Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 40
Filter
1.
Nat Immunol ; 23(4): 605-618, 2022 04.
Article in English | MEDLINE | ID: mdl-35352063

ABSTRACT

Autonomic nerves control organ function through the sympathetic and parasympathetic branches, which have opposite effects. In the bone marrow, sympathetic (adrenergic) nerves promote hematopoiesis; however, how parasympathetic (cholinergic) signals modulate hematopoiesis is unclear. Here, we show that B lymphocytes are an important source of acetylcholine, a neurotransmitter of the parasympathetic nervous system, which reduced hematopoiesis. Single-cell RNA sequencing identified nine clusters of cells that expressed the cholinergic α7 nicotinic receptor (Chrna7) in the bone marrow stem cell niche, including endothelial and mesenchymal stromal cells (MSCs). Deletion of B cell-derived acetylcholine resulted in the differential expression of various genes, including Cxcl12 in leptin receptor+ (LepR+) stromal cells. Pharmacologic inhibition of acetylcholine signaling increased the systemic supply of inflammatory myeloid cells in mice and humans with cardiovascular disease.


Subject(s)
Acetylcholine , Hematopoiesis , Animals , B-Lymphocytes , Cholinergic Agents , Hematopoiesis/genetics , Mice , Stem Cell Niche
2.
Cell ; 169(3): 510-522.e20, 2017 04 20.
Article in English | MEDLINE | ID: mdl-28431249

ABSTRACT

Organ-specific functions of tissue-resident macrophages in the steady-state heart are unknown. Here, we show that cardiac macrophages facilitate electrical conduction through the distal atrioventricular node, where conducting cells densely intersperse with elongated macrophages expressing connexin 43. When coupled to spontaneously beating cardiomyocytes via connexin-43-containing gap junctions, cardiac macrophages have a negative resting membrane potential and depolarize in synchrony with cardiomyocytes. Conversely, macrophages render the resting membrane potential of cardiomyocytes more positive and, according to computational modeling, accelerate their repolarization. Photostimulation of channelrhodopsin-2-expressing macrophages improves atrioventricular conduction, whereas conditional deletion of connexin 43 in macrophages and congenital lack of macrophages delay atrioventricular conduction. In the Cd11bDTR mouse, macrophage ablation induces progressive atrioventricular block. These observations implicate macrophages in normal and aberrant cardiac conduction.


Subject(s)
Heart Conduction System , Macrophages/physiology , Animals , Connexin 43/metabolism , Female , Heart Atria/cytology , Humans , Male , Mice , Mice, Inbred C57BL , Middle Aged , Myocytes, Cardiac/physiology
4.
Immunity ; 51(5): 899-914.e7, 2019 11 19.
Article in English | MEDLINE | ID: mdl-31732166

ABSTRACT

Myocardial infarction, stroke, and sepsis trigger systemic inflammation and organism-wide complications that are difficult to manage. Here, we examined the contribution of macrophages residing in vital organs to the systemic response after these injuries. We generated a comprehensive catalog of changes in macrophage number, origin, and gene expression in the heart, brain, liver, kidney, and lung of mice with myocardial infarction, stroke, or sepsis. Predominantly fueled by heightened local proliferation, tissue macrophage numbers increased systemically. Macrophages in the same organ responded similarly to different injuries by altering expression of tissue-specific gene sets. Preceding myocardial infarction improved survival of subsequent pneumonia due to enhanced bacterial clearance, which was caused by IFNÉ£ priming of alveolar macrophages. Conversely, EGF receptor signaling in macrophages exacerbated inflammatory lung injury. Our data suggest that local injury activates macrophages in remote organs and that targeting macrophages could improve resilience against systemic complications following myocardial infarction, stroke, and sepsis.


