Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 52
Filtrar
1.
J Leukoc Biol ; 2024 May 24.
Artigo em Inglês | MEDLINE | ID: mdl-38785336

RESUMO

Splenic leukocytes, particularly macrophage-expressed lipoxygenases, facilitate the biosynthesis of resolution mediators essential for cardiac repair. Next, we asked whether deletion of 12/15 lipoxygenase (12/15LOX) in macrophages impedes the resolution of inflammation following myocardial infarction (MI). Using 12/15flox/flox and LysMcre scheme, we generated macrophage-specific 12/15LOX (Mɸ-12/15LOX-/-) mice. Young C57BL/6J wild-type and Mɸ-12/15LOX-/- male mice were subjected to permanent coronary ligation micro-surgery. Mice were monitored at day (d)1-d5 (as acute HF; AHF) and to d56 (chronic HF; CHF) post-MI, maintaining no-MI as d0 naïve controls. Post-ligation, Mɸ-12/15LOX-/- mice showed increased survival (88%vs56%) and limited heart dysfunction compared with WT. In AHF, Mɸ-12/15LOX-/- mice have increased biosynthesis of epoxyeicosatrienoic acid (EETs) by 30%, with the decrease in D-series resolvins, protectin, and maresin by 70% in the infarcted heart. Overall, myeloid cell profiling from the heart and spleen indicated that Mɸ-12/15LOX-/- mice showed higher immune cells with reparative Ly6Clow macrophages during AHF. In addition, the detailed immune profiling revealed reparative macrophage phenotype (Ly6Clow) in Mɸ-12/15LOX-/- mice in a splenocardiac manner post-MI. Mɸ-12/15LOX-/- mice showed an increase in myeloid population that coordinated increase of Tregs (CD4+/Foxp3+) in the spleen and injured heart at CHF compared with WT. Thus, macrophage-specific deletion of 12/15LOX directs reparative macrophage phenotype to facilitate cardiac repair. The presented study outlines the complex role of 12/15LOX in macrophage plasticity, and Treg signaling that indicates resolution mediators are viable targets to facilitate cardiac repair in heart failure post-MI.


Macrophage-derived bioactive lipids promote the safe clearance of inflammation (resolution), thus modulating macrophage-specific 12/15 lipoxygenase restores structure, function, and survival after heart attack in mice.

2.
J Mol Cell Cardiol ; 188: 79-89, 2024 03.
Artigo em Inglês | MEDLINE | ID: mdl-38364731

RESUMO

The study investigated the role of volunteer exercise and an obesogenic diet (OBD) in mice, focusing on the splenocardiac axis and inflammation-resolution signaling. Male C57BL/6J mice (2 months old) were assigned to control (CON) or OBD groups for ten months, then randomized into sedentary (Sed) or exercise (Exe) groups for two weeks. Leukocytes, heart function, structure, and spleen tissue examined for inflammation-resolution mediators and macrophage-centric gene transcripts. After two weeks of volunteer exercise, cardiac function shows limited changes, but structural changes were notable in the heart and spleen. Exercise induced cardiac nuclear hyperplasia observed in both CON and OBD groups. OBD-Sed mice showed splenic changes and increased neutrophils, whereas increased neutrophils were noted in the CON post exercise. OBD-Sed increased pro-inflammatory lipid mediators in the heart, reduced by exercise in OBD-Exe, while CON-Exe preserved resolution mediators. Chronic OBD-Sed depletes long chain fatty acids (DHA/EPA) in the heart and spleen, while exercise independently regulates lipid metabolism genes in both organs, affecting macrophage-centric lipid and lipoprotein pathways. Chronic obesity amplified cardiac inflammation, countered by exercise that lowered pro-inflammatory bioactive lipid mediators in the heart. OBD sustained inflammation in the heart and spleen, while exercise conserved resolution mediators in CON mice. In summary, these findings emphasize the interplay of diet with exercise and highlight the intricate connection of diet, exercise, inflammation-resolution signaling in splenocardiac axis and immune health.


Assuntos
Dieta , Baço , Humanos , Masculino , Animais , Camundongos , Lactente , Camundongos Endogâmicos C57BL , Envelhecimento , Ácidos Graxos , Inflamação , Mediadores da Inflamação
3.
Am J Physiol Heart Circ Physiol ; 325(3): H433-H448, 2023 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-37417877

RESUMO

Chronic and uncleared inflammation is the root cause of various cardiovascular diseases. Fundamentally, acute inflammation is supportive when overlapping with safe clearance of inflammation termed resolution; however, if the lifestyle-directed extrinsic factors such as diet, sleep, exercise, or physical activity are misaligned, that results in unresolved inflammation. Although genetics play a critical role in cardiovascular health, four extrinsic risk factors-unhealthy processed diet, sleep disruption or fragmentation, sedentary lifestyle, thereby, subsequent stress-have been identified as heterogeneous and polygenic triggers of heart failure (HF), which can result in several complications with indications of chronic inflammation. Extrinsic risk factors directly impact endogenous intrinsic factors, such as using fatty acids by immune-responsive enzymes [lipoxygenases (LOXs)/cyclooxygenases (COXs)/cytochromes-P450 (CYP450)] to form resolution mediators that activate specific resolution receptors. Thus, the balance of extrinsic factors such as diet, sleep, and physical activity feed-forward the coordination of intrinsic factors such as fatty acids-enzymes-bioactive lipid receptors that modulates the immune defense, metabolic health, inflammation-resolution signaling, and cardiac health. Future research on lifestyle- and aging-associated molecular patterns is warranted in the context of intrinsic and extrinsic factors, immune fitness, inflammation-resolution signaling, and cardiac health.


