Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 29
1.
J Am Heart Assoc ; 12(16): e8711, 2023 08 15.
Article En | MEDLINE | ID: mdl-37581385

Background The association of circulating trimethylamine-N-oxide (TMAO) with stroke has received limited attention. To address this gap, we examined the associations of serial measures of plasma TMAO with incident ischemic stroke. Methods and Results We used a prospective cohort design with data pooled from 2 cohorts. The settings were the CHS (Cardiovascular Health Study), a cohort of older adults, and the MESA (Multi-Ethnic Study of Atherosclerosis), both in the United States. We measured plasma concentrations of TMAO at baseline and again during the follow-up using high-performance liquid chromatography and mass spectrometry. We assessed the association of plasma TMAO with incident ischemic stroke using proportional hazards regression adjusted for risk factors. The combined cohorts included 11 785 participants without a history of stroke, on average 73 (CHS) and 62 (MESA) years old at baseline, including 60% (CHS) and 53% (MESA) women. We identified 1031 total incident ischemic strokes during a median 15-year follow-up in the combined cohorts. In multivariable analyses, TMAO was significantly associated with incident ischemic stroke risk (hazard ratios comparing a doubling of TMAO: 1.11 [1.03-1.18], P=0.004). The association was linear over the range of TMAO concentrations and appeared restricted to those without diagnosed coronary heart disease. An association with hemorrhagic stroke was not found. Conclusions Plasma TMAO levels are associated with incident ischemic stroke in a diverse population. Registration URL: https://www.clinicaltrials.gov. Unique identifier: NCT00005133.


Atherosclerosis , Ischemic Stroke , Stroke , Aged , Female , Humans , Atherosclerosis/diagnosis , Atherosclerosis/epidemiology , Atherosclerosis/complications , Ischemic Stroke/diagnosis , Ischemic Stroke/epidemiology , Ischemic Stroke/complications , Methylamines , Oxides , Prospective Studies , Risk Factors , Stroke/diagnosis , Stroke/epidemiology , United States/epidemiology
2.
Int J Mol Sci ; 24(9)2023 Apr 30.
Article En | MEDLINE | ID: mdl-37175797

Stroke is the second most common cause of cognitive impairment and dementia. Vascular dementia (VaD), a cognitive impairment following a stroke, is common and significantly impacts the quality of life. We recently demonstrated via gut microbe transplant studies that the gut microbe-dependent trimethylamine-N-oxide (TMAO) pathway impacts stroke severity, both infarct size and long-term cognitive outcomes. However, the molecular mechanisms that underly the role of the microbiome in VaD have not been explored in depth. To address this issue, we performed a comprehensive RNA-sequencing analysis to identify differentially expressed (DE) genes in the ischemic cerebral cortex of mouse brains at pre-stroke and post-stroke day 1 and day 3. A total of 4016, 3752 and 7861 DE genes were identified at pre-stroke and post-stroke day 1 and day 3, respectively. The Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analysis indicated pathways of neurodegeneration in multiple diseases, chemokine signaling, calcium signaling, and IL-17 signaling as the key enriched pathways. Inflammatory response genes interleukin-1 beta (Il-1ß), chemokines (C-X-C motif chemokine ligand 10 (Cxcl10), chemokine ligand 2 (Ccl2)), and immune system genes (S100 calcium binding protein 8 (S100a8), lipocalin-2 (Lcn2)) were among the most significantly upregulated genes. Hypocretin neuropeptide precursor (Hcrt), a neuropeptide, and transcription factors such as neuronal PAS domain protein 4 (Npas4), GATA binding protein 3 (Gata3), and paired box 7 (Pax7) were among the most significantly downregulated genes. In conclusion, our results indicate that higher plasma TMAO levels induce differential mRNA expression profiles in the ischemic brain tissue in our pre-clinical stroke model, and the predicted pathways provide the molecular basis for regulating the TMAO-enhanced neuroinflammatory response in the brain.


Dementia, Vascular , Gastrointestinal Microbiome , Stroke , Animals , Mice , Gastrointestinal Microbiome/physiology , Dementia, Vascular/genetics , Transcriptome , Ligands , Quality of Life , Stroke/genetics , Methylamines/metabolism , Basic Helix-Loop-Helix Transcription Factors/metabolism
3.
Nat Microbiol ; 7(1): 73-86, 2022 01.
Article En | MEDLINE | ID: mdl-34949826

The heightened cardiovascular disease (CVD) risk observed among omnivores is thought to be linked, in part, to gut microbiota-dependent generation of trimethylamine-N-oxide (TMAO) from L-carnitine, a nutrient abundant in red meat. Gut microbial transformation of L-carnitine into trimethylamine (TMA), the precursor of TMAO, occurs via the intermediate γ-butyrobetaine (γBB). However, the interrelationship of γBB, red meat ingestion and CVD risks, as well as the gut microbial genes responsible for the transformation of γBB to TMA, are unclear. In the present study, we show that plasma γBB levels in individuals from a clinical cohort (n = 2,918) are strongly associated with incident CVD event risks. Culture of human faecal samples and microbial transplantation studies in gnotobiotic mice with defined synthetic communities showed that the introduction of Emergencia timonensis, a human gut microbe that can metabolize γBB into TMA, is sufficient to complete the carnitine → γBB → TMA transformation, elevate TMAO levels and enhance thrombosis potential in recipients after arterial injury. RNA-sequencing analyses of E. timonensis identified a six-gene cluster, herein named the γBB utilization (gbu) gene cluster, which is upregulated in response to γBB. Combinatorial cloning and functional studies identified four genes (gbuA, gbuB, gbuC and gbuE) that are necessary and sufficient to recapitulate the conversion of γBB to TMA when coexpressed in Escherichia coli. Finally, reanalysis of samples (n = 113) from a clinical, randomized diet, intervention study showed that the abundance of faecal gbuA correlates with plasma TMAO and a red meat-rich diet. Our findings reveal a microbial gene cluster that is critical to dietary carnitine → γBB → TMA → TMAO transformation in hosts and contributes to CVD risk.