Subject(s)
Disease Susceptibility , Macrophages/immunology , Macrophages/metabolism , Animals , Biomarkers , Cell Count , Disease Susceptibility/immunology , ErbB Receptors/metabolism , Gene Expression Profiling , Gene Expression Regulation , Gene Regulatory Networks , Ischemia/etiology , Ischemia/metabolism , Macrophages, Alveolar/immunology , Macrophages, Alveolar/metabolism , Mice , Muscle Cells/immunology , Muscle Cells/metabolism , Myocardial Infarction/etiology , Myocardial Infarction/metabolism , Organ Specificity/genetics , Organ Specificity/immunology , Pneumonia/etiology , Pneumonia/metabolism , Pneumonia/pathology
5.
Circulation ; 150(1): 49-61, 2024 Jul 02.
Article in English | MEDLINE | ID: mdl-38506045

ABSTRACT

BACKGROUND: Viral infections can cause acute respiratory distress syndrome (ARDS), systemic inflammation, and secondary cardiovascular complications. Lung macrophage subsets change during ARDS, but the role of heart macrophages in cardiac injury during viral ARDS remains unknown. Here we investigate how immune signals typical for viral ARDS affect cardiac macrophage subsets, cardiovascular health, and systemic inflammation. METHODS: We assessed cardiac macrophage subsets using immunofluorescence histology of autopsy specimens from 21 patients with COVID-19 with SARS-CoV-2-associated ARDS and 33 patients who died from other causes. In mice, we compared cardiac immune cell dynamics after SARS-CoV-2 infection with ARDS induced by intratracheal instillation of Toll-like receptor ligands and an ACE2 (angiotensin-converting enzyme 2) inhibitor. RESULTS: In humans, SARS-CoV-2 increased total cardiac macrophage counts and led to a higher proportion of CCR2+ (C-C chemokine receptor type 2 positive) macrophages. In mice, SARS-CoV-2 and virus-free lung injury triggered profound remodeling of cardiac resident macrophages, recapitulating the clinical expansion of CCR2+ macrophages. Treating mice exposed to virus-like ARDS with a tumor necrosis factor α-neutralizing antibody reduced cardiac monocytes and inflammatory MHCIIlo CCR2+ macrophages while also preserving cardiac function. Virus-like ARDS elevated mortality in mice with pre-existing heart failure. CONCLUSIONS: Our data suggest that viral ARDS promotes cardiac inflammation by expanding the CCR2+ macrophage subset, and the associated cardiac phenotypes in mice can be elicited by activating the host immune system even without viral presence in the heart.


Subject(s)
COVID-19 , Cardiomyopathies , Respiratory Distress Syndrome , SARS-CoV-2 , COVID-19/immunology , COVID-19/complications , COVID-19/pathology , Animals , Humans , Respiratory Distress Syndrome/immunology , Respiratory Distress Syndrome/etiology , Respiratory Distress Syndrome/pathology , Respiratory Distress Syndrome/virology , Mice , Male , Female , Cardiomyopathies/immunology , Cardiomyopathies/etiology , Cardiomyopathies/pathology , Cardiomyopathies/virology , Macrophages/immunology , Macrophages/pathology , Macrophages/metabolism , Inflammation/pathology , Middle Aged , Myocardium/pathology , Myocardium/immunology , Mice, Inbred C57BL , Aged
6.
Eur Heart J ; 42(39): 4077-4088, 2021 10 14.
Article in English | MEDLINE | ID: mdl-34279021

ABSTRACT

AIMS: Mental stress substantially contributes to the initiation and progression of human disease, including cardiovascular conditions. We aim to investigate the underlying mechanisms of these contributions since they remain largely unclear. METHODS AND RESULTS: Here, we show in humans and mice that leucocytes deplete rapidly from the blood after a single episode of acute mental stress. Using cell-tracking experiments in animal models of acute mental stress, we found that stress exposure leads to prompt uptake of inflammatory leucocytes from the blood to distinct tissues including heart, lung, skin, and, if present, atherosclerotic plaques. Mechanistically, we found that acute stress enhances leucocyte influx into mouse atherosclerotic plaques by modulating endothelial cells. Specifically, acute stress increases adhesion molecule expression and chemokine release through locally derived norepinephrine. Either chemical or surgical disruption of norepinephrine signalling diminished stress-induced leucocyte migration into mouse atherosclerotic plaques. CONCLUSION: Our data show that acute mental stress rapidly amplifies inflammatory leucocyte expansion inside mouse atherosclerotic lesions and promotes plaque vulnerability.