Assuntos
Insuficiência Cardíaca , Humanos , Coração , Inflamação/metabolismo , Fatores de Risco , Ácidos Graxos
4.
FASEB J ; 37(5): e22899, 2023 05.
Artigo em Inglês | MEDLINE | ID: mdl-37002889

RESUMO

Sleep is a fundamental medicine for cardiac homeostasis, and sleep-deprived individuals are prone to higher incidences of heart attack. The lipid-dense diet (obesogenic diet-OBD) is a cumulative risk factor for chronic inflammation in cardiovascular disease; thus, understanding how sleep fragmentation (SF) in an obesity setting impacts immune and cardiac health is an unmet medical need. We hypothesized whether the co-existence of SF with OBD dysregulates gut homeostasis and leukocyte-derived reparative/resolution mediators, thereby impairing cardiac repair. Two-month-old male C57BL/6J mice were randomized first into two groups, then four groups; Control, control + SF, OBD, and OBD + SF mice subjected to myocardial infarction (MI). OBD mice had higher levels of plasma linolenic acid with a decrease in eicosapentaenoic and docosahexaenoic acid. The OBD mice had lower Lactobacillus johnsonii indicating a loss of probiotic microbiota. SF in OBD mice increased Firmicutes/Bacteroidetes ratio indicative of a detrimental change in SF-directed microbiome. OBD + SF group increased in the neutrophil: lymphocyte ratio suggestive of suboptimal inflammation. As a result of SF, resolution mediators (RvD2, RvD3, RvD5, LXA4 , PD1, and MaR1) decreased and inflammatory mediators (PGD2 , PGE2 , PGF2a , 6k-PGF1a ) were increased in OBD mice post-MI. At the site of infarction, the proinflammatory cytokines Ccl2, IL1ß, and IL-6 were amplified in OBD + SF indicating a robust proinflammatory milieu post-MI. Also, brain circadian genes (Bmal1, Clock) were downregulated in SF-subjected control mice, but remained elevated in OBD mice post-MI. SF superimposed on obesity dysregulated physiological inflammation and disrupted resolving response thereby impaired cardiac repair and signs of pathological inflammation.


Assuntos
Insuficiência Cardíaca , Microbiota , Infarto do Miocárdio , Masculino , Camundongos , Animais , Privação do Sono/complicações , Lipidômica , Camundongos Endogâmicos C57BL , Inflamação/complicações , Insuficiência Cardíaca/etiologia , Infarto do Miocárdio/patologia , Citocinas/genética , Obesidade/complicações
5.
Am J Physiol Heart Circ Physiol ; 323(4): H721-H737, 2022 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-36018758

RESUMO

Arachidonate 5-lipoxygenase (ALOX5)-derived leukotrienes are primary signals of leukocyte activation and inflammation in response to ischemic cardiac injury (MI; myocardial infarction). Using risk-free male C57BL/6J and ALOX5-null mice (8-12 wk), we quantitated leukocytes and ALOX5-derived bioactive lipids of the infarcted left ventricle (LV) and spleen to measure the physiological inflammation and cardiac repair. Our results showed that ALOX5 endogenously generates specialized pro-resolving mediators (SPMs) that facilitate cardiac repair post-MI. Deficiency of ALOX5 leads to increase in cyclooxygenase gene expression, 6-keto prostaglandin F1α, and delayed neutrophil clearance with signs of unresolved inflammation post-MI. Consequently, ALOX5 deficiency impaired the resolution of inflammation and cardiac repair, including increased myocardium rupture post-MI in acute heart failure. On-time ALOX5 activation is critical for leukocyte clearance from the infarcted heart, indicating an essential role of ALOX5 in the resolution of inflammation. In addition, to balance the inflammatory responses, ALOX5 is also necessary for fibroblast signaling, as the ALOX5-deficient fibroblast are prone to fibroblast-to-myofibroblast differentiation leading to defective scar formation in post-MI cardiac repair. Consistent with these findings, ALOX5-null mice showed an overly inflammatory response, defective fibrotic signaling, and unresolved inflammation. These findings are indicative of a critical role of ALOX5 in myocardium healing, inflammation-resolution signaling, cardiac repair, and fibroblast pathophysiology.NEW & NOTEWORTHY Arachidonate 5-lipoxygenase (ALOX5) is critical in synthesizing specialized pro-resolving mediators that facilitate cardiac repair after cardiac injury. Thus, ALOX5 orchestrates the overlapping phases of inflammation and resolution to facilitate myocardium healing in cardiac repair postmyocardial infarction.


Assuntos
Insuficiência Cardíaca , Infarto do Miocárdio , Animais , Araquidonato 5-Lipoxigenase/genética , Araquidonato 5-Lipoxigenase/metabolismo , Inflamação/metabolismo , Mediadores da Inflamação/metabolismo , Leucotrienos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Prostaglandina-Endoperóxido Sintases
6.
Am J Physiol Heart Circ Physiol ; 322(6): H953-H970, 2022 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-35333119