Cardiovascular Diseases/genetics , Carnitine/blood , Carnitine/metabolism , Gastrointestinal Microbiome/physiology , Genes, Bacterial/genetics , Multigene Family , Red Meat , Animals , Cardiovascular Diseases/blood , Clostridiales/genetics , Clostridiales/metabolism , Feces/microbiology , Female , Germ-Free Life , Humans , Methylamines/metabolism , Mice , Mice, Inbred C57BL , Observational Studies as Topic
4.
Cell Host Microbe ; 29(7): 1199-1208.e5, 2021 07 14.
Article En | MEDLINE | ID: mdl-34139173

Clinical studies have demonstrated associations between circulating levels of the gut-microbiota-derived metabolite trimethylamine-N-oxide (TMAO) and stroke incident risk. However, a causal role of gut microbes in stroke has not yet been demonstrated. Herein we show that gut microbes, through dietary choline and TMAO generation, directly impact cerebral infarct size and adverse outcomes following stroke. Fecal microbial transplantation from low- versus high-TMAO-producing human subjects into germ-free mice shows that both TMAO generation and stroke severity are transmissible traits. Furthermore, employing multiple murine stroke models and transplantation of defined microbial communities with genetically engineered human commensals into germ-free mice, we demonstrate that the microbial cutC gene (an enzymatic source of choline-to-TMA transformation) is sufficient to transmit TMA/TMAO production, heighten cerebral infarct size, and lead to functional impairment. We thus reveal that gut microbiota in general, specifically the metaorganismal TMAO pathway, directly contributes to stroke severity.


Bacteria/metabolism , Gastrointestinal Microbiome , Methylamines/metabolism , Oxides/metabolism , Stroke/microbiology , Animals , Bacteria/classification , Bacteria/genetics , Bacteria/isolation & purification , Choline/metabolism , Female , Humans , Male , Mice , Mice, Inbred C57BL , Severity of Illness Index , Stroke/metabolism , Stroke/pathology
5.
Cell ; 180(5): 862-877.e22, 2020 03 05.
Article En | MEDLINE | ID: mdl-32142679

Using untargeted metabolomics (n = 1,162 subjects), the plasma metabolite (m/z = 265.1188) phenylacetylglutamine (PAGln) was discovered and then shown in an independent cohort (n = 4,000 subjects) to be associated with cardiovascular disease (CVD) and incident major adverse cardiovascular events (myocardial infarction, stroke, or death). A gut microbiota-derived metabolite, PAGln, was shown to enhance platelet activation-related phenotypes and thrombosis potential in whole blood, isolated platelets, and animal models of arterial injury. Functional and genetic engineering studies with human commensals, coupled with microbial colonization of germ-free mice, showed the microbial porA gene facilitates dietary phenylalanine conversion into phenylacetic acid, with subsequent host generation of PAGln and phenylacetylglycine (PAGly) fostering platelet responsiveness and thrombosis potential. Both gain- and loss-of-function studies employing genetic and pharmacological tools reveal PAGln mediates cellular events through G-protein coupled receptors, including α2A, α2B, and ß2-adrenergic receptors. PAGln thus represents a new CVD-promoting gut microbiota-dependent metabolite that signals via adrenergic receptors.


Cardiovascular Diseases/blood , Gastrointestinal Microbiome/genetics , Glutamine/analogs & derivatives , Thrombosis/metabolism , Animals , Arteries/injuries , Arteries/metabolism , Arteries/microbiology , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Blood Platelets/metabolism , Blood Platelets/microbiology , Cardiovascular Diseases/genetics , Cardiovascular Diseases/microbiology , Cardiovascular Diseases/pathology , Death, Sudden, Cardiac/pathology , Glutamine/blood , Glutamine/genetics , Humans , Male , Metabolome/genetics , Metabolomics/methods , Mice , Myocardial Infarction/blood , Myocardial Infarction/microbiology , Platelet Activation/genetics , Receptors, Adrenergic, alpha/blood , Receptors, Adrenergic, alpha/genetics , Receptors, Adrenergic, beta/blood , Receptors, Adrenergic, beta/genetics , Risk Factors , Stroke/blood , Stroke/microbiology , Stroke/pathology , Thrombosis/genetics , Thrombosis/microbiology , Thrombosis/pathology
6.
Arterioscler Thromb Vasc Biol ; 39(6): 1045-1054, 2019 06.
Article En | MEDLINE | ID: mdl-31070450