Subject(s)
Atherosclerosis , Plaque, Atherosclerotic , Animals , Disease Models, Animal , Endothelial Cells , Inflammation , Mice , Mice, Inbred C57BL , Mice, Knockout
7.
Circulation ; 142(25): 2443-2455, 2020 12 22.
Article in English | MEDLINE | ID: mdl-33092403

ABSTRACT

BACKGROUND: Ibrutinib is a Bruton tyrosine kinase inhibitor with remarkable efficacy against B-cell cancers. Ibrutinib also increases the risk of atrial fibrillation (AF), which remains poorly understood. METHODS: We performed electrophysiology studies on mice treated with ibrutinib to assess inducibility of AF. Chemoproteomic analysis of cardiac lysates identified candidate ibrutinib targets, which were further evaluated in genetic mouse models and additional pharmacological experiments. The pharmacovigilance database, VigiBase, was queried to determine whether drug inhibition of an identified candidate kinase was associated with increased reporting of AF. RESULTS: We demonstrate that treatment of mice with ibrutinib for 4 weeks results in inducible AF, left atrial enlargement, myocardial fibrosis, and inflammation. This effect was reproduced in mice lacking Bruton tyrosine kinase, but not in mice treated with 4 weeks of acalabrutinib, a more specific Bruton tyrosine kinase inhibitor, demonstrating that AF is an off-target side effect. Chemoproteomic profiling identified a short list of candidate kinases that was narrowed by additional experimentation leaving CSK (C-terminal Src kinase) as the strongest candidate for ibrutinib-induced AF. Cardiac-specific Csk knockout in mice led to increased AF, left atrial enlargement, fibrosis, and inflammation, phenocopying ibrutinib treatment. Disproportionality analyses in VigiBase confirmed increased reporting of AF associated with kinase inhibitors blocking Csk versus non-Csk inhibitors, with a reporting odds ratio of 8.0 (95% CI, 7.3-8.7; P<0.0001). CONCLUSIONS: These data identify Csk inhibition as the mechanism through which ibrutinib leads to AF. Registration: URL: https://ww.clinicaltrials.gov; Unique identifier: NCT03530215.


Subject(s)
Adenine/analogs & derivatives , Antineoplastic Agents/toxicity , Atrial Fibrillation/chemically induced , Atrial Function, Left/drug effects , CSK Tyrosine-Protein Kinase/antagonists & inhibitors , Heart Atria/drug effects , Heart Rate/drug effects , Piperidines/toxicity , Protein Kinase Inhibitors/toxicity , Action Potentials/drug effects , Adenine/toxicity , Agammaglobulinaemia Tyrosine Kinase/deficiency , Agammaglobulinaemia Tyrosine Kinase/genetics , Animals , Atrial Fibrillation/enzymology , Atrial Fibrillation/physiopathology , CSK Tyrosine-Protein Kinase/genetics , CSK Tyrosine-Protein Kinase/metabolism , Databases, Genetic , Heart Atria/enzymology , Heart Atria/physiopathology , Humans , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Inbred CBA , Mice, Knockout , Risk Assessment , Risk Factors
8.
Circ Res ; 124(9): 1372-1385, 2019 04 26.
Article in English | MEDLINE | ID: mdl-30782088

ABSTRACT

RATIONALE: After a stroke, patients frequently experience altered systemic immunity resulting in peripheral immunosuppression and higher susceptibility to infections, which is at least partly attributed to lymphopenia. The mechanisms that profoundly change the systemic leukocyte repertoire after stroke are incompletely understood. Emerging evidence indicates that stroke alters hematopoietic output of the bone marrow. OBJECTIVE: To explore the mechanisms that lead to defects of B lymphopoiesis after ischemic stroke. METHODS AND RESULTS: We here report that ischemic stroke triggers brain-bone marrow communication via hormonal long-range signals that regulate hematopoietic B lineage decisions. Bone marrow fluorescence-activated cell sorter analyses and serial intravital microscopy indicate that transient middle cerebral artery occlusion in mice arrests B-cell development beginning at the pro-B-cell stage. This phenotype was not rescued in Myd88-/- and TLR4-/- mice with disrupted TLR (Toll-like receptor) signaling or after blockage of peripheral sympathetic nerves. Mechanistically, we identified stroke-induced glucocorticoid release as the main instigator of B lymphopoiesis defects. B-cell lineage-specific deletion of the GR (glucocorticoid receptor) in CD19-Cre loxP Nr3c1 mice attenuated lymphocytopenia after transient middle cerebral artery. In 20 patients with acute stroke, increased cortisol levels inversely correlated with blood lymphocyte numbers. CONCLUSIONS: Our data demonstrate that the hypothalamic-pituitary-adrenal axis mediates B lymphopoiesis defects after ischemic stroke.