RESUMO

All fats are not created equal, and despite the extensive literature, the effect of fat intake is the most debated question in obesity, cardiovascular, and cardiorenal research. Cellular and molecular mechanisms underlying cardiac dysfunction and consequent heart failure in the setting of obesity are not well understood. Our understanding of how fats are metabolically transformed after nonreperfused myocardial infarction (MI), in particular, is incomplete. Here, using male C57BL/6J mice (2 mo old), we determined the role of omega-6 fatty acids, provided as safflower oil (SO) for 12 wk, followed by supplementation with docosahexaenoic acid (DHA; n-3 fatty acids) for 8 wk before MI. With SO feeding, inflammation resolution was impaired. Specialized proresolving mediators (SPMs) increased in DHA-fed mice to reverse the effects of SO, whereas prostaglandins and thromboxane B2 were reduced in the spleen and amplified multiple resolving mechanisms in heart and kidney post-MI. DHA amplified the number of resolving macrophages and cardiac reparative pathways of the splenocardiac and cardiorenal networks in acute heart failure, with higher Treg cells in chronic heart failure and marked expression of Foxp3+ in the myocardium. Our findings indicate that surplus ingestion of SO intensified systemic, baseline, nonresolving inflammation, and DHA intake dominates splenocardiac resolving phase with the biosynthesis of SPMs and controlled cardiorenal inflammation in heart failure survivor mice.NEW & NOTEWORTHY Chronic and surplus dietary intake of safflower oil (SO) increased plasma creatinine dysregulated post-MI splenocardiac inflammation coincides with the dysfunctional cardiorenal network. In contrast, docosahexaenoic acid (DHA) increases post-MI survival in chronic heart failure. DHA transforms into specialized proresolving mediators (SPMs) and limited proinflammatory prostaglandins and thromboxanes following myocardial infarction (MI). DHA promotes Ly6Clow resolving macrophages and T regulatory cells (Foxp3+) in a splenocardiac manner post-MI.


Assuntos
Insuficiência Cardíaca , Infarto do Miocárdio , Animais , Ácidos Docosa-Hexaenoicos , Fatores de Transcrição Forkhead , Insuficiência Cardíaca/tratamento farmacológico , Insuficiência Cardíaca/etiologia , Insuficiência Cardíaca/metabolismo , Inflamação/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Obesidade/complicações , Prostaglandinas , Óleo de Cártamo
7.
Mol Cell Endocrinol ; 542: 111521, 2022 02 15.
Artigo em Inglês | MEDLINE | ID: mdl-34843898

RESUMO

Heart failure with preserved ejection fraction (HFpEF) has been an emerging type of cardiac disease since the pseudo-left ventricle function is preserved; therefore, challenges in finding the target and treatment. Damage and pathogen-associated molecular patterns (DAMPs and PAMPs) are widely investigated in acute and chronic inflammation in heart failure; however, lifestyle-associated molecular patterns (LAMPs: diet, sleep, exercise), particularly in obesity, remains of interest due to the enormous increase of HFpEF patients. In this review, we covered obesity-related cardiomyopathy, LAMPs, and resolution receptor dysfunction in the context of heart failure with preserved ejection fraction.


Assuntos
Cardiomiopatias , Insuficiência Cardíaca , Humanos , Inflamação , Obesidade/complicações , Volume Sistólico/fisiologia
8.
Geroscience ; 44(3): 1689-1702, 2022 06.
Artigo em Inglês | MEDLINE | ID: mdl-34932185

RESUMO

In the process of physiological cardiac repair, splenic leukocyte-activated lipoxygenases (LOXs) are essential for the biosynthesis of specialized pro-resolving lipid mediators as a segment of an active process of acute inflammation in splenocardiac manner. In contrast, young 12/15LOX-/- mice use a compensatory mechanism that amplifies epoxyeicosatrienoic acid mediators after myocardial infarction, improving cardiac repair, function, and survival. Next, we tested whether deletion of 12/15LOX impacted the genesis of chronic inflammation in progressive aging. To test the risk factor of aging, we used the inter-organ hypothesis and assessed heart and spleen leukocyte population along with the number of inflammation markers in age-related 12/15LOX-/- aging mice (2 months, 6 months, 13 months) and compared with C57BL/6 J (WT; wild type) as controls (2 months). The 12/15LOX-/- aging mice showed an age-related increase in spleen mass (hypersplenism) and decreased marginal zone area. Results suggest increased interstitial fibrosis in the heart marked with the inflammatory mediator (PGD2) level in 12/15LOX-/- aging mice than WT controls. From a cellular perspective, the quantitative measurement of immune cells indicates that heart and spleen leukocytes (CD11b+ and F4/80+ population) were reduced in 12/15LOX-/- aging mice than WT controls. At the molecular level, analyses of cytokines in the heart and spleen suggest amplified IFN-γ, with reduced COX-1, COX-2, and ALOX5 expression in the absence of 12/15LOX-derived mediators in the spleen. Thus, aging of 12/15LOX-/- mice increased spleen mass and altered spleen and heart structure with activation of multiple molecular and cellular pathways contributing to age-related integrative and inter-organ inflammation.


Assuntos
Cardiopatias , Insuficiência Cardíaca , Hiperesplenismo , Envelhecimento , Animais , Cardiopatias/metabolismo , Insuficiência Cardíaca/metabolismo , Hiperesplenismo/metabolismo , Inflamação/metabolismo , Leucócitos/metabolismo , Lipoxigenase/metabolismo , Camundongos , Camundongos Endogâmicos C57BL
9.
Am J Physiol Heart Circ Physiol ; 321(3): H599-H611, 2021 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-34415189