Objective- FMO (flavin-containing monooxygenase) 3 converts bacterial-derived trimethylamine to trimethylamine N-oxide (TMAO), an independent risk factor for cardiovascular disease. We generated FMO3 knockout (FMO3KO) mouse to study its effects on plasma TMAO, lipids, glucose/insulin metabolism, thrombosis, and atherosclerosis. Approach and Results- Previous studies with an antisense oligonucleotide (ASO) knockdown strategy targeting FMO3 in LDLRKO (low-density lipoprotein receptor knockout) mice resulted in major reductions in TMAO levels and atherosclerosis, but also showed effects on plasma lipids, insulin, and glucose. Although FMO3KO mice generated via CRISPR/Cas9 technology bred onto the LDLRKO background did exhibit similar effects on TMAO levels, the effects on lipid metabolism were not as pronounced as with the ASO knockdown model. These differences could result from either off-target effects of the ASO or from a developmental adaptation to the FMO3 deficiency. To distinguish these possibilities, we treated wild-type and FMO3KO mice with control or FMO3 ASOs. FMO3-ASO treatment led to the same extent of lipid-lowering effects in the FMO3KO mice as the wild-type mice, indicating off-target effects. The levels of TMAO in LDLRKO mice fed an atherogenic diet are very low in both wild-type and FMO3KO mice, and no significant effect was observed on atherosclerosis. When FMO3KO and wild-type mice were maintained on a 0.5% choline diet, FMO3KO showed a marked reduction in both TMAO and in vivo thrombosis potential. Conclusions- FMO3KO markedly reduces systemic TMAO levels and thrombosis potential. However, the previously observed large effects of an FMO3 ASO on plasma lipid levels appear to be due partly to off-target effects.


Atherosclerosis/metabolism , Choline/metabolism , Methylamines/metabolism , Oxygenases/genetics , Thrombosis/metabolism , Animals , Atherosclerosis/genetics , Choline/pharmacology , Disease Models, Animal , Lipid Metabolism/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Oxygenases/metabolism , Polymerase Chain Reaction/methods , Random Allocation , Reference Values , Thrombosis/physiopathology
7.
Circ Res ; 123(10): 1164-1176, 2018 10 26.
Article En | MEDLINE | ID: mdl-30359185

RATIONALE: Gut microbes influence cardiovascular disease and thrombosis risks through the production of trimethylamine N-oxide (TMAO). Microbiota-dependent generation of trimethylamine (TMA)-the precursor to TMAO-is rate limiting in the metaorganismal TMAO pathway in most humans and is catalyzed by several distinct microbial choline TMA-lyases, including the proteins encoded by the cutC/D (choline utilization C/D) genes in multiple human commensals. OBJECTIVE: Direct demonstration that the gut microbial cutC gene is sufficient to transmit enhanced platelet reactivity and thrombosis potential in a host via TMA/TMAO generation has not yet been reported. METHODS AND RESULTS: Herein, we use gnotobiotic mice and a series of microbial colonization studies to show that microbial cutC-dependent TMA/TMAO production is sufficient to transmit heightened platelet reactivity and thrombosis potential in a host. Specifically, we examine in vivo thrombosis potential employing germ-free mice colonized with either high TMA-producing stable human fecal polymcrobial communities or a defined CutC-deficient background microbial community coupled with a CutC-expressing human commensal±genetic disruption of its cutC gene (ie, Clostridium sporogenes Δ cutC). CONCLUSIONS: Collectively, these studies point to the microbial choline TMA-lyase pathway as a rational molecular target for the treatment of atherothrombotic heart disease.


Bacterial Proteins/metabolism , Fecal Microbiota Transplantation , Lyases/metabolism , Platelet Activation , Thrombosis/microbiology , Adult , Animals , Bacterial Proteins/genetics , Choline/metabolism , Clostridium/enzymology , Clostridium/genetics , Female , Gastrointestinal Microbiome , Humans , Lyases/genetics , Male , Methylamines/metabolism , Mice , Mice, Inbred C57BL , Middle Aged , Thrombosis/blood
8.
Nat Med ; 24(9): 1407-1417, 2018 09.
Article En | MEDLINE | ID: mdl-30082863

Trimethylamine N-oxide (TMAO) is a gut microbiota-derived metabolite that enhances both platelet responsiveness and in vivo thrombosis potential in animal models, and TMAO plasma levels predict incident atherothrombotic event risks in human clinical studies. TMAO is formed by gut microbe-dependent metabolism of trimethylamine (TMA) moiety-containing nutrients, which are abundant in a Western diet. Here, using a mechanism-based inhibitor approach targeting a major microbial TMA-generating enzyme pair, CutC and CutD (CutC/D), we developed inhibitors that are potent, time-dependent, and irreversible and that do not affect commensal viability. In animal models, a single oral dose of a CutC/D inhibitor significantly reduced plasma TMAO levels for up to 3 d and rescued diet-induced enhanced platelet responsiveness and thrombus formation, without observable toxicity or increased bleeding risk. The inhibitor selectively accumulated within intestinal microbes to millimolar levels, a concentration over 1-million-fold higher than needed for a therapeutic effect. These studies reveal that mechanism-based inhibition of gut microbial TMA and TMAO production reduces thrombosis potential, a critical adverse complication in heart disease. They also offer a generalizable approach for the selective nonlethal targeting of gut microbial enzymes linked to host disease limiting systemic exposure of the inhibitor in the host.