Subject(s)
Adrenal Cortex Hormones/blood , B-Lymphocytes/metabolism , Bone Marrow Cells/metabolism , Lymphopoiesis , Receptors, Glucocorticoid/blood , Stroke/blood , Aged , Animals , B-Lymphocytes/cytology , Bone Marrow/metabolism , Bone Marrow Cells/cytology , Female , Humans , Hypothalamo-Hypophyseal System/physiopathology , Male , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Middle Aged , Pituitary-Adrenal System/physiopathology , Receptors, Glucocorticoid/genetics , Receptors, Glucocorticoid/metabolism , Stroke/genetics , Stroke/physiopathology
9.
Circ Res ; 123(11): 1208-1219, 2018 11 09.
Article in English | MEDLINE | ID: mdl-30571465

ABSTRACT

RATIONALE: Cardiac pacing is a critical technology for the treatment of arrhythmia and heart failure. The impact of specific pacing strategies on myocardial function is an area of intense research and high clinical significance. Mouse models have proven extremely useful for probing mechanisms of heart disease, but there is currently no reliable technology for long-term pacing in the mouse. OBJECTIVE: We sought to develop a device for long-term pacing studies in mice. We evaluated the device for (1) treating third-degree atrioventricular block after macrophage depletion, (2) ventricular pacing-induced cardiomyopathy, and (3) high-rate atrial pacing. METHODS AND RESULTS: We developed a mouse pacemaker by refashioning a 26 mm×6.7 mm clinical device powered by a miniaturized, highly efficient battery. The electrode was fitted with a single flexible lead, and custom software extended the pacing rate to up to 1200 bpm. The wirelessly programmable device was implanted in the dorsal subcutaneous space of 39 mice. The tunneled lead was passed through a left thoracotomy incision and attached to the epicardial surface of the apex (for ventricular pacing) or the left atrium (for atrial pacing). Mice tolerated the implantation and both long-term atrial and ventricular pacing over weeks. We then validated the pacemaker's suitability for the treatment of atrioventricular block after macrophage depletion in Cd11b DTR mice. Ventricular pacing increased the heart rate from 313±59 to 550 bpm ( P<0.05). In addition, we characterized tachypacing-induced cardiomyopathy in mice. Four weeks of ventricular pacing resulted in reduced left ventricular function, fibrosis, and an increased number of cardiac leukocytes and endothelial activation. Finally, we demonstrated the feasibility of chronic atrial pacing at 1200 bpm. CONCLUSIONS: Long-term pacing with a fully implantable, programmable, and battery-powered device enables previously impossible investigations of arrhythmia and heart failure in the mouse.


Subject(s)
Arrhythmias, Cardiac/physiopathology , Pacemaker, Artificial , Telemetry/methods , Animals , Electrodes, Implanted , Equipment Design , Female , Male , Mice , Mice, Inbred C57BL , Miniaturization , Software , Time
11.
Bioconjug Chem ; 30(3): 733-740, 2019 03 20.
Article in English | MEDLINE | ID: mdl-30615425

ABSTRACT

Amiodarone is an effective antiarrhythmic drug used to treat and prevent different types of cardiac arrhythmias. However, amiodarone can have considerable side effects resulting from accumulation in off-target tissues. Cardiac macrophages are highly prevalent tissue-resident immune cells with importance in homeostatic functions, including immune response and modulation of cardiac conduction. We hypothesized that amiodarone could be more efficiently delivered to the heart via cardiac macrophages, an important step toward reducing overall dose and off-target tissue accumulation. Toward this goal, we synthesized a nanoparticle drug carrier composed of l-lysine cross-linked succinyl-ß-cyclodextrin that demonstrates amiodarone binding through supramolecular host-guest interaction as well as a high macrophage affinity. Biodistribution analyses at the organ and single-cell level demonstrate accumulation of nanoparticles in the heart resulting from rapid uptake by cardiac macrophages. Nanoparticle assisted delivery of amiodarone resulted in a 250% enhancement in the selective delivery of the drug to cardiac tissue in part due to a concomitant decrease of pulmonary accumulation, the main source of off-target toxicity.