RESUMO

Sphingosine-1-phosphate (S1P) is a bioactive mediator in inflammation. Dysregulated S1P is demonstrated as a cause of heart failure (HF). However, the time-dependent and integrative role of S1P interaction with receptors in HF is unclear after myocardial infarction (MI). In this study, the sphingolipid mediators were quantified in ischemic human hearts. We also measured the time kinetics of these mediators post-MI in murine spleen and heart as an integrative approach to understand the interaction of S1P and respective S1P receptors in the transition of acute (AHF) to chronic HF (CHF). Risk-free 8-12 wk male C57BL/6 mice were subjected to MI surgery, and MI was confirmed by echocardiography and histology. Mass spectrometry was used to quantify sphingolipids in plasma, infarcted heart, spleen of mice, and ischemic and healthy human heart. The physiological cardiac repair was observed in mice with a notable increase of S1P quantity (pmol/g) in the heart and spleen significantly reduced in patients with ischemic HF. The circulating murine S1P levels were increased during AHF and CHF despite lowered substrate in CHF. The S1PR1 receptor expression was observed to coincide with the respective S1P quantity in mice and human hearts. Furthermore, selective S1P1 agonist limited inflammatory markers CCL2 and TNF-α and accelerated reparative markers ARG-1 and YM-1 in macrophages in the presence of Kdo2-Lipid A (KLA; potent inflammatory stimulant). This report demonstrated the importance of S1P/S1PR1 signaling in physiological inflammation during cardiac repair in mice. Alteration in these axes may serve as the signs of pathological remodeling in patients with ischemia.NEW & NOTEWORTHY Previous studies indicate that sphingosine-1-phosphate (S1P) has some role in cardiovascular disease. This study adds quantitative and integrative systems-based approaches that are necessary for discovery and bedside translation. Here, we quantitated sphinganine, sphingosine, sphingosine-1-phosphate (S1P) in mice and human cardiac pathobiology. Interorgan S1P quantity and respective systems-based receptor activation suggest cardiac repair after myocardial infarction. Thus, S1P serves as a therapeutic target for cardiac protection in clinical translation.


Assuntos
Insuficiência Cardíaca/metabolismo , Lisofosfolipídeos/metabolismo , Infarto do Miocárdio/metabolismo , Miócitos Cardíacos/metabolismo , Esfingosina/análogos & derivados , Baço/metabolismo , Animais , Arginase/metabolismo , Células Cultivadas , Quimiocina CCL2/metabolismo , Humanos , Lectinas/metabolismo , Macrófagos/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Miócitos Cardíacos/fisiologia , Regeneração , Esfingosina/metabolismo , Receptores de Esfingosina-1-Fosfato/genética , Receptores de Esfingosina-1-Fosfato/metabolismo , Fator de Necrose Tumoral alfa/metabolismo , beta-N-Acetil-Hexosaminidases/metabolismo
10.
Biomed Pharmacother ; 139: 111574, 2021 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-33862495

RESUMO

The presistent increase of 12/15 lipoxygenase enzyme activity is correlated with uncontrolled inflammation, leading to organ dysfunction. ML351, a potent 12/15 lipoxygenase (12/15LOX) inhibitor, was reported to reduce infarct size and inflammation in a murine ischemic stroke model. In the presented work, we have applied three complementary experimental approaches, in-vitro, ex-vivo, and in-vivo, to determine whether pharmacological inhibition of 12/15LOX could dampen the inflammatory response in adult mice after Kdo2-Lipid A (KLA) as an endotoxin stimulator or post myocardial infarction (MI). Male C57BL/6 (8-12 weeks) mice were subjected to permanent coronary ligation thereby inducing acute heart failure (MI-d1 and MI-d5) for in-vivo studies. 12/15LOX antagonist ML351 (50 mg/kg) was subcutaneously injected 2 h post-MI, while MI-controls received saline. For ex-vivo experiments, ML351 (25 mg/kg) was injected as bolus after 5 min of inflammatory stimulus (KLA 1 µg/g) injection. Peritoneal macrophages (PMɸ) were harvested after 4 h post KLA. For in-vitro studies, PMɸ were treated with KLA (100 ng/mL), ML351 (10 µM), or KLA + ML351 for 4 h, and inflammatory response was evaluated. In-vivo, 5LOX expression was reduced after ML351 administration, inducing a compensatory increase of 12LOX that sensitized PMɸ toward a proinflammatory state. This was marked by higher inflammatory cytokines and dysregulation of the splenocardiac axis post-MI. ML351 treatment increased CD11b+ and Ly6Chigh populations in spleen and Ly6G+ population in heart, with a decrease in F4/80+ macrophage population at MI-d1. In-vitro results indicated that ML351 suppressed initiation of inflammation while ex-vivo results suggested ML351 overactivated inflammation consequently delaying the resolution process. Collectively, in-vitro, ex-vivo, and in-vivo results indicated that pharmacological blockade of lipoxygenases using ML351 impaired initiation of inflammation thereby dysregulated acute immune response in cardiac repair.


Assuntos
Anti-Inflamatórios não Esteroides/farmacologia , Insuficiência Cardíaca/tratamento farmacológico , Isoxazóis/farmacologia , Isoxazóis/uso terapêutico , Inibidores de Lipoxigenase/farmacologia , Naftalenos/farmacologia , Naftalenos/uso terapêutico , Animais , Anti-Inflamatórios não Esteroides/uso terapêutico , Araquidonato 12-Lipoxigenase , Araquidonato 15-Lipoxigenase , Araquidonato 5-Lipoxigenase/metabolismo , Ecocardiografia , Insuficiência Cardíaca/fisiopatologia , Imunidade Inata , Inflamação/patologia , Inibidores de Lipoxigenase/uso terapêutico , Ativação de Macrófagos/efeitos dos fármacos , Macrófagos Peritoneais/efeitos dos fármacos , Macrófagos Peritoneais/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Infarto do Miocárdio/induzido quimicamente , Infarto do Miocárdio/tratamento farmacológico , Infarto do Miocárdio/patologia
11.
Theranostics ; 11(6): 2742-2754, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33456570