Gastrointestinal Microbiome , Thrombosis/microbiology , Animals , Bacteria/drug effects , Bacteria/metabolism , Choline/pharmacology , Diet , Gastrointestinal Microbiome/drug effects , Hexanols/pharmacology , Mice, Inbred C57BL , Oxidoreductases, N-Demethylating/antagonists & inhibitors , Oxidoreductases, N-Demethylating/metabolism , Platelet Aggregation/drug effects
9.
Arterioscler Thromb Vasc Biol ; 38(9): 2225-2235, 2018 09.
Article En | MEDLINE | ID: mdl-29976769

Objective- Gut microbiota-dependent metabolites, in particular trimethylamine N-oxide (TMAO), have recently been reported to promote atherosclerosis and thrombosis. Here, we examined for the first time the relation of TMAO and the risk of incident cardiovascular events in patients with recent first-ever ischemic stroke in 2 independent prospective cohorts. Moreover, the link between TMAO and proinflammatory monocytes as a potential contributing factor for cardiovascular risk in stroke patients was studied. Approach and Results- In a first study (n=78), higher TMAO plasma levels were linked with an increased risk of incident cardiovascular events including myocardial infarction, recurrent stroke, and cardiovascular death (fourth quartile versus first quartile; hazard ratio, 2.31; 95% CI, 1.25-4.23; P<0.01). In the second independent validation cohort (n=593), high TMAO levels again heralded marked increased risk of adverse cardiovascular events (fourth quartile versus first quartile; hazard ratio, 5.0; 95% CI, 1.7-14.8; P<0.01), and also after adjustments for cardiovascular risk factors including hypertension, diabetes mellitus, LDL (low-density lipoprotein) cholesterol, and estimated glomerular filtration rate (hazard ratio, 3.3; 95% CI, 1.2-10.9; P=0.04). A significant correlation was also found between TMAO levels and percentage of proinflammatory intermediate CD14++CD16+ monocytes ( r=0.70; P<0.01). Moreover, in mice fed a diet enriched with choline to increase TMAO synthesis, levels of proinflammatory murine Ly6Chigh monocytes were higher than in the chow-fed control group (choline: 9.2±0.5×103 per mL versus control: 6.5±0.5×103 per mL; P<0.01). This increase was abolished in mice with depleted gut microbiota (choline+antibiotics: 5.4±0.7×103 per mL; P<0.001 versus choline). Conclusions- The present study demonstrates for the first time a graded relation between TMAO levels and the risk of subsequent cardiovascular events in patients with recent prior ischemic stroke. Our data support the notion that TMAO-related increase of proinflammatory monocytes may add to elevated cardiovascular risk of patients with increased TMAO levels.


Brain Ischemia/complications , Brain Ischemia/metabolism , Cardiovascular Diseases/etiology , Gastrointestinal Microbiome/physiology , Methylamines/blood , Monocytes/metabolism , Stroke/complications , Stroke/metabolism , Animals , Antigens, CD , Antigens, Differentiation, T-Lymphocyte , CD4 Antigens , Death, Sudden, Cardiac/etiology , Female , Humans , Inflammation , Male , Mice, Inbred C57BL , Monocytes/immunology , Prospective Studies , Recurrence , Risk Factors
11.
Cell ; 165(1): 111-124, 2016 Mar 24.
Article En | MEDLINE | ID: mdl-26972052

Normal platelet function is critical to blood hemostasis and maintenance of a closed circulatory system. Heightened platelet reactivity, however, is associated with cardiometabolic diseases and enhanced potential for thrombotic events. We now show gut microbes, through generation of trimethylamine N-oxide (TMAO), directly contribute to platelet hyperreactivity and enhanced thrombosis potential. Plasma TMAO levels in subjects (n > 4,000) independently predicted incident (3 years) thrombosis (heart attack, stroke) risk. Direct exposure of platelets to TMAO enhanced sub-maximal stimulus-dependent platelet activation from multiple agonists through augmented Ca(2+) release from intracellular stores. Animal model studies employing dietary choline or TMAO, germ-free mice, and microbial transplantation collectively confirm a role for gut microbiota and TMAO in modulating platelet hyperresponsiveness and thrombosis potential and identify microbial taxa associated with plasma TMAO and thrombosis potential. Collectively, the present results reveal a previously unrecognized mechanistic link between specific dietary nutrients, gut microbes, platelet function, and thrombosis risk.


Blood Platelets/metabolism , Gastrointestinal Microbiome , Methylamines/metabolism , Thrombosis/metabolism , Animals , Calcium/metabolism , Carotid Artery Injuries/pathology , Cecum/microbiology , Chlorides , Choline/metabolism , Diet , Female , Ferric Compounds , Germ-Free Life , Humans , Methylamines/blood , Mice , Mice, Inbred C57BL , Thrombosis/pathology
12.
J Am Heart Assoc ; 5(2)2016 Feb 22.
Article En | MEDLINE | ID: mdl-26903003

BACKGROUND: The choline-derived metabolite trimethylamine N-oxide (TMAO) has been demonstrated to contribute to atherosclerosis and is associated with coronary artery disease risk. METHODS AND RESULTS: We explored the impact of TMAO on endothelial and smooth muscle cell function in vivo, focusing on disease-relevant outcomes for atherogenesis. Initially, we observed that aortas of LDLR(-/-) mice fed a choline diet showed elevated inflammatory gene expression compared with controls. Acute TMAO injection at physiological levels was sufficient to induce the same inflammatory markers and activate the well-known mitogen-activated protein kinase, extracellular signal-related kinase, and nuclear factor-κB signaling cascade. These observations were recapitulated in primary human aortic endothelial cells and vascular smooth muscle cells. We also found that TMAO promotes recruitment of activated leukocytes to endothelial cells. Through pharmacological inhibition, we further showed that activation of nuclear factor-κB signaling was necessary for TMAO to induce inflammatory gene expression in both of these relevant cell types as well as endothelial cell adhesion of leukocytes. CONCLUSIONS: Our results suggest a likely contributory mechanism for TMAO-dependent enhancement in atherosclerosis and cardiovascular risks.