Subject(s)
Amiodarone/administration & dosage , Anti-Arrhythmia Agents/administration & dosage , Drug Carriers , Heart/drug effects , Amiodarone/pharmacokinetics , Amiodarone/therapeutic use , Animals , Anti-Arrhythmia Agents/pharmacokinetics , Anti-Arrhythmia Agents/therapeutic use , Mice , Mice, Inbred C57BL , Microscopy, Fluorescence , RAW 264.7 Cells , Tissue Distribution , beta-Cyclodextrins/pharmacokinetics
12.
Circ Res ; 119(7): 853-64, 2016 Sep 16.
Article in English | MEDLINE | ID: mdl-27444755

ABSTRACT

RATIONALE: Macrophages reside in the healthy myocardium, participate in ischemic heart disease, and modulate myocardial infarction (MI) healing. Their origin and roles in post-MI remodeling of nonischemic remote myocardium, however, remain unclear. OBJECTIVE: This study investigated the number, origin, phenotype, and function of remote cardiac macrophages residing in the nonischemic myocardium in mice with chronic heart failure after coronary ligation. METHODS AND RESULTS: Eight weeks post MI, fate mapping and flow cytometry revealed that a 2.9-fold increase in remote macrophages results from both increased local macrophage proliferation and monocyte recruitment. Heart failure produced by extensive MI, through activation of the sympathetic nervous system, expanded medullary and extramedullary hematopoiesis. Circulating Ly6C(high) monocytes rose from 64±5 to 108±9 per microliter of blood (P<0.05). Cardiac monocyte recruitment declined in Ccr2(-/-) mice, reducing macrophage numbers in the failing myocardium. Mechanical strain of primary murine and human macrophage cultures promoted cell cycle entry, suggesting that the increased wall tension in post-MI heart failure stimulates local macrophage proliferation. Strained cells activated the mitogen-activated protein kinase pathway, whereas specific inhibitors of this pathway reduced macrophage proliferation in strained cell cultures and in the failing myocardium (P<0.05). Steady-state cardiac macrophages, monocyte-derived macrophages, and locally sourced macrophages isolated from failing myocardium expressed different genes in a pattern distinct from the M1/M2 macrophage polarization paradigm. In vivo silencing of endothelial cell adhesion molecules curbed post-MI monocyte recruitment to the remote myocardium and preserved ejection fraction (27.4±2.4 versus 19.1±2%; P<0.05). CONCLUSIONS: Myocardial failure is influenced by an altered myeloid cell repertoire.


Subject(s)
Biomechanical Phenomena/physiology , Cell Proliferation/physiology , Heart Failure/pathology , Macrophages/physiology , Myocardium/cytology , Animals , Cells, Cultured , Chronic Disease , Female , Heart Failure/physiopathology , Humans , Male , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Transgenic
13.
J Mol Cell Cardiol ; 93: 149-55, 2016 04.
Article in English | MEDLINE | ID: mdl-26593722

ABSTRACT

The mammalian heart contains a population of resident macrophages that expands in response to myocardial infarction and hemodynamic stress. This expansion occurs likely through both local macrophage proliferation and monocyte recruitment. Given the role of macrophages in tissue remodeling, their contribution to adaptive processes in the heart is conceivable but currently poorly understood. In this review, we discuss monocyte and macrophage heterogeneity associated with cardiac stress, the cell's potential contribution to the pathogenesis of cardiac fibrosis, and describe different tools to study and characterize these innate immune cells. Finally, we highlight their potential role as therapeutic targets.


Subject(s)
Macrophages/immunology , Macrophages/metabolism , Monocytes/immunology , Monocytes/metabolism , Ventricular Remodeling , Animals , Biomarkers , Cell Communication , Cell Proliferation , Fibrosis , Humans , Myocardium/metabolism , Myocardium/pathology , Phenotype , Signal Transduction , Translational Research, Biomedical
14.
Circulation ; 132(20): 1880-90, 2015 Nov 17.
Article in English | MEDLINE | ID: mdl-26358260

ABSTRACT

BACKGROUND: Myocardial infarction (MI) is an ischemic wound that recruits millions of leukocytes. MI-associated blood leukocytosis correlates inversely with patient survival, yet the signals driving heightened leukocyte production after MI remain incompletely understood. METHODS AND RESULTS: With the use of parabiosis surgery, this study shows that soluble danger signals, among them interleukin-1ß, increase bone marrow hematopoietic stem cell proliferation after MI. Data obtained in bone marrow reconstitution experiments reveal that interleukin-1ß enhances hematopoietic stem cell proliferation by both direct actions on hematopoietic cells and through modulation of the bone marrow's hematopoietic microenvironment. An antibody that neutralizes interleukin-1ß suppresses these effects. Anti-interleukin-1ß treatment dampens the post-MI increase in hematopoietic stem cell proliferation. Consequently, decreased leukocyte numbers in the blood and infarct reduce inflammation and diminish post-MI heart failure in ApoE(-/-) mice with atherosclerosis. CONCLUSIONS: The presented insight into post-MI bone marrow activation identifies a mechanistic target for muting inflammation in the ischemically damaged heart.


