Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 4 de 4
Filter
Add more filters










Database
Language
Publication year range
1.
J Am Heart Assoc ; 9(8): e015672, 2020 04 21.
Article in English | MEDLINE | ID: mdl-32295449

ABSTRACT

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.


Subject(s)
Inflammation Mediators/metabolism , Leukocytes/metabolism , Myocardial Infarction/metabolism , Myocardium/metabolism , Animals , Disease Models, Animal , Eicosapentaenoic Acid/metabolism , Female , Leukocytes/immunology , Lipoxygenases/metabolism , Macrophages/metabolism , Male , Mice, Inbred C57BL , Myocardial Infarction/immunology , Myocardial Infarction/pathology , Myocardial Infarction/physiopathology , Myocardium/immunology , Myocardium/pathology , Prostaglandin-Endoperoxide Synthases/metabolism , Recovery of Function , Sex Factors , Ventricular Function, Left , Ventricular Remodeling
2.
Mol Metab ; 31: 138-149, 2020 01.
Article in English | MEDLINE | ID: mdl-31918915

ABSTRACT

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.


Subject(s)
Heart Failure/metabolism , Inflammation/metabolism , Lipoxins/metabolism , Receptors, Formyl Peptide/metabolism , Receptors, Lipoxin/metabolism , Age Factors , Animals , Heart Failure/pathology , Humans , Inflammation/pathology , Lipoxins/deficiency , Macrophages/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Receptors, Formyl Peptide/deficiency , Receptors, Lipoxin/deficiency
3.
Heart Fail Rev ; 25(2): 381-391, 2020 03.
Article in English | MEDLINE | ID: mdl-31201605

ABSTRACT

Cardiac injuries, like heart attacks, drive the secondary pathology with advanced heart failure. In this process, non-resolving inflammation is a prime component of accelerated cardiovascular disease and subsequent fatal events associated with imbalanced diet, physical inactivity, disrupted circadian rhythms, neuro-hormonal stress, and poly- or co-medication. Laboratory rodents have established that splenic leukocyte-directed resolution mechanisms are essential for cardiac repair after injury. Here, we discuss the impact of three lifestyle-related factors that are prime causes of derailed cardiac healing, putative non-resolving inflammation-resolution mechanisms in cardiovascular diseases, and progressive heart failure after cardiac injury. The presented review resurfaces the lifestyle-related risks and future research directions required to understand the molecular and cellular mechanisms between the causes of cardiovascular disease and their related consequences of non-resolving inflammation.


Subject(s)
Cardiovascular Diseases/etiology , Inflammation/complications , Leukocytes/pathology , Animals , Cardiovascular Diseases/metabolism , Chronic Disease , Humans , Inflammation/metabolism
4.
Life Sci ; 218: 224-232, 2019 Feb 01.
Article in English | MEDLINE | ID: mdl-30597172

ABSTRACT

Non-Steroidal Anti-Inflammatory Drugs (NSAIDs) are commonly used to control pain, inflammation, and limit the cardinal signs of injury in humans. However, prolonged use of NSAIDs increases the risk of heart attack (myocardial infarction; MI) and the subsequent risk of heart and renal failure. The molecular and cellular mechanism of action for this adverse effect, particularly along the cardiorenal network, is incomplete. To define the mechanism, carprofen (CAP), an NSAID was administered at the dose of 5 mg/kg to C57BL/6 male mice for two weeks. After last dose of CAP treatment mice were subjected to permanent occlusion of coronary artery that induces irreversible cardiac remodeling while maintaining naive and MI-controls. After MI, cardiac pathology and dysfunction were confirmed, along with additional measurements of kidney function, histology, and injury markers, such as plasma creatinine. CAP treatment increased plasma creatinine levels and subsequently, myocardial structural disorganization increased. Kidney neutrophil gelatinase associated lipocalin (NGAL) and protein expression were increased post-MI. After two weeks CAP treatment, the expression of pyrogenic pro-inflammatory cytokines TNF-α and IL-1ß was increased compared to non-CAP treated mice, indicative of amplified inflammatory response. There was also evidence that renal injury of both the post-CAP treatment controls and post-CAP MI were much greater than the non-CAP treated naïve controls, as serum creatinine and NGAL levels were elevated along with obvious structural impairment of the glomerulus. Therefore, CAP treatment tampers with the acute inflammatory response that promotes cardiorenal syndrome and non-resolving inflammation post-MI in acute heart failure.


Subject(s)
Carbazoles/toxicity , Cardio-Renal Syndrome/pathology , Heart Failure/physiopathology , Inflammation/pathology , Myocardial Infarction/pathology , Animals , Anti-Inflammatory Agents, Non-Steroidal/toxicity , Biomarkers/metabolism , Cardio-Renal Syndrome/chemically induced , Cardio-Renal Syndrome/metabolism , Cytokines/metabolism , Disease Models, Animal , Female , Inflammation/chemically induced , Inflammation/metabolism , Male , Mice , Mice, Inbred C57BL , Myocardial Infarction/chemically induced , Myocardial Infarction/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...