Aortitis/chemically induced , Atherosclerosis/chemically induced , Endothelial Cells/drug effects , Methylamines/toxicity , Mitogen-Activated Protein Kinases/metabolism , Muscle, Smooth, Vascular/drug effects , Myocytes, Smooth Muscle/drug effects , NF-kappa B/metabolism , Animals , Aorta/drug effects , Aorta/enzymology , Aorta/pathology , Aortitis/enzymology , Aortitis/genetics , Aortitis/pathology , Atherosclerosis/enzymology , Atherosclerosis/genetics , Atherosclerosis/pathology , Cell Adhesion/drug effects , Cells, Cultured , Choline , Coculture Techniques , Disease Models, Animal , Endothelial Cells/enzymology , Endothelial Cells/pathology , Enzyme Activation , Female , Gene Expression Regulation , Genetic Predisposition to Disease , Humans , Leukocytes/drug effects , Leukocytes/enzymology , Mice, Inbred C57BL , Mice, Knockout , Muscle, Smooth, Vascular/enzymology , Muscle, Smooth, Vascular/pathology , Myocytes, Smooth Muscle/enzymology , Myocytes, Smooth Muscle/pathology , Phenotype , Receptors, LDL/deficiency , Receptors, LDL/genetics , Signal Transduction/drug effects
13.
Cell ; 163(7): 1585-95, 2015 Dec 17.
Article En | MEDLINE | ID: mdl-26687352

Trimethylamine (TMA) N-oxide (TMAO), a gut-microbiota-dependent metabolite, both enhances atherosclerosis in animal models and is associated with cardiovascular risks in clinical studies. Here, we investigate the impact of targeted inhibition of the first step in TMAO generation, commensal microbial TMA production, on diet-induced atherosclerosis. A structural analog of choline, 3,3-dimethyl-1-butanol (DMB), is shown to non-lethally inhibit TMA formation from cultured microbes, to inhibit distinct microbial TMA lyases, and to both inhibit TMA production from physiologic polymicrobial cultures (e.g., intestinal contents, human feces) and reduce TMAO levels in mice fed a high-choline or L-carnitine diet. DMB inhibited choline diet-enhanced endogenous macrophage foam cell formation and atherosclerotic lesion development in apolipoprotein e(-/-) mice without alterations in circulating cholesterol levels. The present studies suggest that targeting gut microbial production of TMA specifically and non-lethal microbial inhibitors in general may serve as a potential therapeutic approach for the treatment of cardiometabolic diseases.


Atherosclerosis/drug therapy , Choline/analogs & derivatives , Gastrointestinal Tract/microbiology , Hexanols/administration & dosage , Lyases/antagonists & inhibitors , Methylamines/metabolism , Animals , Apolipoproteins E/genetics , Atherosclerosis/metabolism , Cholesterol/metabolism , Choline/metabolism , Diet , Feces/chemistry , Foam Cells/metabolism , Humans , Lyases/metabolism , Mice , Mice, Inbred C57BL , Microbiota
14.
PLoS One ; 10(4): e0124494, 2015.
Article En | MEDLINE | ID: mdl-25886360

Scavenger receptor class B, type I (SR-BI) and its adaptor protein PDZK1 mediate responses to HDL cholesterol in endothelium. Whether the receptor-adaptor protein tandem serves functions in other vascular cell types is unknown. The current work determined the roles of SR-BI and PDZK1 in vascular smooth muscle (VSM). To evaluate possible VSM functions of SR-BI and PDZK1 in vivo, neointima formation was assessed 21 days post-ligation in the carotid arteries of wild-type, SR-BI-/- or PDZK1-/- mice. Whereas neointima development was negligible in wild-type and SR-BI-/-, there was marked neointima formation in PDZK1-/- mice. PDZK1 expression was demonstrated in primary mouse VSM cells, and compared to wild-type cells, PDZK1-/- VSM displayed exaggerated proliferation and migration in response to platelet derived growth factor (PDGF). Tandem affinity purification-mass spectrometry revealed that PDZK1 interacts with breakpoint cluster region kinase (Bcr), which contains a C-terminal PDZ binding sequence and is known to enhance responses to PDGF in VSM. PDZK1 interaction with Bcr in VSM was demonstrated by pull-down and by coimmunoprecipitation, and the augmented proliferative response to PDGF in PDZK1-/- VSM was abrogated by Bcr depletion. Furthermore, compared with wild-type Bcr overexpression, the introduction of a Bcr mutant incapable of PDZK1 binding into VSM cells yielded an exaggerated proliferative response to PDGF. Thus, PDZK1 has novel SR-BI-independent function in VSM that affords protection from neointima formation, and this involves PDZK1 suppression of VSM cell proliferation via an inhibitory interaction with Bcr.


Intracellular Signaling Peptides and Proteins/physiology , Muscle, Smooth, Vascular/enzymology , Proto-Oncogene Proteins c-bcr/antagonists & inhibitors , Tunica Intima/growth & development , Animals , Cell Movement , Cell Proliferation , Intracellular Signaling Peptides and Proteins/genetics , Membrane Proteins , Mice , Mice, Inbred C57BL , Mice, Knockout , Muscle, Smooth, Vascular/cytology , Proto-Oncogene Proteins c-bcr/metabolism
15.
J Biol Chem ; 290(9): 5647-60, 2015 Feb 27.
Article En | MEDLINE | ID: mdl-25550161