Subject(s)
Drug Delivery Systems/methods , Interleukin-1beta/antagonists & inhibitors , Leukocytes/pathology , Myocardial Infarction/pathology , Myocardial Infarction/prevention & control , Animals , Cell Proliferation/drug effects , Cell Proliferation/physiology , Hematopoietic Stem Cells/drug effects , Hematopoietic Stem Cells/metabolism , Interleukin-1beta/metabolism , Leukocytes/metabolism , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Myocardial Infarction/metabolism , Recombinant Proteins/administration & dosage
16.
Science ; 381(6654): 231-239, 2023 07 14.
Article in English | MEDLINE | ID: mdl-37440641

ABSTRACT

Atrial fibrillation disrupts contraction of the atria, leading to stroke and heart failure. We deciphered how immune and stromal cells contribute to atrial fibrillation. Single-cell transcriptomes from human atria documented inflammatory monocyte and SPP1+ macrophage expansion in atrial fibrillation. Combining hypertension, obesity, and mitral valve regurgitation (HOMER) in mice elicited enlarged, fibrosed, and fibrillation-prone atria. Single-cell transcriptomes from HOMER mouse atria recapitulated cell composition and transcriptome changes observed in patients. Inhibiting monocyte migration reduced arrhythmia in Ccr2-∕- HOMER mice. Cell-cell interaction analysis identified SPP1 as a pleiotropic signal that promotes atrial fibrillation through cross-talk with local immune and stromal cells. Deleting Spp1 reduced atrial fibrillation in HOMER mice. These results identify SPP1+ macrophages as targets for immunotherapy in atrial fibrillation.


Subject(s)
Atrial Fibrillation , Macrophages , Osteopontin , Animals , Humans , Mice , Atrial Fibrillation/genetics , Atrial Fibrillation/immunology , Heart Atria , Macrophages/immunology , Mitral Valve Insufficiency/genetics , Osteopontin/genetics , Gene Deletion , Cell Movement , Single-Cell Gene Expression Analysis
17.
Nat Cardiovasc Res ; 2(12): 1277-1290, 2023 Dec.
Article in English | MEDLINE | ID: mdl-38344689

ABSTRACT

After myocardial infarction (MI), emergency hematopoiesis produces inflammatory myeloid cells that accelerate atherosclerosis and promote heart failure. Since the balance between glycolysis and mitochondrial metabolism regulates hematopoietic stem cell homeostasis, metabolic cues may influence emergency myelopoiesis. Here, we show in humans and female mice that hematopoietic progenitor cells increase fatty acid metabolism after MI. Blockade of fatty acid oxidation by deleting carnitine palmitoyltransferase (Cpt1A) in hematopoietic cells of Vav1Cre/+Cpt1Afl/fl mice limited hematopoietic progenitor proliferation and myeloid cell expansion after MI. We also observed reduced bone marrow adiposity in humans, pigs and mice following MI. Inhibiting lipolysis in adipocytes using AdipoqCreERT2Atglfl/fl mice or local depletion of bone marrow adipocytes in AdipoqCreERT2iDTR mice also curbed emergency hematopoiesis. Furthermore, systemic and regional sympathectomy prevented bone marrow adipocyte shrinkage after MI. These data establish a critical role for fatty acid metabolism in post-MI emergency hematopoiesis.