RESUMO

Aim: Immune responsive 12/15 lipoxygenase (12/15LOX)-orchestrate biosynthesis of essential inflammation-resolution mediators during acute inflammatory response in post-myocardial infarction (MI). Lack of 12/15LOX dampens proinflammatory mediator 12-(S)-hydroxyeicosatetraenoic acid (12-(S)-HETE), improves post-MI survival, through the biosynthesis of endogenous mediators epoxyeicosatrienoic acids (EETs; cypoxins) to resolve post-MI inflammation. However, the mechanism that amplifies cypoxins-directed cardiac repair in acute heart failure (AHF) and chronic HF (CHF) remains of interest in MI-directed renal inflammation. Therefore, we determined the role of EETs in macrophage-specific receptor activation in facilitating cardiac repair in 12/15LOX deficient mice experiencing HF. Methods and Results: Risk-free young adult (8 -12 week-old) male C57BL/6J wild-type mice (WT; n = 43) and 12/15LOX-/- mice (n = 31) were subjected to permanent coronary artery ligation and monitored at day (d)1, d5 (as acute HF), and d28 to d56 (8 weeks; chronic HF) post-surgery maintaining no-MI mice that served as d0 naïve controls. Left ventricle (LV) infarcted area of 12/15LOX-/- mice displayed an increase in expression of prostanoid receptor EP4 along with monocyte chemoattractant protein-1 CCL2 in AHF and CHF. The transcriptome analysis of isolated leukocytes (macrophages/neutrophils) from infarcted LV revealed a higher expression of EP4 on reparative macrophages expressing MRC-1 in 12/15LOX-/- mice. Deletion of 12/15LOX differentially modulated the miRNA levels, downregulating miR-23a-3p (~20 fold; p < 0.05) and upregulating miR-125a-5p (~160 fold; p < 0.05) in AHF which promoted polarization of the macrophages towards reparative phenotype. Furthermore, 12/15LOX deletion markedly attenuated renal inflammation with reduced levels of NGAL and KIM-1 and apoptotic markers in the kidney during CHF. Conclusion: In risk-free mice during physiological cardiac repair, absence of 12/15LOX promoted reparative macrophages with marked activation of EP4 signaling thereby improving post-MI survival and limiting renal inflammation in acute and advanced HF. The future studies are warranted to advance the role of EETs in macrophage receptor biology.


Assuntos
Insuficiência Cardíaca/metabolismo , Lipoxigenase/deficiência , Receptores de Prostaglandina E Subtipo EP4/metabolismo , Animais , Apoptose/fisiologia , Araquidonato 12-Lipoxigenase/metabolismo , Araquidonato 15-Lipoxigenase/metabolismo , Biomarcadores/metabolismo , Linhagem Celular Tumoral , Doença Crônica , Regulação para Baixo/fisiologia , Coração/fisiologia , Ventrículos do Coração/metabolismo , Humanos , Inflamação/metabolismo , Mediadores da Inflamação/metabolismo , Macrófagos/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , MicroRNAs/metabolismo , Infarto do Miocárdio/metabolismo , Neutrófilos/metabolismo , Regulação para Cima/fisiologia , Remodelação Ventricular/fisiologia
12.
FASEB J ; 34(8): 10560-10573, 2020 08.
Artigo em Inglês | MEDLINE | ID: mdl-32543720

RESUMO

Chronic unresolved inflammation is the primary determinant of cardiovascular disease. Precise mechanisms that define the genesis of unresolved inflammation in heart failure with preserved ejection fraction (HFpEF) are of interest due to the obesity epidemic. To examine the obesity phenotype and its direct/indirect consequences, multiple approaches were employed using the lipoxin receptor (abbreviated as ALX) dysfunction mouse model. Indirect calorimetry analyses revealed that the deletion of ALX dysregulated energy metabolism driving toward age-related obesity. Heart function data suggest that obesity-prone ALX deficient mice had impaired myocardium strain. Comprehensive measurement of chemokines, extracellular matrix, and arrhythmogenic arrays confirmed the dysregulation of multiple ion channels gene expression with amplified inflammatory chemokines and cytokines response at the age of 4 months compared with WT counterparts. Quantitative analyses of leukocytes demonstrated an increase of proinflammatory Ly6Chi CCR2+ macrophages in the spleen and heart at a steady-state resulting in an inflamed splenocardiac axis. Signs of subtle inflammation were marked with cardiorenal, endothelial defects with decreased CD31 and eNOS and an increased iNOS and COX2 expression. Thus, ALX receptor deficiency serves as an experimental model that defines multiple cellular and molecular mechanisms in HFpEF that could be a target for the development of HFpEF therapy in cardiovascular medicine.


Assuntos
Cardiomiopatias/metabolismo , Células Endoteliais/metabolismo , Inflamação/metabolismo , Leucócitos/metabolismo , Obesidade/metabolismo , Receptores de Lipoxinas/metabolismo , Doenças Vasculares/metabolismo , Animais , Cardiomiopatias/patologia , Células Endoteliais/patologia , Coração/fisiopatologia , Insuficiência Cardíaca/metabolismo , Insuficiência Cardíaca/patologia , Inflamação/patologia , Leucócitos/patologia , Macrófagos/metabolismo , Macrófagos/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Obesos , Miocárdio/metabolismo , Miocárdio/patologia , Obesidade/patologia , Baço/metabolismo , Baço/patologia , Doenças Vasculares/patologia
13.
ESC Heart Fail ; 7(4): 1700-1710, 2020 08.
Artigo em Inglês | MEDLINE | ID: mdl-32363774