Recent studies indicate both clinical and mechanistic links between atherosclerotic heart disease and intestinal microbial metabolism of certain dietary nutrients producing trimethylamine N-oxide (TMAO). Here we test the hypothesis that gut microbial transplantation can transmit choline diet-induced TMAO production and atherosclerosis susceptibility. First, a strong association was noted between atherosclerotic plaque and plasma TMAO levels in a mouse diversity panel (n = 22 strains, r = 0.38; p = 0.0001). An atherosclerosis-prone and high TMAO-producing strain, C57BL/6J, and an atherosclerosis-resistant and low TMAO-producing strain, NZW/LacJ, were selected as donors for cecal microbial transplantation into apolipoprotein e null mice in which resident intestinal microbes were first suppressed with antibiotics. Trimethylamine (TMA) and TMAO levels were initially higher in recipients on choline diet that received cecal microbes from C57BL/6J inbred mice; however, durability of choline diet-dependent differences in TMA/TMAO levels was not maintained to the end of the study. Mice receiving C57BL/6J cecal microbes demonstrated choline diet-dependent enhancement in atherosclerotic plaque burden as compared with recipients of NZW/LacJ microbes. Microbial DNA analyses in feces and cecum revealed transplantation of donor microbial community features into recipients with differences in taxa proportions between donor strains that were transmissible to recipients and that tended to show coincident proportions with TMAO levels. Proportions of specific taxa were also identified that correlated with plasma TMAO levels in donors and recipients and with atherosclerotic lesion area in recipients. Atherosclerosis susceptibility may be transmitted via transplantation of gut microbiota. Gut microbes may thus represent a novel therapeutic target for modulating atherosclerosis susceptibility.


Atherosclerosis/microbiology , Cecum/microbiology , Disease Susceptibility/microbiology , Gastrointestinal Tract/microbiology , Microbiota/physiology , Animals , Aorta/metabolism , Aorta/pathology , Atherosclerosis/blood , Atherosclerosis/etiology , Choline/administration & dosage , Diet/adverse effects , Disease Susceptibility/blood , Disease Susceptibility/complications , Female , Host-Pathogen Interactions , Humans , Male , Methylamines/blood , Methylamines/metabolism , Mice, Inbred AKR , Mice, Inbred BALB C , Mice, Inbred C3H , Mice, Inbred C57BL , Mice, Inbred CBA , Mice, Inbred DBA , Mice, Inbred Strains , Mice, Knockout , Mice, Transgenic , Species Specificity
16.
J Vasc Surg ; 58(2): 470-7, 2013 Aug.
Article En | MEDLINE | ID: mdl-23375603

OBJECTIVE: Endothelial dysfunction has been studied in animal models. However, direct evidence of endothelial function from human vessels is limited. Our objectives were to optimize methods in harvesting human arteries from amputation specimens, determine endothelial function, and measure responsiveness to l-arginine, a nitric oxide precursor. METHODS: Fresh amputation specimens were transferred expeditiously from the operating room to the bench laboratory for dissection and arterial harvest in an Investigational Review Board-approved protocol. Popliteal and tibial vessels were examined in pilot experiments leading to the use of the anterior tibial artery in consecutive experiments. Human lower extremity anterior tibial artery segments were harvested from 14 amputation specimens. Specimens were rapidly collected and divided for endothelial-dependent relaxation (EDR) studies in a tissue bath apparatus, immunohistochemistry, and intravascular ultrasound-derived virtual histology. A total of 47 ring segments were studied. The data were compared with two-way analysis of variance. RESULTS: Human lower extremity arteries exhibited low responsiveness to acetylcholine (EDR, 24.9%; acetylcholine, 10(-4)). L-arginine supplementation enhanced EDR by 38.5% (P < .0001). N-nitro-L-arginine methyl ester abrogated EDR (P < .0001) in vessels exposed to L-arginine. Arterial responsiveness was intact in all vessels (endothelial independent relaxation to sodium nitroprusside, 113.2% ± 28.1%). Histology and immunohistochemistry confirmed intact endothelium by morphometric analysis, cluster of differentiation 31, endothelial nitric oxide synthase, and arginase II staining. Intravascular ultrasound-derived virtual histology indicated atheroma burden was 11.9 ± 4.7 mm(3)/cm, and plaque stratification indicated fibrous morphology was predominant (59.9%; necrotic core, 16.9%; calcium, 11.2%). Variations in plaque morphology did not correlate with endothelial function or responsiveness to L-arginine. CONCLUSIONS: Human lower extremity arteries demonstrate low baseline endothelial function in patients requiring amputation. Endothelial dysfunction is improved by L-arginine supplementation in an ex vivo model. These results support strategies to increase local levels of nitric oxide in human vessels.


Endothelium, Vascular/surgery , Lower Extremity/blood supply , Peripheral Arterial Disease/surgery , Tibia/surgery , Tissue and Organ Harvesting/methods , Amputation, Surgical , Arginase/analysis , Biomarkers/analysis , Endothelium, Vascular/chemistry , Endothelium, Vascular/diagnostic imaging , Endothelium, Vascular/drug effects , Endothelium, Vascular/pathology , Endothelium, Vascular/physiopathology , Feasibility Studies , Fibrosis , Humans , Immunohistochemistry , Necrosis , Nitric Oxide Donors/pharmacology , Nitric Oxide Synthase Type III/analysis , Peripheral Arterial Disease/diagnosis , Peripheral Arterial Disease/metabolism , Peripheral Arterial Disease/physiopathology , Plaque, Atherosclerotic , Platelet Endothelial Cell Adhesion Molecule-1/analysis , Tibia/chemistry , Tibia/diagnostic imaging , Tibia/drug effects , Tibia/pathology , Tibia/physiopathology , Ultrasonography, Interventional , Vasodilation , Vasodilator Agents/pharmacology
17.
Blood ; 119(25): 6136-44, 2012 Jun 21.
Article En | MEDLINE | ID: mdl-22431576

Diabetes mellitus has been associated with platelet hyperreactivity, which plays a central role in the hyperglycemia-related prothrombotic phenotype. The mechanisms responsible for this phenomenon are not established. In the present study, we investigated the role of CD36, a class-B scavenger receptor, in this process. Using both in vitro and in vivo mouse models, we demonstrated direct and specific interactions of platelet CD36 with advanced glycation end products (AGEs) generated under hyperglycemic conditions. AGEs bound to platelet CD36 in a specific and dose-dependent manner, and binding was inhibited by the high-affinity CD36 ligand NO(2)LDL. Cd36-null platelets did not bind AGE. Using diet- and drug-induced mouse models of diabetes, we have shown that cd36-null mice had a delayed time to the formation of occlusive thrombi compared with wild-type (WT) in a FeCl(3)-induced carotid artery injury model. Cd36-null mice had a similar level of hyperglycemia and a similar level of plasma AGEs compared with WT mice under this condition, but WT mice had more AGEs incorporated into thrombi. Mechanistic studies revealed that CD36-dependent JNK2 activation is involved in this prothrombotic pathway. Therefore, the results of the present study couple vascular complications in diabetes mellitus with AGE-CD36-mediated platelet signaling and hyperreactivity.


Asymptomatic Diseases , Blood Platelets/metabolism , CD36 Antigens/metabolism , Glycation End Products, Advanced/metabolism , Glycation End Products, Advanced/physiology , Thrombosis/etiology , Animals , Blood Platelets/drug effects , CD36 Antigens/genetics , CD36 Antigens/physiology , Diabetes Mellitus, Experimental/blood , Diabetes Mellitus, Experimental/complications , Diabetes Mellitus, Experimental/genetics , Diabetes Mellitus, Experimental/metabolism , Diabetic Angiopathies/blood , Diabetic Angiopathies/genetics , Diabetic Angiopathies/metabolism , Diabetic Angiopathies/pathology , Diet, Atherogenic , Glycation End Products, Advanced/pharmacology , Hyperglycemia/blood , Hyperglycemia/etiology , Hyperglycemia/genetics , Hyperglycemia/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Phenotype , Platelet Aggregation/genetics , Platelet Aggregation/physiology , Protein Binding , Streptozocin , Thrombosis/metabolism , Thrombosis/pathology
18.
Circ Res ; 109(10): 1132-40, 2011 Oct 28.
Article En | MEDLINE | ID: mdl-21940940

RATIONALE: Elevations in C-reactive protein (CRP) are associated with increased cardiovascular disease risk and endothelial dysfunction. CRP antagonizes endothelial nitric oxide synthase (eNOS) through processes mediated by the IgG receptor Fcγ receptor IIB (FcγRIIB), its immunoreceptor tyrosine-based inhibitory motif, and SH2 domain-containing inositol 5'-phosphatase 1. In mice, CRP actions on eNOS blunt carotid artery re-endothelialization. OBJECTIVE: How CRP activates FcγRIIB in endothelium is not known. We determined the role of Fcγ receptor I (FcγRI) and the basis for coupling of FcγRI to FcγRIIB in endothelium. METHODS AND RESULTS: In cultured endothelial cells, FcγRI-blocking antibodies prevented CRP antagonism of eNOS, and CRP activated Src via FcγRI. CRP-induced increases in FcγRIIB immunoreceptor tyrosine-based inhibitory motif phosphorylation and SH2 domain-containing inositol 5'-phosphatase 1 activation were Src-dependent, and Src inhibition prevented eNOS antagonism by CRP. Similar processes mediated eNOS antagonism by aggregated IgG used to mimic immune complex. Carotid artery re-endothelialization was evaluated in offspring from crosses of CRP transgenic mice (TG-CRP) with either mice lacking the γ subunit of FcγRI (FcRγ(-/-)) or FcγRIIB(-/-) mice. Whereas re-endothelialization was impaired in TG-CRP vs wild-type, it was normal in both FcRγ(-/-); TG-CRP and FcγRIIB(-/-); TG-CRP mice. CONCLUSIONS: CRP antagonism of eNOS is mediated by the coupling of FcγRI to FcγRIIB by Src kinase and resulting activation of SH2 domain-containing inositol 5'-phosphatase 1, and consistent with this mechanism, both FcγRI and FcγRIIB are required for CRP to blunt endothelial repair in vivo. Similar mechanisms underlie eNOS antagonism by immune complex. FcγRI and FcγRIIB may be novel therapeutic targets for preventing endothelial dysfunction in inflammatory or immune complex-mediated conditions.


C-Reactive Protein/metabolism , Carotid Artery Injuries/enzymology , Carotid Artery Injuries/immunology , Endothelial Cells/enzymology , Endothelial Cells/immunology , Receptors, IgG/metabolism , src-Family Kinases/metabolism , Animals , Antigen-Antibody Complex/metabolism , C-Reactive Protein/genetics , Carotid Artery Injuries/genetics , Carotid Artery Injuries/pathology , Cattle , Cell Proliferation , Cells, Cultured , Disease Models, Animal , Endothelial Cells/pathology , Enzyme Activation , Humans , Inositol Polyphosphate 5-Phosphatases , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Nitric Oxide Synthase Type III/metabolism , Phosphoric Monoester Hydrolases/metabolism , Phosphorylation , Rabbits , Receptors, IgG/deficiency , Receptors, IgG/genetics , Signal Transduction , Time Factors , Transfection
19.
Am J Physiol Cell Physiol ; 298(4): C952-60, 2010 Apr.
Article En | MEDLINE | ID: mdl-20032511

Arterial thrombosis is a common disease leading to severe ischemia beyond the obstructing thrombus. Additionally, endothelial dysfunction at the site of thrombosis can be rescued by l-arginine supplementation or arginase blockade in several animal models. Exposure of rat aortic endothelial cells (RAECs) to thrombin upregulates arginase I mRNA and protein levels. In this study, we further investigated the molecular mechanism of thrombin-induced arginase changes in endothelial cells. Thrombin strikingly increased arginase I promoter and enzyme activity in primary cultured RAECs. Using different deletion and point mutations of the promoter, we demonstrated that the activating protein-1 (AP-1) consensus site located at -3,157 bp in the arginase I promoter was a thrombin-responsive element. Electrophoretic mobility shift assay and chromatin immunoprecipitation assay further confirmed that upon thrombin stimulation, c-Jun and activating transcription factor-2 (ATF-2) bound to the AP-1 site, which initiated the transactivation. Moreover, loss-of-function studies using small interfering RNA confirmed that recruitment of these two transcription factors to the AP-1 site was required for thrombin-induced arginase upregulation. In the course of defining the signaling pathway leading to the activation of AP-1 by thrombin, we found thrombin-induced phosphorylation of stress-activated protein kinase/c-Jun-NH(2)-terminal kinase (SAPK/JNK or JNK1/2/3) and p38 mitogen-activated protein kinase, which were followed by the phosphorylation of both c-Jun and ATF-2. These findings reveal the basis for thrombin induction of endothelial arginase I and indicate that arginase inhibition may be an attractive therapeutic alternative in the setting of arterial thrombosis and its associated endothelial dysfunction.


Arginase/metabolism , Endothelial Cells/enzymology , Transcription Factor AP-1/metabolism , Activating Transcription Factor 2/genetics , Activating Transcription Factor 2/metabolism , Animals , Aorta/cytology , Arginase/genetics , Cells, Cultured , Endothelial Cells/cytology , Mitogen-Activated Protein Kinases/metabolism , Proto-Oncogene Proteins c-jun/genetics , Proto-Oncogene Proteins c-jun/metabolism , RNA Interference , Rats , Signal Transduction/physiology , Thrombin/genetics , Thrombin/metabolism , Transcription Factor AP-1/genetics
20.
J Vasc Surg ; 48(2): 441-6, 2008 Aug.
Article En | MEDLINE | ID: mdl-18486420

INTRODUCTION: Acute arterial thrombosis causes endothelial dysfunction due to decreased nitric oxide bioactivity. Increased arginase activity may modulate intracellular L-arginine levels, the substrate for nitric oxide. The purpose of this study was to identify the role of arginase in endothelial dysfunction in cell culture and in the vasomotor response of arteries exposed to thrombus. METHODS: Rat aortic endothelial cells were exposed to thrombin at different time points. The cell extract was analyzed by immunoblotting and real-time polymerase chain reaction. Adult male rats underwent infrarenal aortic thrombosis by clip ligature for 1 hour. Infrarenal aortic ring segments were harvested and placed in physiologic buffer baths, and a force transducer was used to measure endothelial-dependent relaxation (EDR) and endothelial-independent relaxation (EIR). Arginase blockade was performed by incubating infrarenal aortic ring segments with arginase inhibitors for 1 hour before measuring EDR. Whole tissue extracts also underwent immunoblot analysis. The EDR and EIR curves were compared with analyses of variance. RESULTS: A 6.76 +/- 1.4-fold induction in arginase I message levels (P = .001) was found in rat aortic endothelial cells exposed to thrombin (30 U/mL), and arginase I protein levels increased 2.1 times. The eight infrarenal aortic ring segments exposed to thrombosis for 1 hour had diminished EDR curves compared with 14 nonthrombosed normal segments (controls). The maximum (+/- SEM) EDR (acetylcholine 10(-5)M dose) in control infrarenal aortic ring segments was 108% +/- 4.3% compared with 63% +/- 6.2% for thrombosed infrarenal aortic ring segments (P < .001). Exposure to arterial thrombosis resulted in a 3.8-times increase in arginase I protein levels in infrarenal aortic ring segments. Preincubation of nine infrarenal aortic ring segments with the nonspecific (difluoromethylornithine) and six with specific ([S]-[2-boronoethyl]-L-Cysteine-HCl [BEC]) arginase inhibitor for 1 hour significantly increased the maximum EDR compared with untreated thrombosed segments (104 +/- 5.2, 108 +/- 7.6 vs 63% +/- 6.2, P < .001). EDR curves for difluoromethylornithine- and BEC-treated infrarenal aortic ring segments were superimposed on control EDR curves. The EIR and the vasoconstriction with norepinephrine for all groups were similar. CONCLUSION: Endothelial cells exposed to thrombin have increased arginase I messenger RNA and protein levels. Arterial thrombosis causes endothelial dysfunction without affecting smooth muscle responsiveness. Arginase blockade can lead to normalization of arterial vasomotor function.


Arginase/antagonists & inhibitors , Arterial Occlusive Diseases/pathology , Endothelial Cells/physiology , Endothelium, Vascular/physiopathology , Thrombin/pharmacology , Analysis of Variance , Animals , Arginase/metabolism , Arterial Occlusive Diseases/physiopathology , Cells, Cultured , Disease Models, Animal , Endothelial Cells/cytology , Endothelial Cells/drug effects , Endothelium, Vascular/cytology , Immunoblotting , Male , Muscle Contraction/physiology , Muscle, Smooth, Vascular/physiology , Probability , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley , Reverse Transcriptase Polymerase Chain Reaction , Sensitivity and Specificity , Vasoconstriction/physiology
...