18.
Curr Atheroscler Rep ; 14(3): 264-76, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22350585

ABSTRACT

High levels of reactive oxygen species (ROS) are observed in chronic human diseases such as obesity, type 2 diabetes, atherosclerosis, and cardiovascular diseases. In addition to the presence of oxidative stress, these diseases are also characterized by deregulated inflammatory responses. Our first aim is to discuss distinct molecular pathways that determine the rate of mitochondrial ROS (mtROS) production and identify agents and enzymes that disrupt the balance between ROS generation and ROS elimination. Recent studies exploring the mechanisms linking ROS and inflammation found that ROS derived from mitochondria act as signal-transducing molecules that provoke endothelial dysfunction associated with uncoupling of nitric oxide synthase, induce the infiltration and activation of inflammatory cells, and increase apoptosis of endothelial and vascular smooth muscle cells. Therefore, our second aim is to give a comprehensive overview of the role of mtROS in all these processes contributing to atherosclerotic lesion progression and causing plaque erosion and rupture. Our third aim is to emphasize the role of the inflammatory toll-like receptor 2/NF-κB signaling pathway in the induction of pro-inflammatory cytokines and mtROS production in relation to insulin resistance, type 2 diabetes, and atherosclerosis. Because mtROS play an active role in several pathogenic mechanisms there is need for mitochondria-targeted antioxidants. Preliminary experiments in cell and animal models of cardiovascular diseases showed that some mitochondria-targeted antioxidants indeed reduce ROS production. However, wide-spread use in humans requires the development of specific and sensitive assays to evaluate mitochondrial oxidative stress and the development of orally active compounds.


Subject(s)
Atherosclerosis/metabolism , Mitochondria/metabolism , Oxidative Stress , Reactive Oxygen Species/metabolism , Animals , Humans , Risk Factors , Signal Transduction
19.
FASEB J ; 25(8): 2515-27, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21507901

ABSTRACT

A primary event in atherogenesis is the infiltration of activated inflammatory cells into the arterial wall. There they secrete reactive oxygen species and oxidize lipoproteins, inducing foam cell formation and endothelial cell apoptosis, which in turn lead to plaque growth, erosion, and rupture. In addition, there is evidence that this vicious circle between oxidative stress and inflammation occurs not only in the diseased arterial wall but also in adipose tissues in obesity. In this condition, oxidative stress and inflammation impair adipocyte maturation, resulting in defective insulin action and adipocytokine signaling. This observation raises questions regarding what molecules are probably common regulators of these pathogenic processes in adipose and vascular tissues. Candidates are small, noncoding, microRNAs (miRs) that control gene expression by inducing mRNA degradation or blocking translation. This review summarizes recent insights into the roles of miRs in regulation of oxidative stress and inflammation in vascular and adipose tissues. It emphasizes the role of miR-containing microvesicles in the interaction between inflammatory cells and endothelial cells within these tissues and in communication between these tissues, possibly explaining the similarity and the simultaneity of molecular changes and interactions in adipose and vascular tissues.


Subject(s)
Atherosclerosis/genetics , Inflammation/genetics , MicroRNAs/genetics , Obesity/genetics , Oxidative Stress/genetics , Adipocytes/metabolism , Adipocytes/pathology , Animals , Apoptosis/genetics , Atherosclerosis/metabolism , Atherosclerosis/pathology , Cell Differentiation/genetics , Humans , Microvessels/metabolism , Microvessels/pathology , Models, Biological , Neovascularization, Pathologic/genetics , Obesity/metabolism , Obesity/pathology , Signal Transduction
20.
JCI Insight ; 7(19)2022 10 10.
Article in English | MEDLINE | ID: mdl-35998035

ABSTRACT

Obesity is an important risk factor for atrial fibrillation (AF), but a better mechanistic understanding of obesity-related atrial fibrillation is required. Serum glucocorticoid kinase 1 (SGK1) is a kinase positioned within multiple obesity-related pathways, and prior work has shown a pathologic role of SGK1 signaling in ventricular arrhythmias. We validated a mouse model of obesity-related AF using wild-type mice fed a high-fat diet. RNA sequencing of atrial tissue demonstrated substantial differences in gene expression, with enrichment of multiple SGK1-related pathways, and we showed upregulated of SGK1 transcription, activation, and signaling in obese atria. Mice expressing a cardiac specific dominant-negative SGK1 were protected from obesity-related AF, through effects on atrial electrophysiology, action potential characteristics, structural remodeling, inflammation, and sodium current. Overall, this study demonstrates the promise of targeting SGK1 in a mouse model of obesity-related AF.


Subject(s)
Atrial Fibrillation , Protein Serine-Threonine Kinases , Animals , Mice , Atrial Fibrillation/genetics , Atrial Fibrillation/prevention & control , Disease Models, Animal , Glucocorticoids/metabolism , Heart Atria/pathology , Obesity/complications , Obesity/genetics , Obesity/metabolism , Sodium/metabolism , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL