role for white matter connections. Moreover, differences were most pronounced in females. This departure from a hippocampal centric to a brain network approach may open avenues for identifying regions linked to increased risk for Alzheimer's disease, before overt disease manifestation. Further exploration of novel biomarkers based on spatial navigation strategies may enlarge the windows of opportunity for interventions. The proposed framework will be significant in dissecting vulnerable circuits associated with cognitive changes in prodromal Alzheimer's disease.

5.
Vascul Pharmacol ; 133-134: 106777, 2020.
Article in English | MEDLINE | ID: mdl-32750408

ABSTRACT

Atherosclerosis is a systemic chronic inflammatory disease. Many antioxidants including alpha-lipoic acid (LA), a product of lipoic acid synthase (Lias), have proven to be effective for treatment of this disease. However, the question remains whether LA regulates the immune response as a protective mechanism against atherosclerosis. We initially investigated whether enhanced endogenous antioxidant can retard the development of atherosclerosis via immunomodulation. To explore the impact of enhanced endogenous antioxidant on the retardation of atherosclerosis via immune regulation, our laboratory has recently created a double mutant mouse model, using apolipoprotein E-deficient (Apoe-/-) mice crossbred with mice overexpressing lipoic acid synthase gene (LiasH/H), designated as LiasH/HApoe-/- mice. Their littermates, Lias+/+Apoe-/- mice, served as a control. Distinct redox environments between the two strains of mice have been established and they can be used to facilitate identification of antioxidant targets in the immune response. At 6 months of age, LiasH/HApoe-/- mice had profoundly decreased atherosclerotic lesion size in the aortic sinus compared to their Lias+/+Apoe-/- littermates, accompanied by significantly enhanced numbers of regulatory T cells (Tregs) and anti-oxidized LDL autoantibody in the vascular system, and reduced T cell infiltrates in aortic walls. Our results represent a novel exploration into an environment with increased endogenous antioxidant and its ability to alleviate atherosclerosis, likely through regulation of the immune response. These outcomes shed light on a new therapeutic strategy using antioxidants to lessen atherosclerosis.


Subject(s)
Aorta/enzymology , Aortic Diseases/prevention & control , Atherosclerosis/prevention & control , Plaque, Atherosclerotic , Sulfurtransferases/biosynthesis , Animals , Aorta/immunology , Aorta/pathology , Aortic Diseases/enzymology , Aortic Diseases/immunology , Aortic Diseases/pathology , Atherosclerosis/enzymology , Atherosclerosis/immunology , Atherosclerosis/pathology , Autoantibodies/blood , Disease Models, Animal , Enzyme Induction , Lipoproteins, LDL/immunology , Male , Mice, Inbred C57BL , Mice, Knockout, ApoE , Oxidation-Reduction , Oxidative Stress , Sulfurtransferases/genetics , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism
7.
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
8.
Sci Rep ; 9(1): 11416, 2019 08 06.
Article in English | MEDLINE | ID: mdl-31388055

ABSTRACT

The development of cardiovascular disease is intimately linked to elevated levels of low-density lipoprotein (LDL) cholesterol in the blood. Hepatic LDL receptor (LDLR) levels regulate the amount of plasma LDL. We identified the secreted zinc metalloproteinase, bone morphogenetic protein 1 (BMP1), as responsible for the cleavage of human LDLR within its extracellular ligand-binding repeats at Gly171↓Asp172. The resulting 120 kDa membrane-bound C-terminal fragment (CTF) of LDLR had reduced capacity to bind LDL and when expressed in LDLR null cells had compromised LDL uptake as compared to the full length receptor. Pharmacological inhibition of BMP1 or siRNA-mediated knockdown prevented the generation of the 120 kDa CTF and resulted in an increase in LDL uptake into cells. The 120 kDa CTF was detected in the livers from humans and mice expressing human LDLR. Collectively, these results identify that BMP1 regulates cellular LDL uptake and may provide a target to modulate plasma LDL cholesterol.


Subject(s)
Bone Morphogenetic Protein 1/metabolism , Lipoproteins, LDL/metabolism , Receptors, LDL/metabolism , Animals , Atherosclerosis/blood , Atherosclerosis/drug therapy , Atherosclerosis/metabolism , Atherosclerosis/pathology , Biopsy , Bone Morphogenetic Protein 1/antagonists & inhibitors , Bone Morphogenetic Protein 1/genetics , CHO Cells , Cricetulus , Gene Knockdown Techniques , Hep G2 Cells , Humans , Lipoproteins, LDL/blood , Liver/chemistry , Liver/metabolism , Liver/pathology , Mice , Mice, Transgenic , Oxadiazoles/pharmacology , Proteolysis/drug effects , RNA, Small Interfering/metabolism , Receptors, LDL/analysis , Receptors, LDL/genetics , Recombinant Proteins/genetics , Recombinant Proteins/metabolism
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.
Arterioscler Thromb Vasc Biol ; 38(8): e145-e158, 2018 08.
Article in English | MEDLINE | ID: mdl-29880490

ABSTRACT

Objective- Dyslipidemia is a component of the metabolic syndrome, an established risk factor for atherosclerotic cardiovascular disease, and is also observed in various autoimmune and chronic inflammatory conditions. However, there are limited opportunities to study the impact of acquired dyslipidemia on cardiovascular and immune pathology. Approach and Results- We designed a model system that allows for the conversion to a state of acute hyperlipidemia in adult life, so that the consequences of such a transition could be observed, through conditionally deleting APOE (apolipoprotein E) in the adult mouse. The transition to hypercholesterolemia was accompanied by adaptive immune responses, including the expansion of T lymphocyte helper cell 1, T follicular helper cell, and T regulatory subsets and the formation of germinal centers. Unlike steady-state Apoe-/- mice, abrupt loss of APOE induced rapid production of antibodies recognizing rheumatoid disease autoantigens. Genetic ablation of the germinal center reduced both autoimmunity and atherosclerosis, indicating that the immune response that follows loss of APOE is independent of atherosclerosis but nevertheless promotes plaque development. Conclusions- Our findings suggest that immune activation in response to hyperlipidemia could contribute to a wide range of inflammatory autoimmune diseases, including atherosclerosis.


Subject(s)
Adaptive Immunity , Aorta/immunology , Aortic Diseases/immunology , Apolipoproteins E/immunology , Atherosclerosis/immunology , Autoimmunity , Dyslipidemias/immunology , Inflammation/immunology , Animals , Aorta/metabolism , Aorta/pathology , Aortic Diseases/genetics , Aortic Diseases/metabolism , Aortic Diseases/pathology , Apolipoproteins E/deficiency , Apolipoproteins E/genetics , Atherosclerosis/genetics , Atherosclerosis/metabolism , Atherosclerosis/pathology , B-Lymphocytes/immunology , B-Lymphocytes/metabolism , Cells, Cultured , Disease Models, Animal , Disease Progression , Dyslipidemias/genetics , Dyslipidemias/metabolism , Dyslipidemias/pathology , Germinal Center/immunology , Germinal Center/metabolism , Immunity, Humoral , Inflammation/genetics , Inflammation/metabolism , Inflammation/pathology , Mice, Inbred C57BL , Mice, Knockout, ApoE , Plaque, Atherosclerotic , Signal Transduction , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Time Factors
11.
Sci Rep ; 8(1): 6939, 2018 05 02.
Article in English | MEDLINE | ID: mdl-29720615

ABSTRACT

Patients born with congenital heart defects frequently encounter arrhythmias due to defects in the ventricular conduction system (VCS) development. Although recent studies identified transcriptional networks essential for the heart development, there is scant information on the mechanisms regulating VCS development. Based on the association of atrial natriuretic peptide (ANP) expression with VCS forming regions, it was reasoned that ANP could play a critical role in differentiation of cardiac progenitor cells (CPCs) and cardiomyocytes (CMs) toward a VCS cell lineage. The present study showed that treatment of embryonic ventricular cells with ANP or cell permeable 8-Br-cGMP can induce gene expression of important VCS markers such as hyperpolarization-activated cyclic nucleotide-gated channel-4 (HCN4) and connexin 40 (Cx40). Inhibition of protein kinase G (PKG) via Rp-8-pCPT-cGMPS further confirmed the role of ANP/NPRA/cGMP/PKG pathway in the regulation of HCN4 and Cx40 gene expression. Additional experiments indicated that ANP may regulate VCS marker gene expression by modulating levels of miRNAs that are known to control the stability of transcripts encoding HCN4 and Cx40. Genetic ablation of NPRA revealed significant decreases in VCS marker gene expression and defects in Purkinje fiber arborisation. These results provide mechanistic insights into the role of ANP/NPRA signaling in VCS formation.


Subject(s)
Atrial Natriuretic Factor/metabolism , Heart Conduction System/embryology , Heart Conduction System/metabolism , Signal Transduction , Animals , Biomarkers , Cell Differentiation , Cells, Cultured , Connexins/genetics , Connexins/metabolism , Fluorescent Antibody Technique , Gene Expression , Genes, Reporter , Genotype , Hyperpolarization-Activated Cyclic Nucleotide-Gated Channels/genetics , Hyperpolarization-Activated Cyclic Nucleotide-Gated Channels/metabolism , Mice , Mice, Knockout , Muscle Proteins/genetics , Muscle Proteins/metabolism , Myocytes, Cardiac/cytology , Myocytes, Cardiac/metabolism , Potassium Channels/genetics , Potassium Channels/metabolism , Protein Kinase Inhibitors/pharmacology
13.
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
14.
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
15.
Mol Neurodegener ; 12(1): 14, 2017 02 07.
Article in English | MEDLINE | ID: mdl-28173832

ABSTRACT

BACKGROUND: Alzheimer's disease (AD) is characterized by progressive cognitive decline and memory loss. Environmental factors and gene-environment interactions (GXE) may increase AD risk, accelerate cognitive decline, and impair learning and memory. However, there is currently little direct evidence supporting this hypothesis. METHODS: In this study, we assessed for a GXE between lead and ApoE4 on cognitive behavior using transgenic knock-in (KI) mice that express the human Apolipoprotein E4 allele (ApoE4-KI) or Apolipoprotein E3 allele (ApoE3-KI). We exposed 8-week-old male and female ApoE3-KI and ApoE4-KI mice to 0.2% lead acetate via drinking water for 12 weeks and assessed for cognitive behavior deficits during and after the lead exposure. In addition, we exposed a second (cellular) cohort of animals to lead and assessed for changes in adult hippocampal neurogenesis as a potential underlying mechanism for lead-induced learning and memory deficits. RESULTS: In the behavior cohort, we found that lead reduced contextual fear memory in all animals; however, this decrease was greatest and statistically significant only in lead-treated ApoE4-KI females. Similarly, only lead-treated ApoE4-KI females exhibited a significant decrease in spontaneous alternation in the T-maze. Furthermore, all lead-treated animals developed persistent spatial working memory deficits in the novel object location test, and this deficit manifested earlier in ApoE4-KI mice, with female ApoE4-KI mice exhibiting the earliest deficit onset. In the cellular cohort, we observed that the maturation, differentiation, and dendritic development of adult-born neurons in the hippocampus was selectively impaired in lead-treated female ApoE4-KI mice. CONCLUSIONS: These data suggest that GXE between ApoE4 and lead exposure may contribute to cognitive impairment and that impaired adult hippocampal neurogenesis may contribute to these deficits in cognitive behavior. Together, these data suggest a role for GXE and sex differences in AD risk.


Subject(s)
Alzheimer Disease/chemically induced , Alzheimer Disease/genetics , Apolipoprotein E4/genetics , Gene-Environment Interaction , Organometallic Compounds/toxicity , Animals , Behavior, Animal/drug effects , Brain/drug effects , Brain/pathology , Disease Models, Animal , Female , Gene Knock-In Techniques , Humans , Immunohistochemistry , Male , Mice , Mice, Transgenic
16.
Proc Natl Acad Sci U S A ; 113(47): 13450-13455, 2016 11 22.
Article in English | MEDLINE | ID: mdl-27821757

ABSTRACT

Preeclampsia (PE) complicates ∼5% of human pregnancies and is one of the leading causes of pregnancy-related maternal deaths. The only definitive treatment, induced delivery, invariably results in prematurity, and in severe early-onset cases may lead to fetal death. Many currently available antihypertensive drugs are teratogenic and therefore precluded from use. Nonteratogenic antihypertensives help control maternal blood pressure in PE, but results in preventing preterm delivery and correcting fetal growth restriction (FGR) that also occurs in PE have been disappointing. Here we show that dietary nicotinamide, a nonteratogenic amide of vitamin B3, improves the maternal condition, prolongs pregnancies, and prevents FGR in two contrasting mouse models of PE. The first is caused by endotheliosis due to excess levels in the mothers of a soluble form of the receptor for vascular endothelial growth factor (VEGF), which binds to and inactivates VEGF. The second is caused by genetic absence of Ankiryn-repeat-and-SOCS-box-containing-protein 4, a factor that contributes to the differentiation of trophoblast stem cells into the giant trophoblast cells necessary for embryo implantation in mice; its absence leads to impaired placental development. In both models, fetal production of ATP is impaired and FGR is observed. We show here that nicotinamide decreases blood pressure and endotheliosis in the mothers, probably by inhibiting ADP ribosyl cyclase (ADPRC), and prevents FGR, probably by normalizing fetal ATP synthesis via the nucleotide salvage pathway. Because nicotinamide benefits both dams and pups, it merits evaluation for preventing or treating PE in humans.


Subject(s)
Niacinamide/therapeutic use , Pre-Eclampsia/drug therapy , Abortion, Spontaneous/blood , Abortion, Spontaneous/physiopathology , Albuminuria/blood , Albuminuria/complications , Albuminuria/physiopathology , Animals , Animals, Newborn , Blood Pressure/drug effects , Body Weight/drug effects , Disease Models, Animal , Embryo Loss/drug therapy , Embryo Loss/prevention & control , Female , Fetal Growth Retardation/blood , Fetal Growth Retardation/drug therapy , Hypertension/blood , Hypertension/complications , Hypertension/physiopathology , Kidney/abnormalities , Kidney/drug effects , Kidney/pathology , Kidney/ultrastructure , Male , Mice, Inbred C57BL , Niacinamide/pharmacology , Organ Size/drug effects , Placenta/drug effects , Placenta/metabolism , Placenta/pathology , Placenta Growth Factor/blood , Pre-Eclampsia/blood , Pre-Eclampsia/pathology , Pre-Eclampsia/physiopathology , Pregnancy , Pregnancy Outcome , Suppressor of Cytokine Signaling Proteins/deficiency , Suppressor of Cytokine Signaling Proteins/metabolism , Uterus/drug effects , Uterus/pathology , Vascular Endothelial Growth Factor A/blood , Vascular Endothelial Growth Factor Receptor-1/blood , Vascular Endothelial Growth Factor Receptor-1/metabolism
17.
PLoS One ; 11(10): e0163208, 2016.
Article in English | MEDLINE | ID: mdl-27706190

ABSTRACT

Oxidative stress is implicated in the pathogenesis of diabetic nephropathy (DN) but outcomes of many clinical trials are controversial. To define the role of antioxidants in kidney protection during the development of diabetic nephropathy, we have generated a novel genetic antioxidant mouse model with over- or under-expression of lipoic acid synthase gene (Lias). These models have been mated with Ins2Akita/+ mice, a type I diabetic mouse model. We compare the major pathologic changes and oxidative stress status in two new strains of the mice with controls. Our results show that Ins2Akita/+ mice with under-expressed Lias gene, exhibit higher oxidative stress and more severe DN features (albuminuria, glomerular basement membrane thickening and mesangial matrix expansion). In contrast, Ins2Akita/+ mice with highly-expressed Lias gene display lower oxidative stress and less DN pathologic changes. Our study demonstrates that strengthening endogenous antioxidant capacity could be an effective strategy for prevention and treatment of DN.


Subject(s)
Diabetic Nephropathies/pathology , Sulfurtransferases/metabolism , 3' Untranslated Regions , Albumins/analysis , Animals , Blood Glucose/analysis , Blood Pressure , Chemokine CCL2/urine , Creatinine/urine , Diabetic Nephropathies/metabolism , Disease Models, Animal , Female , Gene Expression , Insulin/genetics , Insulin/metabolism , Kidney/pathology , Male , Mice , Mice, Inbred C57BL , Microscopy, Electron , Oxidative Stress , Sulfurtransferases/genetics
18.
Sci Rep ; 6: 26646, 2016 05 25.
Article in English | MEDLINE | ID: mdl-27221351

ABSTRACT

PPARγ modulates energy metabolism and inflammation. However, its specific functions in the balance of immunity in vivo have been explored incompletely. In this study, by the age of 14 mo, Pparg(C/-) mice with PPARγ expression at 25% of the normal level exhibited high autoantibody levels and developed mesangial proliferative glomerulonephritis, which resembled systemic lupus erythematosus (SLE)-like autoimmune disease. These symptoms were preceded by splenomegaly at an early age, which was associated with increases in splenocyte accumulation and B-cell activation but not with relocation of hematopoiesis to the spleen. The mechanism of splenic lymphocyte accumulation involved reduced sphingosine-1-phosphate receptor 1 (S1P1) expression and diminished migration toward S1P in the Pparg(C/-) splenocytes, which impeded lymphocyte egression. Mechanistically, increased Th17 polarization and IL-17 signaling in the Pparg(C/-) CD4(+) T cells contributed to B-cell hyperactivation in the spleen. Finally, the activation of the remaining PPARγ in Pparg(C/-) mice by pioglitazone increased S1P1 levels, reduced the Th17 population in the spleen, and ameliorated splenomegaly. Taken together, our data demonstrated that reduction of Pparg expression in T-helper cells is critical for spontaneous SLE-like autoimmune disease development; we also revealed a novel function of PPARγ in lymphocyte trafficking and cross talk between Th17 and B cells.


Subject(s)
B-Lymphocytes/immunology , Cell Movement/immunology , Gene Expression Regulation/immunology , Immune Tolerance , Immunity, Cellular , PPAR gamma/immunology , Th17 Cells/immunology , Animals , B-Lymphocytes/metabolism , B-Lymphocytes/pathology , Cell Movement/genetics , Lupus Erythematosus, Systemic/genetics , Lupus Erythematosus, Systemic/immunology , Lupus Erythematosus, Systemic/pathology , Mice , Mice, Inbred NOD , Mice, Knockout , Mice, SCID , PPAR gamma/biosynthesis , PPAR gamma/genetics , Th17 Cells/metabolism , Th17 Cells/pathology
19.
Hypertension ; 67(6): 1298-308, 2016 06.
Article in English | MEDLINE | ID: mdl-27045031

ABSTRACT

Loss of integrity and massive disruption of elastic fibers are key features of abdominal aortic aneurysm (AAA). Peroxisome proliferator-activated receptor γ (PPARγ) has been shown to attenuate AAA through inhibition of inflammation and proteolytic degradation. However, its involvement in elastogenesis during AAA remains unclear. PPARγ was highly expressed in human AAA within all vascular cells, including inflammatory cells and fibroblasts. In the aortas of transgenic mice expressing PPARγ at 25% normal levels (Pparg(C) (/-) mice), we observed the fragmentation of elastic fibers and reduced expression of vital elastic fiber components of elastin and fibulin-5. These were not observed in mice with 50% normal PPARγ expression (Pparg(+/-) mice). Infusion of a moderate dose of angiotensin II (500 ng/kg per minute) did not induce AAA but Pparg(+/-) aorta developed flattened elastic lamellae, whereas Pparg(C/-) aorta showed severe destruction of elastic fibers. After infusion of angiotensin II at 1000 ng/kg per minute, 73% of Pparg(C/-) mice developed atypical suprarenal aortic aneurysms: superior mesenteric arteries were dilated with extensive collagen deposition in adventitia and infiltrations of inflammatory cells. Although matrix metalloproteinase inhibition by doxycycline somewhat attenuated the dilation of aneurysm, it did not reduce the incidence nor elastic lamella deterioration in angiotensin II-infused Pparg(C/-) mice. Furthermore, PPARγ antagonism downregulated elastin and fibulin-5 in fibroblasts, but not in vascular smooth muscle cells. Chromatin immunoprecipitation assay demonstrated PPARγ binding in the genomic sequence of fibulin-5 in fibroblasts. Our results underscore the importance of PPARγ in AAA development though orchestrating proper elastogenesis and preserving elastic fiber integrity.


Subject(s)
Aorta, Abdominal/drug effects , Aortic Aneurysm, Abdominal/genetics , Gene Expression Regulation , Matrix Metalloproteinases/metabolism , PPAR gamma/genetics , Analysis of Variance , Angiotensin II/pharmacology , Animals , Aorta, Abdominal/metabolism , Aorta, Abdominal/pathology , Cells, Cultured , Disease Models, Animal , Elastin/metabolism , Humans , Inflammation/metabolism , Male , Mice , Mice, Knockout , Middle Aged , Muscle, Smooth, Vascular/metabolism
20.
Biochim Biophys Acta ; 1860(1 Pt A): 36-45, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26344063

ABSTRACT

BACKGROUND: S-nitrosylation of mitochondrial enzymes involved in energy transfer under nitrosative stress may result in ATP deficiency. We investigated whether α-lipoic acid, a powerful antioxidant, could alleviate nitrosative stress by regulating S-nitrosylation, which could result in retaining the mitochondrial enzyme activity. METHODS: In this study, we have identified the S-nitrosylated forms of subunit 1 of dihydrolipoyllysine succinyltransferase (complex III), and subunit 2 of the α-ketoglutarate dehydrogenase complex by implementing a fluorescence-based differential quantitative proteomics method. RESULTS: We found that the activities of these two mitochondrial enzymes were partially but reversibly inhibited by S-nitrosylation in cultured endothelial cells, and that their activities were partially restored by supplementation of α-lipoic acid. We show that protein S-nitrosylation affects the activity of mitochondrial enzymes that are central to energy supply, and that α-lipoic acid protects mitochondrial enzymes by altering S-nitrosylation levels. CONCLUSIONS: Inhibiting protein S-nitrosylation with α-lipoic acid seems to be a protective mechanism against nitrosative stress. GENERAL SIGNIFICANCE: Identification and characterization of these new protein targets should contribute to expanding the therapeutic power of α-lipoic acid and to a better understanding of the underlying antioxidant mechanisms.


Subject(s)
Electron Transport Complex III/metabolism , Ketoglutarate Dehydrogenase Complex/metabolism , Nitric Oxide/metabolism , Oxidative Stress , Thioctic Acid/pharmacology , Adenosine Triphosphate/biosynthesis , Animals , Cells, Cultured , Mice , Mice, Inbred C57BL , Mitochondria/drug effects , Mitochondria/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...