RESUMO

AIMS: Leucocyte-directed specialized pro-resolving mediators (SPMs) are essential for cardiac repair, and their biosynthesis coincides with the expression of pro-inflammatory mediators; however, the precise quantitation during an acute myocardial infarction (MI) event is poorly understood in race-specific and sex-specific manner. Coronary heart disease is the leading cause of death and disability in the USA. Although the prevalence of coronary heart disease is similar between Black and White patients, cardiovascular events (including MI), rehospitalization, and mortality are disproportionately higher in Black patients. Therefore, understanding differences in inflammation and resolution can enable the development of predictive, personalized, and precise treatment and attenuate sex/racial disparities. Thus, herein, we assess differences in bioactive lipids and SPMs, between Black and White patients experiencing an acute MI. METHODS AND RESULTS: From the PRiME-GGAT cohort, we collected plasma after MI within 24-48 h from 22 Black (15 male and 7 female) and 31 White (23 male and 8 female) subjects for a comparative race-based and sex-based analyses. MI was confirmed using a biochemical measurement of plasma troponin and ST elevation. Plasma levels of three essential polyunsaturated fatty acids [arachidonic acid (AA), docosahexaenoic acid (DHA), and eicosapentaenoic acid (EPA)] and a set of 40 bioactive lipid mediators with major emphasis on SPMs were quantified by liquid chromatography-mass spectrometry. AA and DHA were higher in White male and female patients, and EPA was noted higher only in White male patients compared with White female and Black male and female patients. Lipoxygenase-mediated AA-derived 12-hydroxyeicosatetraenoic acid (29-63%) and 15-hydroxyeicosatetraenoic acid (3-9%) and DHA-derived 17-hydroxydocosahexaenoic acid (3-22%) and 14-hydroxydocosahexaenoic acid (7-10%) were major bioactive lipid mediators in plasma. The SPM signature resolvin E1 was significantly lower in Black patients compared with White male and female patients, whereas protectin D1 was lower in White male patients compared with White female and Black male and female patients. CONCLUSION: Our comparative analyses of fatty acids and respective cyclooxygenase-derived and lipoxygenase-derived SPM signatures capture the heterogeneity of disease pathology and elucidate potential mechanisms underlying sex-based and race-based differences following MI.


Assuntos
Infarto do Miocárdio , Feminino , Humanos , Inflamação , Masculino , Espectrometria de Massas , Infarto do Miocárdio/epidemiologia , Caracteres Sexuais
14.
J Am Heart Assoc ; 9(8): e015672, 2020 04 21.
Artigo em Inglês | MEDLINE | ID: mdl-32295449

RESUMO

Background Leukocyte-directed biosynthesis of specialized proresolving mediators (SPMs) orchestrates physiological inflammation after myocardial infarction. Deficiency of SPMs drives pathological and nonresolving inflammation, leading to heart failure (HF). Differences in SPMs and inflammatory responses caused by sex-specific differences are of interest. We differentiated leukocyte-directed biosynthesis of lipid mediators in male and female mice, focusing on leukocyte populations, structural remodeling, functional recovery, and survival rates. Methods and Results Risk-free male and female C57BL/6 mice were selected as naïve controls or subjected to myocardial infarction surgery. Molecular and cellular mechanisms that differentiate survival, heart function, and structure and leukocyte-directed lipid mediators were quantified to describe physiological inflammation after myocardial infarction. Female mice show improved survival in acute HF but no statistical difference during chronic HF compared with male mice. Female mice improved survival is marked with functional recovery and limited remodeling compared with male mice. Male and female mice are similarly responsive to arachidonate lipoxygenase (LOX-5, LOX-12, LOX-15) or cyclooxygenase (COX-1, COX-2) in acute HF and particularly male infarcted heart had overall increased SPMs. Female cardiac healing is marked with the biosynthesis of differential p450-derived product, particularly 11,12 epoxyeicosatrienoic acid in acute HF. A sex-specific difference of dendritic cells in acute HF is distinct, with limited changes in chronic HF. Conclusions Cardiac repair is marked with increased SPM biosynthesis in male mice and amplified epoxyeicosatrienoic acid in female mice. Female mice showed improved survival, functional recovery, and limited remodeling, which are signs of fine-tuned physiological inflammation after myocardial infarction. These results rationalize the sex-specific precise therapies and differential treatments in acute and chronic HF.


Assuntos
Mediadores da Inflamação/metabolismo , Leucócitos/metabolismo , Infarto do Miocárdio/metabolismo , Miocárdio/metabolismo , Animais , Modelos Animais de Doenças , Ácido Eicosapentaenoico/metabolismo , Feminino , Leucócitos/imunologia , Lipoxigenases/metabolismo , Macrófagos/metabolismo , Masculino , Camundongos Endogâmicos C57BL , Infarto do Miocárdio/imunologia , Infarto do Miocárdio/patologia , Infarto do Miocárdio/fisiopatologia , Miocárdio/imunologia , Miocárdio/patologia , Prostaglandina-Endoperóxido Sintases/metabolismo , Recuperação de Função Fisiológica , Fatores Sexuais , Função Ventricular Esquerda , Remodelação Ventricular
15.
Mol Metab ; 31: 138-149, 2020 01.
Artigo em Inglês | MEDLINE | ID: mdl-31918915

RESUMO

OBJECTIVE: Recently, we observed that the specialized proresolving mediator (SPM) entity resolvin D1 activates lipoxin A4/formyl peptide receptor 2 (ALX/FPR2), which facilitates cardiac healing and persistent inflammation is a hallmark of impaired cardiac repair in aging. Splenic leukocyte-directed SPMs are essential for the safe clearance of inflammation and cardiac repair after injury; however, the target of SPMs remains undefined in cardiac healing and repair. METHODS: To define the mechanistic basis of ALX/FPR2 as a resolvin D1 target, ALX/FPR2-null mice were examined extensively. The systolic-diastolic heart function was assessed using echocardiography, leukocytes were phenotyped using flow cytometry, and SPMs were quantitated using mass spectrometry. The presence of cardiorenal syndrome was validated using histology and renal markers. RESULTS: Lack of ALX/FPR2 led to the development of spontaneous obesity and diastolic dysfunction with reduced survival with aging. After cardiac injury, ALX/FPR2-/- mice showed lower expression of lipoxygenases (-5, -12, -15) and a reduction in SPMs in the infarcted left ventricle and spleen, indicating nonresolving inflammation. Reduced SPM levels in the infarcted heart and spleen are suggestive of impaired cross-talk between the injured heart and splenic leukocytes, which are required for the resolution of inflammation. In contrast, cyclooxygenases (-1 and -2) were over amplified in the infarcted heart. Together, these results suggest interorgan signaling in which the spleen acts as both an SPM biosynthesizer and supplier in acute heart failure. ALX/FPR2 dysfunction magnified obesogenic cardiomyopathy and renal inflammation (↑NGAL, ↑TNF-α, ↑CCL2, ↑IL-1ß) with elevated plasma creatinine levels in aging mice. At the cellular level, ALX/FPR2-/- mice showed impairment of macrophage phagocytic function ex-vivo with expansion of neutrophils after myocardial infarction. CONCLUSIONS: Lack of ALX/FPR2 induced obesity, reduced the life span, amplified leukocyte dysfunction, and facilitated profound interorgan nonresolving inflammation. Our study shows the integrative and indispensable role of ALX/FPR2 in lipid metabolism, cardiac inflammation-resolution processes, obesogenic aging, and renal homeostasis.


Assuntos
Insuficiência Cardíaca/metabolismo , Inflamação/metabolismo , Lipoxinas/metabolismo , Receptores de Formil Peptídeo/metabolismo , Receptores de Lipoxinas/metabolismo , Fatores Etários , Animais , Insuficiência Cardíaca/patologia , Humanos , Inflamação/patologia , Lipoxinas/deficiência , Macrófagos/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Receptores de Formil Peptídeo/deficiência , Receptores de Lipoxinas/deficiência
16.
Pharmacol Ther ; 205: 107424, 2020 01.
Artigo em Inglês | MEDLINE | ID: mdl-31629005

RESUMO

Amplified innate leukocytes (neutrophils and monocytes/macrophages) are associated with advanced ischemic and non-ischemic heart failure (HF). Intensified neutrophilic leukocytosis (neutrophilia) and sustained activation of neutrophils is the predominant factor that determines over activated inflammation in acute HF and the outcome of long-term chronic HF. After heart attack, the first wave of innate responsive and short-lived neutrophils is essential for the initiation of inflammation, resolution of inflammation, and cardiac repair, however uncontrolled and long-term activation of neutrophils leads to collateral damage of myocardium. In the presented review, we highlighted the interactive and integrative role of neutrophil phenotypes in cellular and molecular events of ischemic HF. In addition, we discussed the current, nonimmune, immune, and novel paradigms of neutrophils in HF associated with differential factors with a specific interest in non-resolving inflammation and resolution physiology.


Assuntos
Insuficiência Cardíaca/fisiopatologia , Isquemia Miocárdica/fisiopatologia , Neutrófilos/metabolismo , Doença Aguda , Animais , Doença Crônica , Insuficiência Cardíaca/imunologia , Humanos , Inflamação/imunologia , Inflamação/fisiopatologia , Isquemia Miocárdica/imunologia , Neutrófilos/imunologia , Fenótipo
17.
Circ Res ; 125(11): 957-968, 2019 11 08.
Artigo em Inglês | MEDLINE | ID: mdl-31588864

RESUMO

RATIONALE: Lower NP (natriuretic peptide) levels may contribute to the development of cardiometabolic diseases. Blacks have lower NP levels than middle-aged and older white adults. A high-carbohydrate challenge causes an upregulation of a negative ANP regulator microRNA-425 (miR-425), which reduces ANP (atrial-NP) levels in whites. OBJECTIVES: We designed a prospective trial to study racial differences in (1) NP levels among young adults, (2) NP response to a high-carbohydrate challenge, and (3) explore underlying mechanisms for race-based differences. METHODS AND RESULTS: Healthy self-identified blacks and whites received 3 days of study diet followed by a high-carbohydrate challenge. Gene expression from whole blood RNA was assessed in the trial participants. Additionally, atrial and ventricular tissue samples from the Myocardial Applied Genomics Network repository were examined for NP system gene expression. Among 72 healthy participants, we found that B-type-NP, NT-proBNP (N-terminal-pro-B-type NP), and MRproANP (midregional-pro-ANP) levels were 30%, 47%, and 18% lower in blacks compared with whites (P≤0.01), respectively. The decrease in MRproANP levels in response to a high-carbohydrate challenge differed by race (blacks 23% [95% CI, 19%-27%] versus whites 34% [95% CI, 31%-38]; Pinteraction<0.001), with no change in NT-proBNP levels. We did not observe any racial differences in expression of genes encoding for NPs (NPPA/NPPB) or NP signaling (NPR1) in atrial and ventricular tissues. NP processing (corin), clearance (NPR3), and regulation (miR-425) genes were ≈3.5-, ≈2.5-, and ≈2-fold higher in blacks than whites in atrial tissues, respectively. We also found a 2-and 8-fold higher whole blood RNA expression of gene encoding for Neprilysin (MME) and miR-425 among blacks than whites. CONCLUSIONS: Racial differences in NP levels are evident in young, healthy adults suggesting a state of NP deficiency exists in blacks. Impaired NP processing and clearance may contribute to race-based NP differences. Higher miR-425 levels in blacks motivate additional studies to understand differences in NP downregulation after physiological perturbations. CLINICAL TRIAL REGISTRATION: URL: https://clinicaltrials.gov/ct2/show/NCT03072602. Unique identifier: NCT03072602.


Assuntos
Fator Natriurético Atrial/sangue , Negro ou Afro-Americano , Carboidratos da Dieta/administração & dosagem , Disparidades nos Níveis de Saúde , Peptídeo Natriurético Encefálico/sangue , Fragmentos de Peptídeos/sangue , População Branca , Adulto , Alabama , Fator Natriurético Atrial/genética , Biomarcadores/sangue , Linhagem Celular , Carboidratos da Dieta/metabolismo , Regulação para Baixo , Feminino , Voluntários Saudáveis , Humanos , Células-Tronco Pluripotentes Induzidas/metabolismo , Masculino , Miócitos Cardíacos/metabolismo , Peptídeo Natriurético Encefálico/genética , Fragmentos de Peptídeos/genética , Estudos Prospectivos , Fatores Raciais , Fatores de Tempo
19.
Pharmacol Res ; 146: 104295, 2019 08.
Artigo em Inglês | MEDLINE | ID: mdl-31216426

RESUMO

Lifestyle or age-related risk factors over-activate the inflammation that triggers acute heart failure (HF)-related mortality following myocardial infarction (MI). Post-MI activated leukocytes express formyl peptide receptor 2 (FPR2) that is essential for inflammation-resolution and in cardiac healing. However, the role of FPR2 in acute HF is incomplete and remain of interest. Here, we aimed to determine whether pharmacological inhibition of FPR2 perturb leukocyte trafficking in acute HF. Male C57BL/6 (8-12 weeks) mice were subjected to acute HF (MI-d1) using permanent coronary artery ligation that develops irreversible acute and chronic heart failure. FPR2 antagonist WRW4 (1 µg/kg/day) was subcutaneously injected 3 h post-MI maintaining saline-injected MI-controls. Leukocytes were quantitated using flow cytometry, and acute decompensated HF was confirmed using echocardiography and histology. FPR2 inhibition decreased the expression of FPR2 in the LV and spleen tissues. Administration of WRW4 inhibitor to mice primed immature and inactive neutrophils infiltration Ly6Gint and intensified the Ccl2 expression compared to MI-control in the infarcted LV post-MI. Leukocyte profiling revealed an overall decrease in monocytes (23.3 ± 2%) in WRW4-injected mice compared with MI-control (49.1 ± 2%) in infarcted LV. FPR2 inhibition increased F4/80+/Ly6Chi pro-inflammatory macrophages (14.8 ± 2%) compared with MI-control (10 ± 1%) with increased transcripts of pro-inflammatory markers TNF-α and IL-1ß, and decreased Arg-1 expression in the infarcted LV compared to MI-controls is suggestive of the impaired acute inflammatory response. Inhibition of FPR2 using WRW4 also disturbed splenocardiac leukocytes recruitment by priming immature neutrophils leading to the onset of incomplete resolution signaling in acute decompensated HF post-MI.


Assuntos
Insuficiência Cardíaca/metabolismo , Inflamação/metabolismo , Leucócitos/metabolismo , Receptores de Formil Peptídeo/metabolismo , Animais , Biomarcadores/metabolismo , Vasos Coronários/metabolismo , Macrófagos/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Monócitos/metabolismo , Infarto do Miocárdio/metabolismo , Neutrófilos/metabolismo , Baço/metabolismo
20.
Metabolism ; 96: 22-32, 2019 07.
Artigo em Inglês | MEDLINE | ID: mdl-30999004

RESUMO

BACKGROUND: After myocardial infarction (MI), delayed progression or reversal of cardiac remodeling is a prime target to limit advanced chronic heart failure (HF). However, the temporal kinetics of lipidomic and systemic metabolic signaling is unclear in HF. There is no consensus on metabolic and lipidomic signatures that influence structure, function, and survival in HF. Here we use genetic knock out model to delineate lipidomic, and metabolic changes to describe the role of lipoxygenase in advancing ischemic HF driven by leukocyte activation with signs of non-resolving inflammation. Bioactive lipids and metabolites are implicated in acute and chronic HF, and the goal of this study was to define the role of lipoxygenase in temporal kinetics of lipidomic and metabolic reprogramming in HF. MATERIALS AND METHODS: To address this question, we used a permanent coronary ligation mouse model which showed profound metabolic and lipidomic reprogramming in acute HF. Additionally, we defined the lipoxygenase-mediated changes in cardiac pathophysiology in acute and chronic HF. For this, we quantitated systemic metabolic changes and lipidomic profiling in infarcted heart tissue with obvious structural remodeling and cardiac dysfunction progressing from acute to chronic HF in the survival cohort. RESULTS: After MI, lipoxygenase-derived specialized pro-resolving mediators were quantitated and showed lipoxygenase-deficient mice (12/15LOX-/-) biosynthesize epoxyeicosatrienoic acid (EETs; cypoxins) to facilitate cardiac healing. Lipoxygenase-deficient mice reduced diabetes risk biomarker 2-aminoadipic acid with profound alterations of plasma metabolic signaling of hexoses, amino acids, biogenic amines, acylcarnitines, glycerophospholipids, and sphingolipids in acute HF, thereby improved survival. CONCLUSION: Specific lipoxygenase deletion alters lipidomic and metabolic signatures, with modified leukocyte profiling that delayed HF progression and improved survival. Future studies are warranted to define the molecular network of lipidome and metabolome in acute and chronic HF patients.


Assuntos
Insuficiência Cardíaca/metabolismo , Metabolismo dos Lipídeos/fisiologia , Lipidômica , Lipoxigenases/metabolismo , Ácido 2-Aminoadípico/análise , Ácido 2-Aminoadípico/metabolismo , Animais , Biomarcadores , Vasos Coronários , Coração/fisiopatologia , Insuficiência Cardíaca/enzimologia , Insuficiência Cardíaca/genética , Cinética , Leucócitos/metabolismo , Leucócitos/patologia , Ligadura , Metabolismo dos Lipídeos/genética , Lipoxigenases/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Isquemia Miocárdica/enzimologia , Isquemia Miocárdica/genética , Isquemia Miocárdica/metabolismo , Miocárdio/patologia , Análise de Sobrevida
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA