1
|
Zhao H, Guo M, Yang C, Xing F. The relationship between serum Omega-6 fatty acids and cardiovascular disease mortality: A competing risks and multivariate Mendelian randomization analysis. Clin Nutr ESPEN 2025; 66:372-380. [PMID: 39894349 DOI: 10.1016/j.clnesp.2025.01.053] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2024] [Revised: 12/15/2024] [Accepted: 01/23/2025] [Indexed: 02/04/2025]
Abstract
BACKGROUND & AIMS The impact of serum Omega-6 fatty acids on cardiovascular health is debated, with evidence supporting both protective and harmful effects. To investigate the association between serum Omega-6 fatty acid and mortality from all causes and cardiovascular disease (CVD), utilizing advanced statistical methodologies including competing risk models and multivariate Mendelian randomization. METHODS Data of 5070 participants from National Health and Nutrition Examination Survey (NHANES) in 2011-2014 wave were analyzed, with follow-up data on mortality sourced from the National Death Index. Serum Omega-6 fatty acids level was measured at baseline. Cox proportional hazards regression and competing risks models was used to estimate hazard ratios (HRs) and 95 % confidence intervals (CIs) for all-cause and CVD mortality by baseline Omega-6 fatty acids level. Restricted cubic splines were used to explore the nonlinearity. Mendelian randomization to assess the causal relationships between Omega-6 levels and mortality. RESULTS 438 all-cause deaths and 137 CVD deaths were observed during an 83 months median follow-up. Restricted cubic spline analysis demonstrated a U-shaped correlation between baseline serum Omega-6 fatty acid levels with all-cause and CVD mortality risks. Subgroup analysis indicated that for the low-level Omega-6 fatty acid participants, the hazard ratios were 0.68 (95 % CI, 0.55-0.85) for all-cause mortality and 0.62 (95 % CI, 0.40-0.95) for CVD mortality. Conversely, for the high-level participants, the hazard ratios were 1.14 (95 % CI, 1.01-1.28) for all-cause mortality and 1.23 (95 % CI, 1.05-1.44) for CVD mortality. Both univariate and multivariate Mendelian randomization analyses confirmed a positive causal relationship between higher serum Omega-6 fatty acid levels and increased CVD mortality risk. CONCLUSIONS The findings suggest a U-shaped relationship between serum Omega-6 fatty acid levels and mortality risks, with elevated levels linked causally to increased CVD mortality. These results underscore the need for balanced dietary Omega-6 fatty acid intake to optimize cardiovascular health.
Collapse
Affiliation(s)
- Huimin Zhao
- Department of General Medicine, No. 970 Hospital of the People's Liberation Army Joint Logistics Support Force, Yantai, Shandong, China
| | - Meirong Guo
- Department of General Medicine, No. 970 Hospital of the People's Liberation Army Joint Logistics Support Force, Yantai, Shandong, China
| | - Changlin Yang
- Department of General Medicine, No. 970 Hospital of the People's Liberation Army Joint Logistics Support Force, Yantai, Shandong, China
| | - Fangkai Xing
- Department of General Medicine, No. 970 Hospital of the People's Liberation Army Joint Logistics Support Force, Yantai, Shandong, China.
| |
Collapse
|
2
|
Shan F, Xiong Y, Pai P, Liu M. Systemic immune inflammation mediates the association of serum omega-3 and omega-6 polyunsaturated fatty acids with biological aging: a national population-based study. Aging Clin Exp Res 2025; 37:74. [PMID: 40057623 PMCID: PMC11890405 DOI: 10.1007/s40520-025-02964-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2024] [Accepted: 02/08/2025] [Indexed: 05/13/2025]
Abstract
OBJECTIVE This study aimed to explore the association between serum omega-3 (n-3) and omega-6 (n-6) polyunsaturated fatty acids (PUFAs) and biological aging, along with the potential mediating role of systemic immune inflammation (SII). METHODS Data from the National Health and Nutrition Examination Survey (NHANES) 2011-2014 were used for analyses. Accelerated aging in participants was assessed by calculating the difference between phenotypic age (PhenoAge) and chronological age. Weighted multivariate linear regression models and subgroup analysis were used to investigate the correlation between serum n-3 and n-6 PUFAs and accelerated aging, and restricted cubic spline (RCS) model was applied to explore potential nonlinear relationships. We further conducted mediation analyses to assess the role of SII in these relationships. Additionally, weighted quantile sum (WQS) regression and quantile g-computation (QGC) models were conducted to investigate the mixed effects of serum PUFAs and identify the key contributor. RESULTS A total of 3376 participants were enrolled in this study. In multivariate linear regression models, eight of the twelve individual serum PUFAs showed a significantly negative association with PhenoAge acceleration, Specifically, per-unit increases in linoleic acid (LA), gamma-linolenic acid (GLA), arachidonic acid (AA), alpha-linolenic acid (ALA), stearidonic acid (SDA), eicosapentaenoic acid (EPA), docosapentaenoic acid (n-3 DPA), and docosahexaenoic acid (DHA) were all associated with reduced PhenoAge acceleration (P < 0.05, respectively). Subgroup analysis demonstrated robust consistence results when stratified by age, sex, and race/ethnicity. L-shaped nonlinear relationships were observed between PhenoAge acceleration with total n-6 PUFAs, LA and ALA (all P for nonlinear < 0.05). Mediation analyses indicated that SII mediated the relationship between serum PUFAs and reduced PhenoAge acceleration. Mixed-effects analysis using WQS and QGC models revealed that the combined effect of serum PUFAs on reducing PhenoAge acceleration, with DHA showing the strongest significant contribution. CONCLUSIONS This study demonstrated that higher levels of certain PUFAs were associated with a reduction in PhenoAge acceleration either individually or in combination, with DHA having the most prominent effect in mixed effects. The SII mediated these relationships, suggesting that PUFAs may slow biological aging by reducing inflammation. These findings highlighted the potential role of PUFAs in mitigating accelerated aging and their implications for aging-related health interventions.
Collapse
Affiliation(s)
- Fei Shan
- Department of Cardiology, University of Hong Kong-Shenzhen Hospital, No.1, Haiyuan 1st Road, Futian District, Shenzhen, Guangdong, China
| | - Yu Xiong
- Department of Neurology, University of Hong Kong-Shenzhen Hospital, No.1, Haiyuan 1st Road, Futian District, Shenzhen, Guangdong, China
| | - Pearl Pai
- Department of Medicine, University of Hong Kong-Shenzhen Hospital, No.1, Haiyuan 1st Road, Futian District, Shenzhen, Guangdong, China.
| | - Mingya Liu
- Department of Cardiology, University of Hong Kong-Shenzhen Hospital, No.1, Haiyuan 1st Road, Futian District, Shenzhen, Guangdong, China.
| |
Collapse
|
3
|
Pan W, Zhang C, Du X, Su X, Lin J, Jiang T, Chen W. Association between epigenetic aging and atrioventricular block: a two-sample Mendelian randomization study. Epigenomics 2025; 17:223-234. [PMID: 39829373 PMCID: PMC11853617 DOI: 10.1080/17501911.2025.2454894] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2024] [Accepted: 01/14/2025] [Indexed: 01/22/2025] Open
Abstract
AIMS Atrioventricular block (AVB) is a prevalent bradyarrhythmia. This study aims to investigate the causal effects of epigenetic aging, as inferred from DNA methylation profiles on the prevalence of AVB by Mendelian randomization (MR) analysis. METHODS Genetic instruments for epigenetic aging and AVB were obtained from genome-wide association study data in the Edinburgh DataShare and FinnGen biobanks. Univariable and multivariable MR analyses were conducted to evaluate causal associations. Additionally, we employed sensitivity tests to assess the robustness of the MR findings. RESULTS MR analysis showed that genetically predicted GrimAge acceleration was significantly associated with a higher risk of AVB (inverse variance-weighted: p = 0.010, 95% confidence interval (CI) = 1.024-1.196; weighted median: p = 0.031, 95% CI = 1.009-1.215). However, no evidence supported a causal relationship between AVB and epigenetic aging. The association between epigenetic aging and AVB was established using multivariate MR analysis after adjusting for various risk factors. Sensitivity analyses confirmed the reliability and robustness of the results. CONCLUSION Our findings suggest that epigenetic aging in GrimAge may increase the risk of AVB, emphasizing the importance of addressing epigenetic aging in strategies for AVB prevention.
Collapse
Affiliation(s)
- Wanqian Pan
- Department of Cardiology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, P. R. China
| | - Chi Zhang
- Department of Cardiology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, P. R. China
| | - Xiaojiao Du
- Department of Cardiology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, P. R. China
| | - Xiong Su
- Department of Biochemistry and Molecular Biology, Suzhou Medical College of Soochow University, Suzhou, Jiangsu, P. R. China
| | - Jia Lin
- Department of Cardiology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, P. R. China
| | - Tingbo Jiang
- Department of Cardiology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, P. R. China
| | - Weixiang Chen
- Department of Cardiology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, P. R. China
| |
Collapse
|
4
|
Mao J, Sun H, Shen Q, Zou C, Yang Y, Du Q. Impact of pre-pregnancy body mass index on preeclampsia. Front Med (Lausanne) 2025; 12:1529966. [PMID: 39975682 PMCID: PMC11835700 DOI: 10.3389/fmed.2025.1529966] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2024] [Accepted: 01/20/2025] [Indexed: 02/21/2025] Open
Abstract
Background It remains unclear how pre-pregnancy body mass index (BMI) affects preeclampsia in the Chinese population, primarily due to insufficient large-scale research on this topic. Objective The study aimed to determine the relationship between pre-pregnancy BMI and (severe) preeclampsia in the Chinese population, providing a detailed description of the findings. Methods The retrospective study included a total of 75,773 pregnant women registered between 2016 and 2020. These participants were categorized into four groups based on their pre-pregnancy BMI: underweight (BMI < 18.5 kg/m2), normal weight (BMI 18.5-<24 kg/m2), overweight (BMI 24-<28 kg/m2), and obese (BMI ≥ 28 kg/m2). The relationship between risks of preeclampsia or severe preeclampsia and pre-pregnancy BMI were further explored, with an evaluation of potential modification by maternal age. Results A lower risk of developing preeclampsia was observed in the underweight population, with an OR of 0.604 (95%CI, 0.507-0.719). In contrast, women who were overweight or obese during the pre-pregnancy period demonstrated a significantly higher risk of preeclampsia, with ORs of 2.211 (95%CI, 1.967-2.486) and 3.662 (95%CI, 3.026-4.431), respectively. After adjusting for confounding factors, the elevated risk of preeclampsia persisted, showing ORs of 2.152(95%CI, 1.911-2.425) for the overweight population and 3.493 (95%CI, 2.874-4.245) for those who were obese, while the risk for underweight women remained lower, with an OR of 0.609(95%CI, 0.511-0.727). For severe preeclampsia, the risk was also higher in the overweight and obese participants after adjusting for confounders, demonstrating ORs of 1.652(95%CI, 1.364-2.001) and 2.762(95%CI, 2.014-3.788), respectively. The underweight population exhibited a lower risk of severe preeclampsia, with an OR of 0.720(95%CI, 0.565-0.919). In addition, these risks were not significantly associated with maternal age. Conclusion Regardless of adjustment for confounders, underweight women demonstrated a lower risk of preeclampsia, whereas the overweight/obese population exhibited a higher occurrence of both preeclampsia and severe preeclampsia. These associations were not influenced by maternal age.
Collapse
Affiliation(s)
| | | | | | | | | | - Qiaoling Du
- Department of Obstetrics, Shanghai Key Laboratory of Maternal Fetal Medicine, Shanghai Institute of Maternal-Fetal Medicine and Gynecologic Oncology, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, Shanghai, China
| |
Collapse
|
5
|
Capece U, Gugliandolo S, Morciano C, Avolio A, Splendore A, Di Giuseppe G, Ciccarelli G, Soldovieri L, Brunetti M, Mezza T, Pontecorvi A, Giaccari A, Cinti F. Erythrocyte Membrane Fluidity and Omega-3 Fatty Acid Intake: Current Outlook and Perspectives for a Novel, Nutritionally Modifiable Cardiovascular Risk Factor. Nutrients 2024; 16:4318. [PMID: 39770939 PMCID: PMC11676811 DOI: 10.3390/nu16244318] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2024] [Revised: 12/07/2024] [Accepted: 12/12/2024] [Indexed: 01/11/2025] Open
Abstract
Omega-3 fatty acids reduce triglycerides and have several positive effects on different organs and systems. They are also found in the plasma membrane in variable amounts in relation to genetics and diet. However, it is still unclear whether omega-3 supplementation can reduce the occurrence of major cardiovascular events (MACEs). Two trials, REDUCE-IT (Reduction of Cardiovascular Events with Icosapent Ethyl-Intervention Trial), with highly purified EPA, and STRENGTH (Effect of High-Dose Omega-3 Fatty Acids vs. Corn Oil on Major Adverse Cardiovascular Events in Patients at High Cardiovascular Risk), with a combination of EPA and DHA, have produced different outcomes, triggering a scientific debate on possible explanations for the discrepancies. Furthermore, doubts have arisen as to the anti-inflammatory and anti-aggregating activity of these compounds. Recent studies have, however, highlighted interesting effects of EPA and DHA on erythrocyte membrane fluidity (EMF). EMF is governed by a complex and dynamic biochemical framework, with fatty acids playing a central role. Furthermore, it can be easily measured in erythrocytes from a blood sample using fluorescent probes. Recent research has also shown that EMF could act as a possible cardiovascular risk factor biomarker. This review aims to synthetize the latest evidence on erythrocyte membrane fluidity, exploring its potential role as a biomarker of residual cardiovascular risk and discussing its clinical relevance. Further, we aim to dissect the possible biological mechanisms that link omega-3 modifiable membrane fluidity to cardiovascular health.
Collapse
Affiliation(s)
- Umberto Capece
- Endocrinologia e Diabetologia, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00136 Rome, Italy
- Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
| | - Shawn Gugliandolo
- Endocrinologia e Diabetologia, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00136 Rome, Italy
- Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
| | - Cassandra Morciano
- Endocrinologia e Diabetologia, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00136 Rome, Italy
- Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
| | - Adriana Avolio
- Endocrinologia e Diabetologia, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00136 Rome, Italy
- Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
| | - Amelia Splendore
- Endocrinologia e Diabetologia, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00136 Rome, Italy
- Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
| | - Gianfranco Di Giuseppe
- Endocrinologia e Diabetologia, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00136 Rome, Italy
- Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
| | - Gea Ciccarelli
- Endocrinologia e Diabetologia, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00136 Rome, Italy
- Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
| | - Laura Soldovieri
- Endocrinologia e Diabetologia, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00136 Rome, Italy
- Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
| | - Michela Brunetti
- Endocrinologia e Diabetologia, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00136 Rome, Italy
- Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
| | - Teresa Mezza
- Endocrinologia e Diabetologia, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00136 Rome, Italy
- Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
| | - Alfredo Pontecorvi
- Endocrinologia e Diabetologia, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00136 Rome, Italy
- Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
| | - Andrea Giaccari
- Endocrinologia e Diabetologia, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00136 Rome, Italy
- Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
| | - Francesca Cinti
- Endocrinologia e Diabetologia, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00136 Rome, Italy
- Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
| |
Collapse
|
6
|
Pergande MR, Osterbauer KJ, Buck KM, Roberts DS, Wood NN, Balasubramanian P, Mann MW, Rossler KJ, Diffee GM, Colman RJ, Anderson RM, Ge Y. Mass Spectrometry-Based Multiomics Identifies Metabolic Signatures of Sarcopenia in Rhesus Monkey Skeletal Muscle. J Proteome Res 2024; 23:2845-2856. [PMID: 37991985 PMCID: PMC11109024 DOI: 10.1021/acs.jproteome.3c00474] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2023]
Abstract
Sarcopenia is a progressive disorder characterized by age-related loss of skeletal muscle mass and function. Although significant progress has been made over the years to identify the molecular determinants of sarcopenia, the precise mechanisms underlying the age-related loss of contractile function remains unclear. Advances in "omics" technologies, including mass spectrometry-based proteomic and metabolomic analyses, offer great opportunities to better understand sarcopenia. Herein, we performed mass spectrometry-based analyses of the vastus lateralis from young, middle-aged, and older rhesus monkeys to identify molecular signatures of sarcopenia. In our proteomic analysis, we identified proteins that change with age, including those involved in adenosine triphosphate and adenosine monophosphate metabolism as well as fatty acid beta oxidation. In our untargeted metabolomic analysis, we identified metabolites that changed with age largely related to energy metabolism including fatty acid beta oxidation. Pathway analysis of age-responsive proteins and metabolites revealed changes in muscle structure and contraction as well as lipid, carbohydrate, and purine metabolism. Together, this study discovers new metabolic signatures and offers new insights into the molecular mechanisms underlying sarcopenia for the evaluation and monitoring of a therapeutic treatment of sarcopenia.
Collapse
Affiliation(s)
- Melissa R. Pergande
- Department of Cell and Regenerative Biology, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Katie J. Osterbauer
- Department of Medicine, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Kevin M. Buck
- Department of Chemistry, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - David S. Roberts
- Department of Chemistry, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Nina N. Wood
- Department of Medicine, University of Wisconsin-Madison, Madison, WI 53705, USA
| | | | - Morgan W. Mann
- Department of Medicine, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Kalina J. Rossler
- Department of Cell and Regenerative Biology, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Gary M. Diffee
- Department of Kinesiology, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Ricki J. Colman
- Department of Cell and Regenerative Biology, University of Wisconsin-Madison, Madison, WI 53705, USA
- Wisconsin National Primate Research Center, University of Wisconsin-Madison, Madison, WI 53715, USA
| | - Rozalyn M. Anderson
- Department of Medicine, University of Wisconsin-Madison, Madison, WI 53705, USA
- Geriatric Research Education and Clinical Center, William S. Middleton Memorial Veterans Hospital, Madison, WI 53705, USA
| | - Ying Ge
- Department of Cell and Regenerative Biology, University of Wisconsin-Madison, Madison, WI 53705, USA
- Department of Medicine, University of Wisconsin-Madison, Madison, WI 53705, USA
- Human Proteomics Program, University of Wisconsin-Madison, Madison, WI 53705, USA
| |
Collapse
|
7
|
Hawley AL, Baum JI. Nutrition as the foundation for successful aging: a focus on dietary protein and omega-3 polyunsaturated fatty acids. Nutr Rev 2024; 82:389-406. [PMID: 37319363 DOI: 10.1093/nutrit/nuad061] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/17/2023] Open
Abstract
Skeletal muscle plays a critical role throughout the aging process. People living with sarcopenia, a progressive and generalized loss of skeletal muscle mass and function, often experience diminished quality of life, which can be attributed to a long period of decline and disability. Therefore, it is important to identify modifiable factors that preserve skeletal muscle and promote successful aging (SA). In this review, SA was defined as (1) low cardiometabolic risk, (2) preservation of physical function, and (3) positive state of wellbeing, with nutrition as an integral component. Several studies identify nutrition, specifically high-quality protein (eg, containing all essential amino acids), and long-chain omega-3 polyunsaturated fatty acids (n-3 PUFAs), eicosapentaenoic acid (EPA), and docosahexaenoic acid (DHA), as positive regulators of SA. Recently, an additive anabolic effect of protein and n-3 PUFAs has been identified in skeletal muscle of older adults. Evidence further suggests that the additive effect of protein and n-3 PUFAs may project beyond skeletal muscle anabolism and promote SA. The key mechanism(s) behind the enhanced effects of intake of protein and n-3 PUFAs needs to be defined. The first objective of this review is to evaluate skeletal muscle as a driver of cardiometabolic health, physical function, and wellbeing to promote SA. The second objective is to examine observational and interventional evidence of protein and n-3 PUFAs on skeletal muscle to promote SA. The final objective is to propose mechanisms by which combined optimal intake of high-quality protein and n-3 PUFAs likely play a key role in SA. Current evidence suggests that increased intake of protein above the Recommended Dietary Allowance and n-3 PUFAs above the Dietary Guidelines for Americans recommendations for late middle-aged and older adults is required to maintain skeletal muscle mass and to promote SA, potentially through the mechanistical target of rapamycin complex 1 (mTORC1).
Collapse
Affiliation(s)
- Aubree L Hawley
- School of Human and Environmental Sciences, University of Arkansas, Fayetteville, AR, USA
| | - Jamie I Baum
- Center for Human Nutrition, Department of Food Science, University of Arkansas System Division of Agriculture, Fayetteville, AR, USA
| |
Collapse
|
8
|
Li J, Xin Y, Wang Z, Li J, Li W, Li H. The role of cardiac resident macrophage in cardiac aging. Aging Cell 2023; 22:e14008. [PMID: 37817547 PMCID: PMC10726886 DOI: 10.1111/acel.14008] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2023] [Revised: 09/20/2023] [Accepted: 09/22/2023] [Indexed: 10/12/2023] Open
Abstract
Advancements in longevity research have provided insights into the impact of cardiac aging on the structural and functional aspects of the heart. Notable changes include the gradual remodeling of the myocardium, the occurrence of left ventricular hypertrophy, and the decline in both systolic and diastolic functions. Macrophages, a type of immune cell, play a pivotal role in innate immunity by serving as vigilant agents against pathogens, facilitating wound healing, and orchestrating the development of targeted acquired immune responses. Distinct subsets of macrophages are present within the cardiac tissue and demonstrate varied functions in response to myocardial injury. The differentiation of cardiac macrophages according to their developmental origin has proven to be a valuable strategy in identifying reparative macrophage populations, which originate from embryonic cells and reside within the tissue, as well as inflammatory macrophages, which are derived from monocytes and recruited to the heart. These subsets of macrophages possess unique characteristics and perform distinct functions. This review aims to summarize the current understanding of the roles and phenotypes of cardiac macrophages in various conditions, including the steady state, aging, and other pathological conditions. Additionally, it will highlight areas that require further investigation to expand our knowledge in this field.
Collapse
Affiliation(s)
- Jiayu Li
- Department of Cardiology, Cardiovascular Center, Beijing Friendship HospitalCapital Medical UniversityBeijingChina
- Laboratory for Clinical MedicineBeijing Friendship Hospital, Capital Medical UniversityBeijingChina
| | - Yanguo Xin
- Department of Cardiology, Cardiovascular Center, Beijing Friendship HospitalCapital Medical UniversityBeijingChina
- Laboratory for Clinical MedicineBeijing Friendship Hospital, Capital Medical UniversityBeijingChina
| | - Zhaojia Wang
- Department of Cardiology, Cardiovascular Center, Beijing Friendship HospitalCapital Medical UniversityBeijingChina
- Laboratory for Clinical MedicineBeijing Friendship Hospital, Capital Medical UniversityBeijingChina
| | - Jingye Li
- Department of Cardiology, Cardiovascular Center, Beijing Friendship HospitalCapital Medical UniversityBeijingChina
| | - Weiping Li
- Department of Cardiology, Cardiovascular Center, Beijing Friendship HospitalCapital Medical UniversityBeijingChina
- Laboratory for Clinical MedicineBeijing Friendship Hospital, Capital Medical UniversityBeijingChina
| | - Hongwei Li
- Department of Cardiology, Cardiovascular Center, Beijing Friendship HospitalCapital Medical UniversityBeijingChina
- Laboratory for Clinical MedicineBeijing Friendship Hospital, Capital Medical UniversityBeijingChina
- Beijing Key Laboratory of Metabolic Disorder Related Cardiovascular DiseaseBeijingChina
| |
Collapse
|
9
|
Lopes KLS, Figueiredo N, Kattah FM, Lima GC, Oliveira ES, Horst MA, Oyama LM, Dâmaso AR, Whitton RGM, de Souza Abreu V, Duarte ACS, Pimentel GD, Corgosinho FC. The degree of food processing can influence serum fatty acid and lipid profiles in women with severe obesity. Front Nutr 2023; 10:1046710. [PMID: 37786791 PMCID: PMC10541961 DOI: 10.3389/fnut.2023.1046710] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2022] [Accepted: 08/18/2023] [Indexed: 10/04/2023] Open
Abstract
Background The increase in the prevalence of obesity is associated with the increase in the consumption of ultra-processed foods and may be related to the increase in the disorders involving metabolism and the transport and storage of fatty acids. Objective To evaluate the effect of processed food consumption according to the degree of processing on the serum fatty acid levels and lipid profile of women with severe obesity. Methods This was a cross-sectional study. Data were collected from anthropometric assessments, the food frequency questionnaire (FFQ), and blood tests for lipidogram studies and serum fatty acid measurements. The foods consumed were identified through the FFQ and classified according to the degree of processing based on the NOVA rating, and the frequencies of consumption were transformed into scores, as proposed by Fornés methodology. Data were analyzed using IBM SPSS Statistics, version 21. The significance level for the analysis was set at 5%. Results This study included 44 women with a mean age of 40.59 years and mean body mass index of 48.61 kg/m2. An inverse association was observed between the consumption of unprocessed and the occurrence of hypertriglyceridemia (p = 0.021) and levels of triglycerides (p = 0.047), total cholesterol (p = 0.030), and very low-density lipoprotein-cholesterol (p = 0.039). The consumption of processed foods was positively associated with the presence of hypertriglyceridemia (p = 0.044) and omega 6/3 ratio (p = 0.001) and negatively associated with total omega 3 levels (p = 0.011). The consumption of processed foods was positively associated with total cholesterol (p = 0.041) and negatively associated with the omega 3/6 ratio (p = 0.001). A negative correlation was found between the average consumption of ultra-processed foods (at least once a week) and serum level of high-density lipoprotein (p = 0.035). Conclusion The consumption of processed and ultra-processed foods was associated with unfavorable lipid profiles and fatty acid levels in women with severe obesity. These results emphasize the importance of promoting the consumption of unprocessed food to mitigate metabolic disorders linked to processed food intake.
Collapse
Affiliation(s)
- Karem Lays Soares Lopes
- Post-Graduation Program in Nutrition and Health, Faculty of Nutrition, Federal University of Goiás – PPGNUT-FANUT-UFG, Goiânia, Goiás, Brazil
| | - Nayra Figueiredo
- Post-Graduation Program in Health Sciences, Faculty of Medicine, Federal University of Goiás, Goiânia, Goiás, Brazil
| | - Fabiana Martins Kattah
- Post-Graduation Program in Nutrition and Health, Faculty of Nutrition, Federal University of Goiás – PPGNUT-FANUT-UFG, Goiânia, Goiás, Brazil
| | - Glaucia Carielo Lima
- Post-Graduation Program in Nutrition and Health, Faculty of Nutrition, Federal University of Goiás – PPGNUT-FANUT-UFG, Goiânia, Goiás, Brazil
| | - Emilly Santos Oliveira
- Post-Graduation Program in Nutrition and Health, Faculty of Nutrition, Federal University of Goiás – PPGNUT-FANUT-UFG, Goiânia, Goiás, Brazil
| | - Maria Aderuza Horst
- Post-Graduation Program in Nutrition and Health, Faculty of Nutrition, Federal University of Goiás – PPGNUT-FANUT-UFG, Goiânia, Goiás, Brazil
| | - Lila Missae Oyama
- Nutrition Physiology Laboratory, Federal University of São Paulo, São Paulo, Brazil
| | - Ana Raimunda Dâmaso
- Post-Graduation Program in Nutrition, Paulista Medicine School, Federal University of São Paulo, São Paulo, Brazil
| | | | | | - Amélia Cristina Stival Duarte
- Post-Graduation Program in Nutrition and Health, Faculty of Nutrition, Federal University of Goiás – PPGNUT-FANUT-UFG, Goiânia, Goiás, Brazil
| | - Gustavo Duarte Pimentel
- Post-Graduation Program in Nutrition and Health, Faculty of Nutrition, Federal University of Goiás – PPGNUT-FANUT-UFG, Goiânia, Goiás, Brazil
- Post-Graduation Program in Health Sciences, Faculty of Medicine, Federal University of Goiás, Goiânia, Goiás, Brazil
| | - Flávia Campos Corgosinho
- Post-Graduation Program in Nutrition and Health, Faculty of Nutrition, Federal University of Goiás – PPGNUT-FANUT-UFG, Goiânia, Goiás, Brazil
- Post-Graduation Program in Health Sciences, Faculty of Medicine, Federal University of Goiás, Goiânia, Goiás, Brazil
| |
Collapse
|
10
|
Rao A, Gupta A, Kain V, Halade GV. Extrinsic and intrinsic modulators of inflammation-resolution signaling in heart failure. Am J Physiol Heart Circ Physiol 2023; 325:H433-H448. [PMID: 37417877 PMCID: PMC10538986 DOI: 10.1152/ajpheart.00276.2023] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/11/2023] [Revised: 06/27/2023] [Accepted: 07/03/2023] [Indexed: 07/08/2023]
Abstract
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.
Collapse
Affiliation(s)
- Archana Rao
- Division of Cardiovascular Sciences, Department of Internal Medicine, Heart Institute, University of South Florida, Tampa, Florida, United States
| | - Akul Gupta
- Division of Cardiovascular Sciences, Department of Internal Medicine, Heart Institute, University of South Florida, Tampa, Florida, United States
| | - Vasundhara Kain
- Division of Cardiovascular Sciences, Department of Internal Medicine, Heart Institute, University of South Florida, Tampa, Florida, United States
| | - Ganesh V Halade
- Division of Cardiovascular Sciences, Department of Internal Medicine, Heart Institute, University of South Florida, Tampa, Florida, United States
| |
Collapse
|
11
|
Mercola J, D'Adamo CR. Linoleic Acid: A Narrative Review of the Effects of Increased Intake in the Standard American Diet and Associations with Chronic Disease. Nutrients 2023; 15:3129. [PMID: 37513547 PMCID: PMC10386285 DOI: 10.3390/nu15143129] [Citation(s) in RCA: 40] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2023] [Revised: 06/20/2023] [Accepted: 06/25/2023] [Indexed: 07/30/2023] Open
Abstract
The intake of linoleic acid (LA) has increased dramatically in the standard American diet. LA is generally promoted as supporting human health, but there exists controversy regarding whether the amount of LA currently consumed in the standard American diet supports human health. The goal of this narrative review is to explore the mechanisms that underlie the hypothesis that excessive LA intake may harm human health. While LA is considered to be an essential fatty acid and support health when consumed in modest amounts, an excessive intake of LA leads to the formation of oxidized linoleic acid metabolites (OXLAMs), impairments in mitochondrial function through suboptimal cardiolipin composition, and likely contributes to many chronic diseases that became an epidemic in the 20th century, and whose prevalence continues to increase. The standard American diet comprises 14 to 25 times more omega-6 fatty acids than omega-3 fatty acids, with the majority of omega-6 intake coming from LA. As LA consumption increases, the potential for OXLAM formation also increases. OXLAMs have been associated with various illnesses, including cardiovascular disease, cancer, and Alzheimer's disease, among others. Lowering dietary LA intake can help reduce the production and accumulation of OXLAMs implicated in chronic diseases. While there are other problematic components in the standard American diet, the half-life of LA is approximately two years, which means the damage can be far more persistent than other dietary factors, and the impact of reducing excessive LA intake takes time. Therefore, additional research-evaluating approaches to reduce OXLAM formation and cardiolipin derangements following LA consumption are warranted.
Collapse
Affiliation(s)
- Joseph Mercola
- Natural Health Partners, LLC, 125 SW 3rd Place, Cape Coral, FL 33991, USA
| | - Christopher R D'Adamo
- Department of Family and Community Medicine, Center for Integrative Medicine, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| |
Collapse
|
12
|
Nair D, Zarei M, Halami P, Talahalli R. Lactobacillus fermentum MCC2760 abrogate high-fat induced perturbations in the enterohepatic circulation of bile acids in rats. Life Sci 2023; 320:121563. [PMID: 36907323 DOI: 10.1016/j.lfs.2023.121563] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2022] [Revised: 02/24/2023] [Accepted: 03/06/2023] [Indexed: 03/12/2023]
Abstract
AIM This study in hyperlipidemic rats elucidated the effect of Lactobacillus fermentum MCC2760 on intestinal bile acid (BA) uptake, hepatic BA synthesis, and enterohepatic BA transporters. MAIN METHODS Diets rich in saturated fatty acids [coconut oil (CO)] and omega-6 fatty acids [sunflower oil (SFO)] at 25 g fat/100 g diet were fed to rats with or without MCC2760 (109 cells/kg body weight). After 60 days of feeding, intestinal BA uptake and expression of Asbt, Osta/b mRNA and protein, and hepatic expression of Ntcp, Bsep, Cyp7a1, Fxr, Shp, Lrh-1, and Hnf4a mRNA were measured. Hepatic expression of HMG-CoA reductase protein and its activity and total BAs in serum, liver, and feces were assessed. KEY FINDINGS Hyperlipidaemic groups (HF-CO and HF-SFO) had: 1) increased intestinal BA uptake, Asbt and Osta/b mRNA expression, and ASBT staining 2) increased BA in serum, 3) decreased hepatic expression of Ntcp, Bsep, and Cyp7a1 mRNA, and NTCP staining 4) increased activity of HMG-CoA reductase, 5) increased hepatic expression of Fxr and Shp mRNA, 6) decreased hepatic expression of Lrh-1 and Hnf4a mRNA, and 7) decreased BA in Feces when compared to their respective controls (N-CO and N-SFO) and experimental groups (HF-CO + LF and HF-SFO + LF). Immunostaining revealed increased intestinal Asbt and hepatic Ntcp protein expression in the HF-CO and HF-SFO groups compared to control and experimental groups. SIGNIFICANCE Incorporating probiotics like MCC2760 abrogated hyperlipidemia-induced changes in the intestinal uptake, hepatic synthesis, and enterohepatic transporters of BA in rats. Probiotic MCC2760 can be used to modulate lipid metabolism in high-fat-induced hyperlipidemic conditions.
Collapse
Affiliation(s)
- Devika Nair
- Dept. of Biochemistry, CSIR-Central Food Technological Research Institute, Mysore 570020, Karnataka, India.; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad-201002, India
| | - Mehrdad Zarei
- Dept. of Biochemistry, CSIR-Central Food Technological Research Institute, Mysore 570020, Karnataka, India
| | - Prakash Halami
- Dept. of Microbiology & Fermentation Technology, CSIR-Central Food Technological Research Institute, Mysore 570020, Karnataka, India
| | - Ramaprasad Talahalli
- Dept. of Biochemistry, CSIR-Central Food Technological Research Institute, Mysore 570020, Karnataka, India.; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad-201002, India..
| |
Collapse
|
13
|
da Silva Batista E, Nakandakari SCBR, Ramos da Silva AS, Pauli JR, Pereira de Moura L, Ropelle ER, Camargo EA, Cintra DE. Omega-3 pleiad: The multipoint anti-inflammatory strategy. Crit Rev Food Sci Nutr 2022; 64:4817-4832. [PMID: 36382659 DOI: 10.1080/10408398.2022.2146044] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Omega 3 (ω3) fatty acids have been described since the 1980s as promising anti-inflammatory substances. Prostaglandin and leukotriene modulation were exhaustively explored as the main reason for ω3 beneficial outcomes. However, during the early 2000s, after the human genome decoding advent, the nutrigenomic approaches exhibited an impressive plethora of ω3 targets, now under the molecular point of view. Different G protein-coupled receptors (GPCRs) recognizing ω3 and its derivatives appear to be responsible for blocking inflammation and insulin-sensitizing effects. A new class of ω3-derived substances, such as maresins, resolvins, and protectins, increases ω3 actions. Inflammasome disruption, the presence of GPR120 on immune cell surfaces, and intracellular crosstalk signaling mediated by PPARγ compose the last discoveries regarding the multipoint anti-inflammatory targets for this nutrient. This review shows a detailed mechanistic proposal to understand ω3 fatty acid action over the inflammatory environment in the background of several chronic diseases.
Collapse
Affiliation(s)
- Ellencristina da Silva Batista
- Graduate Program of Health Sciences (PPGCS), Federal University of Sergipe, Aracaju, Sergipe, Brazil
- Nutritional Genomics Laboratory, LabGeN, School of Applied Sciences, UNICAMP, São Paulo, Brazil
- Nutrigenomics and Lipids Research Center, CELN, School of Applied Sciences, UNICAMP, São Paulo, Brazil
- Nutrition Department, Federal University of Sergipe, Lagarto, Sergipe, Brazil
| | - Susana Castelo Branco Ramos Nakandakari
- Nutritional Genomics Laboratory, LabGeN, School of Applied Sciences, UNICAMP, São Paulo, Brazil
- Nutrigenomics and Lipids Research Center, CELN, School of Applied Sciences, UNICAMP, São Paulo, Brazil
| | | | - José Rodrigo Pauli
- Nutrigenomics and Lipids Research Center, CELN, School of Applied Sciences, UNICAMP, São Paulo, Brazil
- Laboratory of Molecular Biology of Exercise, School of Applied Sciences, UNICAMP, São Paulo, Brazil
| | - Leandro Pereira de Moura
- Laboratory of Molecular Biology of Exercise, School of Applied Sciences, UNICAMP, São Paulo, Brazil
| | - Eduardo Rochete Ropelle
- Nutrigenomics and Lipids Research Center, CELN, School of Applied Sciences, UNICAMP, São Paulo, Brazil
- Laboratory of Molecular Biology of Exercise, School of Applied Sciences, UNICAMP, São Paulo, Brazil
| | - Enilton A Camargo
- Graduate Program of Health Sciences (PPGCS), Federal University of Sergipe, Aracaju, Sergipe, Brazil
- Department of Physiology, Federal University of Sergipe, São Cristóvão, Sergipe, Brazil
| | - Dennys Esper Cintra
- Nutritional Genomics Laboratory, LabGeN, School of Applied Sciences, UNICAMP, São Paulo, Brazil
- Nutrigenomics and Lipids Research Center, CELN, School of Applied Sciences, UNICAMP, São Paulo, Brazil
- OCRC - Obesity and Comorbidities Research Center, UNICAMP, São Paulo, Brazil
| |
Collapse
|
14
|
Turpin W, Dong M, Sasson G, Raygoza Garay JA, Espin-Garcia O, Lee SH, Neustaeter A, Smith MI, Leibovitzh H, Guttman DS, Goethel A, Griffiths AM, Huynh HQ, Dieleman LA, Panaccione R, Steinhart AH, Silverberg MS, Aumais G, Jacobson K, Mack D, Murthy SK, Marshall JK, Bernstein CN, Abreu MT, Moayyedi P, Paterson AD, Xu W, Croitoru K. Mediterranean-Like Dietary Pattern Associations With Gut Microbiome Composition and Subclinical Gastrointestinal Inflammation. Gastroenterology 2022; 163:685-698. [PMID: 35643175 DOI: 10.1053/j.gastro.2022.05.037] [Citation(s) in RCA: 71] [Impact Index Per Article: 23.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Revised: 05/17/2022] [Accepted: 05/18/2022] [Indexed: 12/15/2022]
Abstract
BACKGROUND & AIMS Case-control studies have shown that patients with Crohn's disease (CD) have a microbial composition different from healthy individuals. Although the causes of CD are unknown, epidemiologic studies suggest that diet is an important contributor to CD risk, potentially via modulation of bacterial composition and gut inflammation. We hypothesized that long-term dietary clusters (DCs) are associated with gut microbiome compositions and gut inflammation. Our objectives were to identify dietary patterns and assess whether they are associated with alterations in specific gut microbial compositions and subclinical levels of gut inflammation in a cohort of healthy first-degree relatives (FDRs) of patients with CD. METHODS As part of the Genetic, Environmental, Microbial (GEM) Project, we recruited a cohort of 2289 healthy FDRs of patients with CD. Individuals provided stool samples and answered a validated food frequency questionnaire reflecting their habitual diet during the year before sample collection. Unsupervised analysis identified 3 dietary and 3 microbial composition clusters. RESULTS DC3, resembling the Mediterranean diet, was strongly associated with a defined microbial composition, with an increased abundance of fiber-degrading bacteria, such as Ruminococcus, as well as taxa such as Faecalibacterium. The DC3 diet was also significantly associated with lower levels of subclinical gut inflammation, defined by fecal calprotectin, compared with other dietary patterns. No significant associations were found between individual food items and fecal calprotectin, suggesting that long-term dietary patterns rather than individual food items contribute to subclinical gut inflammation. Additionally, mediation analysis demonstrated that DC3 had a direct effect on subclinical inflammation that was partially mediated by the microbiota. CONCLUSIONS Overall, these results indicated that Mediterranean-like dietary patterns are associated with microbiome and lower intestinal inflammation. This study will help guide future dietary strategies that affect microbial composition and host gut inflammation to prevent diseases.
Collapse
Affiliation(s)
- Williams Turpin
- Zane Cohen Centre for Digestive Diseases, Mount Sinai Hospital, Toronto, Ontario, Canada
| | - Mei Dong
- Department of Biostatistics, Dalla Lana School of Public Health, University of Toronto, Toronto, Ontario, Canada
| | - Gila Sasson
- Zane Cohen Centre for Digestive Diseases, Mount Sinai Hospital, Toronto, Ontario, Canada
| | - Juan Antonio Raygoza Garay
- Zane Cohen Centre for Digestive Diseases, Mount Sinai Hospital, Toronto, Ontario, Canada; Department of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Osvaldo Espin-Garcia
- Division of Biostatistics, Dalla Lana School of Public Health, University of Toronto, and Department of Biostatistics, Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
| | - Sun-Ho Lee
- Zane Cohen Centre for Digestive Diseases, Mount Sinai Hospital, Toronto, Ontario, Canada; Department of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Anna Neustaeter
- Zane Cohen Centre for Digestive Diseases, Mount Sinai Hospital, Toronto, Ontario, Canada
| | - Michelle I Smith
- Zane Cohen Centre for Digestive Diseases, Mount Sinai Hospital, Toronto, Ontario, Canada
| | - Haim Leibovitzh
- Zane Cohen Centre for Digestive Diseases, Mount Sinai Hospital, Toronto, Ontario, Canada
| | - David S Guttman
- Department of Cell & Systems Biology, University of Toronto, Toronto, Ontario, Canada; Centre for the Analysis of Genome Evolution & Function, University of Toronto, Toronto, Ontario, Canada
| | - Ashleigh Goethel
- Zane Cohen Centre for Digestive Diseases, Mount Sinai Hospital, Toronto, Ontario, Canada
| | - Anne M Griffiths
- Division of Gastroenterology, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Hien Q Huynh
- Division of Gastroenterology and Nutrition, Department of Pediatrics, University of Alberta, Edmonton, Alberta, Canada
| | - Levinus A Dieleman
- Division of Gastroenterology and the Centre of Excellence for Gastrointestinal Inflammation and Immunity Research (CEGIIR), Department of Medicine, University of Alberta, Edmonton, Alberta, Canada
| | - Remo Panaccione
- Inflammatory Bowel Disease Clinic, Division of Gastroenterology and Hepatology, University of Calgary, Calgary, Alberta, Canada
| | - A Hillary Steinhart
- Zane Cohen Centre for Digestive Diseases, Mount Sinai Hospital, Toronto, Ontario, Canada; Division of Biostatistics, Dalla Lana School of Public Health, University of Toronto, and Department of Biostatistics, Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
| | - Mark S Silverberg
- Zane Cohen Centre for Digestive Diseases, Mount Sinai Hospital, Toronto, Ontario, Canada
| | - Guy Aumais
- Department of Medicine, Hôpital Maisonneuve-Rosemont, Montreal University, Montreal, Quebec, Canada
| | - Kevan Jacobson
- Canadian Gastro-Intestinal Epidemiology Consortium (CanGIEC); British Columbia Children's Hospital Research Institute, University of British Columbia, Vancouver, British Columbia, Canada
| | - David Mack
- Division of Gastroenterology, Hepatology & Nutrition, Children's Hospital of Eastern Ontario and University of Ottawa, Ottawa, Ontario, Canada
| | - Sanjay K Murthy
- The Ottawa Hospital Inflammatory Bowel Disease Centre, Department of Medicine, University of Ottawa, Ottawa, Ontario, Canada
| | - John K Marshall
- Department of Medicine, McMaster University, Farncombe Family Digestive Health Research Institute, Hamilton, Ontario, Canada
| | - Charles N Bernstein
- Inflammatory Bowel Disease Clinical and Research Centre, and Department of Internal Medicine, Max Rady College of Medicine, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Maria T Abreu
- Department of Medicine, Crohn's and Colitis Center, University of Miami Miller School of Medicine, Miami, Florida
| | - Paul Moayyedi
- Department of Medicine, McMaster University, Farncombe Family Digestive Health Research Institute, Hamilton, Ontario, Canada
| | - Andrew D Paterson
- Department of Biostatistics, Dalla Lana School of Public Health, University of Toronto, Toronto, Ontario, Canada; Genetics and Genome Biology, The Hospital for Sick Children Research Institute, The Hospital for Sick Children, Toronto, Ontario, Canada
| | | | - Wei Xu
- Division of Biostatistics, Dalla Lana School of Public Health, University of Toronto, and Department of Biostatistics, Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
| | - Kenneth Croitoru
- Zane Cohen Centre for Digestive Diseases, Mount Sinai Hospital, Toronto, Ontario, Canada.
| |
Collapse
|
15
|
Broughton P, Troncoso M, Corker A, Williams A, Bolus D, Munoz G, McWhorter C, Roerden H, Huebsch P, DeLeon-Pennell KY. Riding the wave: a quantitative report of electrocardiogram utilization for myocardial infarction confirmation. Am J Physiol Heart Circ Physiol 2022; 323:H378-H387. [PMID: 35802516 PMCID: PMC9359650 DOI: 10.1152/ajpheart.00201.2022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/25/2022] [Revised: 06/22/2022] [Accepted: 07/07/2022] [Indexed: 11/22/2022]
Abstract
The purpose of this study was to generate a quantitative profile of electrocardiograms (ECGs) for confirming surgical success of permanent coronary artery ligation. An ECG was recorded at baseline, and 0, 1, and 5 min after ligation and analyzed using iWorkx LabScribe software. Cohort 1 (C57Bl6/J, n = 8/sex) was enrolled to determine ECG characteristics that were confirmed in cohort 2 (C57Bl6/J, n = 6/sex; CD8-/-n = 6 males/4 females). Of the 16 mice in cohort 1, 12 (6/sex) had an infarct ≥35% and four mice (2/sex) had <35% based on 2,3,5-triphenyltetrazolium chloride staining. After ligation, the QRS complex and R-S amplitude were significantly different compared with baseline. No differences were observed in the R-S amplitude between mice with infarcts ≥35% versus <35% at any time point, whereas the QRS complex was significant 1 min after ligation. Receiver operating characteristic (ROC) curve linked changes in the QRS complex but not the R-S amplitude at 1 and 5 min with surgical success. Data were normalized to baseline values to calculate fold change. ROC analysis of the normalized QRS data indicated strong sensitivity and specificity for infarcts ≥35%; normalized R-S amplitude remained nonsignificant. With a cutoff generated by ROC analysis of cohort 1 (>80% sensitivity; >90% specificity), the non-normalized QRS complex of cohort 2 had an 86% success rate (2 false positives; 1 false negative). The normalized data had a 77% success rate (2 false positives; 3 false negatives). Neither sex nor genotype was associated with false predictions (P = 0.18). Our data indicate that the area under the QRS complex 1 min after ligation can improve reproducibility in MI surgeries.NEW & NOTEWORTHY Our study describes a quantitative method for using an electrocardiogram (ECG) to determine which animals have infarcts that reflect coronary artery ligation. Using a quantitative ECG, investigators will have the benefit of having real-time feedback during the procedure, which will ultimately decrease the amount of time investigators spend performing experiments. This overall increase in efficiency will help investigators decrease animal numbers used due to better surgical outcomes.
Collapse
Affiliation(s)
- Philip Broughton
- Division of Cardiology, Department of Medicine, Medical University of South Carolina, Charleston, South Carolina
| | - Miguel Troncoso
- Research Service, Ralph H. Johnson Veterans Affairs Medical Center, Charleston, South Carolina
| | - Alexa Corker
- Division of Cardiology, Department of Medicine, Medical University of South Carolina, Charleston, South Carolina
| | - Alexus Williams
- Division of Cardiology, Department of Medicine, Medical University of South Carolina, Charleston, South Carolina
| | - Dawson Bolus
- College of Medicine, Medical University of South Carolina, Charleston, South Carolina
| | - Gualberto Munoz
- College of Medicine, Medical University of South Carolina, Charleston, South Carolina
| | - Caroline McWhorter
- College of Medicine, Medical University of South Carolina, Charleston, South Carolina
| | - Hallie Roerden
- College of Medicine, Medical University of South Carolina, Charleston, South Carolina
| | - Penny Huebsch
- College of Medicine, Medical University of South Carolina, Charleston, South Carolina
| | - Kristine Y DeLeon-Pennell
- Division of Cardiology, Department of Medicine, Medical University of South Carolina, Charleston, South Carolina
- Research Service, Ralph H. Johnson Veterans Affairs Medical Center, Charleston, South Carolina
| |
Collapse
|
16
|
Halade GV, Kain V, De La Rosa X, Lindsey ML. Metabolic transformation of fat in obesity determines the inflammation resolving capacity of splenocardiac and cardiorenal networks in heart failure. Am J Physiol Heart Circ Physiol 2022; 322:H953-H970. [PMID: 35333119 PMCID: PMC9054267 DOI: 10.1152/ajpheart.00684.2021] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Revised: 03/23/2022] [Accepted: 03/23/2022] [Indexed: 01/02/2023]
Abstract
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.
Collapse
Affiliation(s)
- Ganesh V Halade
- Division of Cardiovascular Sciences, Department of Medicine, The University of South Florida, Tampa, Florida
| | - Vasundhara Kain
- Division of Cardiovascular Sciences, Department of Medicine, The University of South Florida, Tampa, Florida
| | - Xavier De La Rosa
- Department of Anesthesiology, Perioperative and Pain Medicine, Center for Experimental Therapeutics and Reperfusion Injury, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Merry L Lindsey
- Department of Cellular and Integrative Physiology, Center for Heart and Vascular Research, University of Nebraska Medical Center, Omaha, Nebraska
- Research Service, Nebraska-Western Iowa Health Care System, Omaha, Nebraska
| |
Collapse
|
17
|
Halade GV, Lee DH. Inflammation and resolution signaling in cardiac repair and heart failure. EBioMedicine 2022; 79:103992. [PMID: 35405389 PMCID: PMC9014358 DOI: 10.1016/j.ebiom.2022.103992] [Citation(s) in RCA: 128] [Impact Index Per Article: 42.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2021] [Revised: 03/22/2022] [Accepted: 03/24/2022] [Indexed: 12/11/2022] Open
Abstract
Unresolved inflammation is a key mediator of advanced heart failure. Especially, damage, pathogen, and lifestyle-associated molecular patterns are the major factors in initiating baseline inflammatory diseases, particularly in cardiac pathology. After a significant cardiac injury like a heart attack, splenic and circulating leukocytes begin a highly optimized sequence of immune cell recruitment (neutrophils and monocytes) to coordinate effective tissue repair. An injured cardiac tissue repair and homeostasis are dependent on clearance of cellular debris where the recruited leukocytes transition from a pro-inflammatory to a reparative program through resolution process. After a cardiac injury, macrophages play a decisive role in cardiac repair through the biosynthesis of endogenous lipid mediators that ensure a timely tissue repair while avoiding chronic inflammation and impaired cardiac repair. However, dysregulation of resolution of inflammation processes due to cardiometabolic defects (obesity, hypertension, and diabetes), aging, or co-medication(s) lead to impaired cardiac repair. Hence, the presented review demonstrates the fundamental role of leukocytes, in particular macrophages orchestrate the inflammation and resolution biology, focusing on the biosynthesis of specialized lipid mediators in cardiac repair and heart failure. This work was supported by research funds from National Institutes of Health (AT006704, HL132989, and HL144788) to G.V.H. The authors acknowledges the use of Servier Medical Art image bank and Biorender that is used to create schematic Figures 1–3.
Collapse
Affiliation(s)
- Ganesh V Halade
- Division of Cardiovascular Sciences, Department of Medicine, Heart Institute, University of South Florida, 560 Channelside Dr, Tampa, FL 33602, United States.
| | - Dae Hyun Lee
- Division of Cardiovascular Sciences, Department of Medicine, Heart Institute, University of South Florida, 560 Channelside Dr, Tampa, FL 33602, United States
| |
Collapse
|
18
|
Kain V, Halade GV. Dysfunction of resolution receptor triggers cardiomyopathy of obesity and signs of non-resolving inflammation in heart failure. Mol Cell Endocrinol 2022; 542:111521. [PMID: 34843898 PMCID: PMC10515100 DOI: 10.1016/j.mce.2021.111521] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/17/2021] [Revised: 11/03/2021] [Accepted: 11/23/2021] [Indexed: 10/19/2022]
Abstract
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.
Collapse
Affiliation(s)
- Vasundhara Kain
- Division of Cardiovascular Sciences, Department of Medicine, University of South Florida, Tampa, FL, 33602, USA
| | - Ganesh V Halade
- Division of Cardiovascular Sciences, Department of Medicine, University of South Florida, Tampa, FL, 33602, USA.
| |
Collapse
|
19
|
Collins HE, Anderson JC, Wende AR, Chatham JC. Cardiomyocyte stromal interaction molecule 1 is a key regulator of Ca 2+ -dependent kinase and phosphatase activity in the mouse heart. Physiol Rep 2022; 10:e15177. [PMID: 35179826 PMCID: PMC8855923 DOI: 10.14814/phy2.15177] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2021] [Revised: 01/04/2022] [Accepted: 01/07/2022] [Indexed: 04/26/2023] Open
Abstract
Stromal interaction molecule 1 (STIM1) is a major regulator of store-operated calcium entry in non-excitable cells. Recent studies have suggested that STIM1 plays a role in pathological hypertrophy; however, the physiological role of STIM1 in the heart is not well understood. We have shown that mice with a cardiomyocyte deletion of STIM1 (cr STIM1-/- ) develop ER stress, mitochondrial, and metabolic abnormalities, and dilated cardiomyopathy. However, the specific signaling pathways and kinases regulated by STIM1 are largely unknown. Therefore, we used a discovery-based kinomics approach to identify kinases differentially regulated by STIM1. Twelve-week male control and cr STIM1-/- mice were injected with saline or phenylephrine (PE, 15 mg/kg, s.c, 15 min), and hearts obtained for analysis of the Serine/threonine kinome. Primary analysis was performed using BioNavigator 6.0 (PamGene), using scoring from the Kinexus PhosphoNET database and GeneGo network modeling, and confirmed using standard immunoblotting. Kinomics revealed significantly lower PKG and protein kinase C (PKC) signaling in the hearts of the cr STIM1-/- in comparison to control hearts, confirmed by immunoblotting for the calcium-dependent PKC isoform PKCα and its downstream target MARCKS. Similar reductions in cr STIM1-/- hearts were found for the kinases: MEK1/2, AMPK, and PDPK1, and in the activity of the Ca2+ -dependent phosphatase, calcineurin. Electrocardiogram analysis also revealed that cr STIM1-/- mice have significantly lower HR and prolonged QT interval. In conclusion, we have shown several calcium-dependent kinases and phosphatases are regulated by STIM1 in the adult mouse heart. This has important implications in understanding how STIM1 contributes to the regulation of cardiac physiology and pathophysiology.
Collapse
Affiliation(s)
- Helen E. Collins
- Division of Environmental MedicineDepartment of MedicineUniversity of LouisvilleLouisvilleKentuckyUSA
| | - Joshua C. Anderson
- Department of Radiation OncologyUniversity of Alabama at BirminghamBirminghamAlabamaUSA
| | - Adam R. Wende
- Division of Molecular and Cellular PathologyDepartment of PathologyUniversity of Alabama at BirminghamBirminghamAlabamaUSA
| | - John C. Chatham
- Division of Molecular and Cellular PathologyDepartment of PathologyUniversity of Alabama at BirminghamBirminghamAlabamaUSA
| |
Collapse
|
20
|
Firoozi D, Masoumi SJ, Ranjbar S, Shivappa N, Hebert JR, Zare M, Poustchi H, Hoseini FS. The Association between Energy-Adjusted Dietary Inflammatory Index, Body Composition, and Anthropometric Indices in COVID-19-Infected Patients: A Case-Control Study in Shiraz, Iran. Int J Clin Pract 2022; 2022:5452488. [PMID: 35685606 PMCID: PMC9159238 DOI: 10.1155/2022/5452488] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/06/2021] [Accepted: 02/24/2022] [Indexed: 11/17/2022] Open
Abstract
BACKGROUND AND AIMS Inflammation is strongly associated with the severity and mortality rate of SARS-CoV-2 disease (COVID-19). Dietary factors have a crucial role in preventing chronic and systemic inflammation. This study aimed to evaluate the association between energy-adjusted dietary inflammatory index (E-DII) scores and body composition parameters in COVID-19-infected patients compared to noninfected controls. METHODS A total of 133 COVID-19-infected patients and 322 noninfected controls were selected and enrolled from the Cohort Study of Employees of Shiraz University of Medical Sciences. E-DII score was calculated based on a validated food frequency questionnaire (FFQ) and body composition was measured using In-Body 770 equipment. Logistic regression models were utilized to estimate the odds ratio (OR). RESULTS In the control group, the mean E-DII score was significantly lower than the case group (-2.05 vs. -0.30, P ≤ 0.001), indicating that the diet of COVID-19-infected subjects was more proinflammatory than the controls. For every 1 unit increase in E-DII score, the odds of infection with COVID-19 was nearly triple (OR: 2.86, CI: 2.30, 3.35, P ≤ 0.001). Moreover, for each unit increase in body mass index (BMI), the odds of infection to COVID-19 increased by 7% (OR: 1.07, CI: 1.01, 1.13, P = 0.02). No significant difference was observed for other anthropometric parameters. CONCLUSION The findings revealed that obese people and those consuming a more proinflammatory diet were more susceptible to coronavirus infection. Therefore, maintaining ideal body weight and consuming a more anti-inflammatory diet can decrease the probability of COVID-19 infection.
Collapse
Affiliation(s)
- Donya Firoozi
- Student Research Committee, School of Nutrition and Food Sciences, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Seyed Jalil Masoumi
- Nutrition Research Center, School of Nutrition and Food Sciences, Shiraz University of Medical Science, Shiraz, Iran
- Center for Cohort Study of SUMS Employees' Health, Shiraz University of Medical Sciences, Shiraz, Iran
- Gastroenterohepatology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Sara Ranjbar
- Nutrition Research Center, School of Nutrition and Food Sciences, Shiraz University of Medical Science, Shiraz, Iran
| | - Nitin Shivappa
- Cancer Prevention and Control Program and Department of Epidemiology and Biostatistics, Arnold School of Public Health, University of South Carolina, Columbia, SC, USA
- Department of Nutrition, Connecting Health Innovations LLC, Columbia, SC, USA
| | - James R. Hebert
- Cancer Prevention and Control Program and Department of Epidemiology and Biostatistics, Arnold School of Public Health, University of South Carolina, Columbia, SC, USA
- Department of Nutrition, Connecting Health Innovations LLC, Columbia, SC, USA
| | - Morteza Zare
- Nutrition Research Center, School of Nutrition and Food Sciences, Shiraz University of Medical Science, Shiraz, Iran
| | - Hossein Poustchi
- Digestive Oncology Research Center, Digestive Diseases Research Institute, Shariati Hospital, Tehran University of Medical Sciences, Tehran, Iran
- Digestive Diseases Research Center, Digestive Diseases Research Institute, Shariati Hospital, Tehran University of Medical Sciences, Tehran, Iran
- Liver and Pancreatobiliary Diseases Research Center, Digestive Diseases Research Institute, Shariati Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | - Faeze Sadat Hoseini
- Department of Nutrition, Science and Research Branch, Islamic Azad University, Tehran, Iran
| |
Collapse
|
21
|
Ciumărnean L, Milaciu MV, Negrean V, Orășan OH, Vesa SC, Sălăgean O, Iluţ S, Vlaicu SI. Cardiovascular Risk Factors and Physical Activity for the Prevention of Cardiovascular Diseases in the Elderly. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2021; 19:207. [PMID: 35010467 PMCID: PMC8751147 DOI: 10.3390/ijerph19010207] [Citation(s) in RCA: 71] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/06/2021] [Revised: 12/19/2021] [Accepted: 12/23/2021] [Indexed: 12/12/2022]
Abstract
Cardiovascular diseases create an important burden on the public health systems, especially in the elderly, mostly because this group of patients frequently suffer from multiple comorbidities. Accumulating cardiovascular risk factors during their lifetime has a detrimental effect on an older adult's health status. The modifiable and non-modifiable cardiovascular risk factors are very diverse, and are frequently in a close relationship with the metabolic comorbidities of the elderly, mainly obesity and Diabetes Mellitus. In this review, we aim to present the most important cardiovascular risk factors which link aging and cardiovascular diseases, starting from the pathophysiological links between these factors and the aging process. Next, we will further review the main interconnections between obesity and Diabetes Mellitus and cardiovascular diseases of the elderly. Lastly, we consider the most important aspects related to prevention through lifestyle changes and physical activity on the occurrence of cardiovascular diseases in the elderly.
Collapse
Affiliation(s)
- Lorena Ciumărnean
- Department 5 Internal Medicine, 4th Medical Clinic, Faculty of Medicine, “Iuliu Haţieganu” University of Medicine and Pharmacy, 400015 Cluj-Napoca, Romania; (L.C.); (M.V.M.); (V.N.); (O.H.O.)
| | - Mircea Vasile Milaciu
- Department 5 Internal Medicine, 4th Medical Clinic, Faculty of Medicine, “Iuliu Haţieganu” University of Medicine and Pharmacy, 400015 Cluj-Napoca, Romania; (L.C.); (M.V.M.); (V.N.); (O.H.O.)
| | - Vasile Negrean
- Department 5 Internal Medicine, 4th Medical Clinic, Faculty of Medicine, “Iuliu Haţieganu” University of Medicine and Pharmacy, 400015 Cluj-Napoca, Romania; (L.C.); (M.V.M.); (V.N.); (O.H.O.)
| | - Olga Hilda Orășan
- Department 5 Internal Medicine, 4th Medical Clinic, Faculty of Medicine, “Iuliu Haţieganu” University of Medicine and Pharmacy, 400015 Cluj-Napoca, Romania; (L.C.); (M.V.M.); (V.N.); (O.H.O.)
| | - Stefan Cristian Vesa
- Department 2 Functional Sciences, Discipline of Pharmacology, Toxicology and Clinical Pharmacology, Faculty of Medicine, “Iuliu Haţieganu” University of Medicine and Pharmacy, 400337 Cluj-Napoca, Romania
| | - Octavia Sălăgean
- Regional Institute of Gastroenterology and Hepatology ‘Octavian Fodor’ Cluj-Napoca, 400162 Cluj-Napoca, Romania;
| | - Silvina Iluţ
- Department 10 Neurosciences, Discipline of Neurology, Faculty of Medicine, “Iuliu Haţieganu” University of Medicine and Pharmacy, 400012 Cluj-Napoca, Romania;
| | - Sonia Irina Vlaicu
- Department 5 Internal Medicine, 1st Medical Clinic, Faculty of Medicine, “Iuliu Haţieganu” University of Medicine and Pharmacy, 400012 Cluj-Napoca, Romania;
| |
Collapse
|
22
|
Batista ES, da Silva Rios T, Muñoz VR, Jesus JS, Vasconcelos MM, da Cunha DT, Marques-Rocha JL, Nakandakari SCBR, Lara R, da Silva ASR, Pauli JR, Ropelle ER, Mekary RA, de Moura LP, Camargo EA, Cintra DE. Omega-3 mechanism of action in inflammation and endoplasmic reticulum stress in mononuclear cells from overweight non-alcoholic fatty liver disease participants: study protocol for the "Brazilian Omega Study" (BROS)-a randomized controlled trial. Trials 2021; 22:927. [PMID: 34922604 PMCID: PMC8684080 DOI: 10.1186/s13063-021-05702-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Accepted: 10/08/2021] [Indexed: 12/03/2022] Open
Abstract
Abstract The low-grade inflammation is pivotal in obesity and its comorbidities; however, the inflammatory proteins are out of target for traditional drug therapy. Omega-3 (ω3) fatty acids can modulate the downstream signaling of Toll-like receptor (TLR) and tumor necrosis factor-α receptor (TNFα) through GPR120, a G-protein-coupled receptor, a mechanism not yet elucidated in humans. This work aims to investigate if the ω3 supplementation, at a feasible level below the previously recommended level in the literature, is enough to disrupt the inflammation and endoplasmic reticulum stress (ER-stress), and also if in acute treatment (3 h) ω3 can activate the GPR120 in peripheral blood mononuclear cells (PBMC) and leukocytes from overweight non-alcoholic fatty liver disease (NAFLD) participants. The R270H variant of the Ffar4 (GPR120 gene) will also be explored about molecular responses and blood lipid profiles. A triple-blind, prospective clinical trial will be conducted in overweight men and women, aged 19–75 years, randomized into placebo or supplemented (2.2 g of ω3 [EPA+DHA]) groups for 28 days. For sample calculation, it was considered the variation of TNFα protein and a 40% dropout rate, obtaining 22 individuals in each group. Volunteers will be recruited among patients with NAFLD diagnosis. Anthropometric parameters, food intake, physical activity, total serum lipids, complete fatty acid blood profile, and glycemia will be evaluated pre- and post-supplementation. In the PBMC and neutrophils, the protein content and gene expression of markers related to inflammation (TNFα, MCP1, IL1β, IL6, IL10, JNK, and TAK1), ER-stress (ATF1, ATF6, IRE1, XBP1, CHOP, eIF2α, eIF4, HSP), and ω3 pathway (GPR120, β-arrestin2, Tab1/2, and TAK1) will be evaluated using Western blot and RT-qPCR. Participants will be genotyped for the R270H (rs116454156) variant using the TaqMan assay. It is hypothesized that attenuation of inflammation and ER-stress signaling pathways in overweight and NAFLD participants will be achieved through ω3 supplementation through binding to the GPR120 receptor. Trial registration ClinicalTrials.gov #RBR-7x8tbx. Registered on May 10, 2018, with the Brazilian Registry of Clinical Trials. Supplementary Information The online version contains supplementary material available at 10.1186/s13063-021-05702-x.
Collapse
Affiliation(s)
- Ellencristina Silva Batista
- Graduate Program of Health Sciences (PPGCS), Federal University of Sergipe, Aracaju, Sergipe, Brazil.,Laboratory of Nutritional Genomics, School of Applied Sciences, University of Campinas, Pedro Zaccaria, 1300 Zip, Limeira, 13484-350, Brazil.,Nutrition Department, Federal University of Sergipe, Lagarto, Sergipe, Brazil.,Lipids and Nutrigenomics Research Center, School of Applied Sciences, University of Campinas, Limeira, Brazil
| | - Thaiane da Silva Rios
- Laboratory of Nutritional Genomics, School of Applied Sciences, University of Campinas, Pedro Zaccaria, 1300 Zip, Limeira, 13484-350, Brazil.,Lipids and Nutrigenomics Research Center, School of Applied Sciences, University of Campinas, Limeira, Brazil
| | - Vitor Rosetto Muñoz
- Lipids and Nutrigenomics Research Center, School of Applied Sciences, University of Campinas, Limeira, Brazil.,Laboratory of Molecular Biology of Exercise, School of Applied Sciences, University of Campinas, Limeira, Brazil
| | - Joyce Santos Jesus
- Nutrition Department, Federal University of Sergipe, Lagarto, Sergipe, Brazil
| | | | - Diogo Thimóteo da Cunha
- Multidisciplinary Laboratory of Food and Health, School of Applied Sciences, University of Campinas, Limeira, Brazil
| | - Jose Luis Marques-Rocha
- Department of Integrated Health Education, Federal University of Espírito Santo, Vitoria, Brazil
| | - Susana Castelo Branco Ramos Nakandakari
- Laboratory of Nutritional Genomics, School of Applied Sciences, University of Campinas, Pedro Zaccaria, 1300 Zip, Limeira, 13484-350, Brazil.,Lipids and Nutrigenomics Research Center, School of Applied Sciences, University of Campinas, Limeira, Brazil
| | - Roberta Lara
- Laboratory of Nutritional Genomics, School of Applied Sciences, University of Campinas, Pedro Zaccaria, 1300 Zip, Limeira, 13484-350, Brazil
| | - Adelino Sanchez Ramos da Silva
- School of Physical Education and Sport of Ribeirão Preto, University of São Paulo (USP), Ribeirão Preto, São Paulo, Brazil
| | - José Rodrigo Pauli
- Lipids and Nutrigenomics Research Center, School of Applied Sciences, University of Campinas, Limeira, Brazil.,Laboratory of Molecular Biology of Exercise, School of Applied Sciences, University of Campinas, Limeira, Brazil
| | - Eduardo Rochete Ropelle
- Lipids and Nutrigenomics Research Center, School of Applied Sciences, University of Campinas, Limeira, Brazil.,Laboratory of Molecular Biology of Exercise, School of Applied Sciences, University of Campinas, Limeira, Brazil
| | - Rania Angelina Mekary
- Massachusetts College of Pharmacy and Health Sciences (MCPHS) University, Boston, MA, USA.,Department of Neurosurgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Leandro Pereira de Moura
- Laboratory of Molecular Biology of Exercise, School of Applied Sciences, University of Campinas, Limeira, Brazil
| | | | - Dennys Esper Cintra
- Laboratory of Nutritional Genomics, School of Applied Sciences, University of Campinas, Pedro Zaccaria, 1300 Zip, Limeira, 13484-350, Brazil. .,Lipids and Nutrigenomics Research Center, School of Applied Sciences, University of Campinas, Limeira, Brazil.
| |
Collapse
|
23
|
B Gowda S, Gowda D, Kain V, Chiba H, Hui SP, Chalfant CE, Parcha V, Arora P, Halade GV. Sphingosine-1-phosphate interactions in the spleen and heart reflect extent of cardiac repair in mice and failing human hearts. Am J Physiol Heart Circ Physiol 2021; 321:H599-H611. [PMID: 34415189 DOI: 10.1152/ajpheart.00314.2021] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
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.
Collapse
Affiliation(s)
| | - Divyavani Gowda
- Faculty of Health Sciences, Hokkaido University, Sapporo, Japan
| | - Vasundhara Kain
- Division of Cardiovascular Sciences, Department of Medicine, University of South Florida, Tampa, Florida
| | - Hitoshi Chiba
- Department of Nutrition, Sapporo University of Health Sciences, Sapporo, Japan
| | - Shu-Ping Hui
- Faculty of Health Sciences, Hokkaido University, Sapporo, Japan
| | - Charles E Chalfant
- Department of Cell Biology, Microbiology and Molecular Biology, University of South Florida, Tampa, Florida.,Research Service, James A. Haley Veterans' Hospital, Tampa, Florida
| | - Vibhu Parcha
- Division of Cardiovascular Disease, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama
| | - Pankaj Arora
- Division of Cardiovascular Disease, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama
| | - Ganesh V Halade
- Division of Cardiovascular Sciences, Department of Medicine, University of South Florida, Tampa, Florida
| |
Collapse
|
24
|
Tourki B, Halade GV. Heart Failure Syndrome With Preserved Ejection Fraction Is a Metabolic Cluster of Non-resolving Inflammation in Obesity. Front Cardiovasc Med 2021; 8:695952. [PMID: 34409075 PMCID: PMC8367012 DOI: 10.3389/fcvm.2021.695952] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2021] [Accepted: 06/08/2021] [Indexed: 12/20/2022] Open
Abstract
Heart failure with preserved ejection fraction (HFpEF) is an emerging disease with signs of nonresolving inflammation, endothelial dysfunction, and multiorgan defects. Moreover, based on the clinical signs and symptoms and the rise of the obesity epidemic, the number of patients developing HFpEF is increasing. From recent molecular and cellular studies, it becomes evident that HFpEF is not a single and homogenous disease but a cluster of heterogeneous pathophysiology with aging at the base of the pyramid. Obesity superimposed on aging drives the number of inflammatory pathways that intersect with metabolic dysfunction and suboptimal inflammation. Here, we compiled information on obesity-directed macrophage dysfunction that coincide with metabolic defects. Obesity-associated proinflammatory stimuli facilitates heart and interorgan inflammation in HFpEF. Furthermore, diversified mechanisms that drive heart failure urge the need of studying pervasive and unresolved inflammation in animal models to understand HFpEF. A broad and system-based approach will help to study major translational aspects of HFpEF, since no single animal model recapitulates all signs of differential HFpEF stages in the clinical setting. Here, we covered experimental models that target HFpEF and emphasized the advances observed with formyl peptide 2 (FPR2) receptor, a prime sensor that is important in inflammation-resolution signaling. Dysfunction of FPR2 led to the development of spontaneous obesity, impaired macrophage function, and triggered kidney fibrosis, providing evidence of multiorgan defects in HFpEF in an obesogenic aging experimental model.
Collapse
Affiliation(s)
- Bochra Tourki
- Division of Cardiovascular Sciences, Department of Medicine, The University of South Florida, Tampa, FL, United States
| | - Ganesh V Halade
- Division of Cardiovascular Sciences, Department of Medicine, The University of South Florida, Tampa, FL, United States
| |
Collapse
|
25
|
Abstract
Macrophages are essential components of the immune system and play a role in the normal functioning of the cardiovascular system. Depending on their origin and phenotype, cardiac macrophages perform various functions. In a steady-state, these cells play a beneficial role in maintaining cardiac homeostasis by defending the body from pathogens and eliminating apoptotic cells, participating in electrical conduction, vessel patrolling, and arterial tone regulation. However, macrophages also take part in adverse cardiac remodeling that could lead to the development and progression of heart failure (HF) in such HF comorbidities as hypertension, obesity, diabetes, and myocardial infarction. Nevertheless, studies on detailed mechanisms of cardiac macrophage function are still in progress, and could enable potential therapeutic applications of these cells. This review aims to present the latest reports on the origin, heterogeneity, and functions of cardiac macrophages in the healthy heart and in cardiovascular diseases leading to HF. The potential therapeutic use of macrophages is also briefly discussed.
Collapse
|
26
|
Dallio M, Romeo M, Gravina AG, Masarone M, Larussa T, Abenavoli L, Persico M, Loguercio C, Federico A. Nutrigenomics and Nutrigenetics in Metabolic- (Dysfunction) Associated Fatty Liver Disease: Novel Insights and Future Perspectives. Nutrients 2021; 13:1679. [PMID: 34063372 PMCID: PMC8156164 DOI: 10.3390/nu13051679] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Revised: 05/12/2021] [Accepted: 05/13/2021] [Indexed: 12/12/2022] Open
Abstract
Metabolic- (dysfunction) associated fatty liver disease (MAFLD) represents the predominant hepatopathy and one of the most important systemic, metabolic-related disorders all over the world associated with severe medical and socio-economic repercussions due to its growing prevalence, clinical course (steatohepatitis and/or hepatocellular-carcinoma), and related extra-hepatic comorbidities. To date, no specific medications for the treatment of this condition exist, and the most valid recommendation for patients remains lifestyle change. MAFLD has been associated with metabolic syndrome; its development and progression are widely influenced by the interplay between genetic, environmental, and nutritional factors. Nutrigenetics and nutrigenomics findings suggest nutrition's capability, by acting on the individual genetic background and modifying the specific epigenetic expression as well, to influence patients' clinical outcome. Besides, immunity response is emerging as pivotal in this multifactorial scenario, suggesting the interaction between diet, genetics, and immunity as another tangled network that needs to be explored. The present review describes the genetic background contribution to MAFLD onset and worsening, its possibility to be influenced by nutritional habits, and the interplay between nutrients and immunity as one of the most promising research fields of the future in this context.
Collapse
Affiliation(s)
- Marcello Dallio
- Department of Precision Medicine, University of Campania Luigi Vanvitelli, Via S. Pansini 5, 80131 Naples, Italy; (M.R.); (A.G.G.); (C.L.); (A.F.)
| | - Mario Romeo
- Department of Precision Medicine, University of Campania Luigi Vanvitelli, Via S. Pansini 5, 80131 Naples, Italy; (M.R.); (A.G.G.); (C.L.); (A.F.)
| | - Antonietta Gerarda Gravina
- Department of Precision Medicine, University of Campania Luigi Vanvitelli, Via S. Pansini 5, 80131 Naples, Italy; (M.R.); (A.G.G.); (C.L.); (A.F.)
| | - Mario Masarone
- Department of Medicine and Surgery, University of Salerno, Via Allende, 84081 Baronissi, Italy; (M.M.); (M.P.)
| | - Tiziana Larussa
- Department of Health Sciences, University Magna Graecia, viale Europa, 88100 Catanzaro, Italy; (T.L.); (L.A.)
| | - Ludovico Abenavoli
- Department of Health Sciences, University Magna Graecia, viale Europa, 88100 Catanzaro, Italy; (T.L.); (L.A.)
| | - Marcello Persico
- Department of Medicine and Surgery, University of Salerno, Via Allende, 84081 Baronissi, Italy; (M.M.); (M.P.)
| | - Carmelina Loguercio
- Department of Precision Medicine, University of Campania Luigi Vanvitelli, Via S. Pansini 5, 80131 Naples, Italy; (M.R.); (A.G.G.); (C.L.); (A.F.)
| | - Alessandro Federico
- Department of Precision Medicine, University of Campania Luigi Vanvitelli, Via S. Pansini 5, 80131 Naples, Italy; (M.R.); (A.G.G.); (C.L.); (A.F.)
| |
Collapse
|
27
|
Role of polyunsaturated fatty acids in ischemic stroke - A perspective of specialized pro-resolving mediators. Clin Nutr 2021; 40:2974-2987. [PMID: 33509668 DOI: 10.1016/j.clnu.2020.12.037] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2020] [Revised: 12/14/2020] [Accepted: 12/26/2020] [Indexed: 12/17/2022]
Abstract
Polyunsaturated fatty acids (PUFAs) have been proposed as beneficial for cardiovascular health. However, results from both epidemiological studies and clinical trials have been inconsistent, whereas most of the animal studies showed promising benefits of PUFAs in the prevention and treatment of ischemic stroke. In recent years, it has become clear that PUFAs are metabolized into various types of bioactive derivatives, including the specialized pro-resolving mediators (SPMs). SPMs exert multiple biofunctions, such as to limit excessive inflammatory responses, regulate lipid metabolism and immune cell functions, decrease production of pro-inflammatory factors, increase anti-inflammatory mediators, as well as to promote tissue repair and homeostasis. Inflammation has been recognised as a key contributor to the pathophysiology of acute ischemic stroke. Owing to their potent pro-resolving actions, SPMs are potential for development of novel anti-stroke therapy. In this review, we will summarize current knowledge of epidemiological studies, basic research and clinical trials concerning PUFAs in stroke prevention and treatment, with special attention to SPMs as the unsung heroes behind PUFAs.
Collapse
|
28
|
Kain V, Ingle KA, Rajasekaran NS, Halade GV. Activation of EP4 receptor limits transition of acute to chronic heart failure in lipoxygenase deficient mice. Am J Cancer Res 2021; 11:2742-2754. [PMID: 33456570 PMCID: PMC7806484 DOI: 10.7150/thno.51183] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2020] [Accepted: 11/20/2020] [Indexed: 12/20/2022] Open
Abstract
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.
Collapse
|
29
|
Lafuse WP, Wozniak DJ, Rajaram MVS. Role of Cardiac Macrophages on Cardiac Inflammation, Fibrosis and Tissue Repair. Cells 2020; 10:E51. [PMID: 33396359 PMCID: PMC7824389 DOI: 10.3390/cells10010051] [Citation(s) in RCA: 222] [Impact Index Per Article: 44.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2020] [Revised: 12/22/2020] [Accepted: 12/28/2020] [Indexed: 12/17/2022] Open
Abstract
The immune system plays a pivotal role in the initiation, development and resolution of inflammation following insult or damage to organs. The heart is a vital organ which supplies nutrients and oxygen to all parts of the body. Heart failure (HF) has been conventionally described as a disease associated with cardiac tissue damage caused by systemic inflammation, arrhythmia and conduction defects. Cardiac inflammation and subsequent tissue damage is orchestrated by the infiltration and activation of various immune cells including neutrophils, monocytes, macrophages, eosinophils, mast cells, natural killer cells, and T and B cells into the myocardium. After tissue injury, monocytes and tissue-resident macrophages undergo marked phenotypic and functional changes, and function as key regulators of tissue repair, regeneration and fibrosis. Disturbance in resident macrophage functions such as uncontrolled production of inflammatory cytokines, growth factors and inefficient generation of an anti-inflammatory response or unsuccessful communication between macrophages and epithelial and endothelial cells and fibroblasts can lead to aberrant repair, persistent injury, and HF. Therefore, in this review, we discuss the role of cardiac macrophages on cardiac inflammation, tissue repair, regeneration and fibrosis.
Collapse
Affiliation(s)
- William P. Lafuse
- Department of Microbial Infection and Immunity, College of Medicine, Ohio State University, Columbus, OH 43210, USA; (W.P.L.); (D.J.W.)
| | - Daniel J. Wozniak
- Department of Microbial Infection and Immunity, College of Medicine, Ohio State University, Columbus, OH 43210, USA; (W.P.L.); (D.J.W.)
- Department of Microbiology, Ohio State University, Columbus, OH 43210, USA
| | - Murugesan V. S. Rajaram
- Department of Microbial Infection and Immunity, College of Medicine, Ohio State University, Columbus, OH 43210, USA; (W.P.L.); (D.J.W.)
| |
Collapse
|
30
|
Schulte F, Asbeutah AA, Benotti PN, Wood GC, Still C, Bistrian BR, Hardt M, Welty FK. The relationship between specialized pro-resolving lipid mediators, morbid obesity and weight loss after bariatric surgery. Sci Rep 2020; 10:20128. [PMID: 33208757 PMCID: PMC7674470 DOI: 10.1038/s41598-020-75353-6] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2020] [Accepted: 09/22/2020] [Indexed: 12/21/2022] Open
Abstract
Obesity and diabetes are associated with chronic inflammation. Specialized pro-resolving lipid mediators (SPMs)—resolvins (Rv), protectins (PD) and maresins (MaR)—actively resolve inflammation. Bariatric surgery achieves remission of diabetes, but mechanisms are unclear. We measured SPMs and proinflammatory eicosanoid levels using liquid chromatography-tandem mass spectrometry in 29 morbidly obese subjects (13 with diabetes) and 15 nondiabetic, mildly obese subjects. Compared to the mildly obese, the morbidly obese had higher levels of SPMs—RvD3, RvD4 and PD1—and white blood cells (WBC) and platelets. Post-surgery, SPM and platelet levels decreased in morbidly obese nondiabetic subjects but not in diabetic subjects, suggesting continued inflammation. Despite similar weight reductions 1 year after surgery (44.6% vs. 46.6%), 8 diabetes remitters had significant reductions in WBC and platelet counts whereas five non-remitters did not. Remitters had a 58.2% decrease (p = 0.03) in 14-HDHA, a maresin pathway marker; non-remitters had an 875.7% increase in 14-HDHA but a 36.9% decrease in MaR1 to a median of 0. In conclusion, higher levels of RvD3, PD1 and their pathway marker, 17-HDHA, are markers of leukocyte activation and inflammation in morbid obesity and diabetes and diminish with weight loss in nondiabetic but not diabetic subjects, possibly representing sustained inflammation in the latter. Lack of diabetes remission after surgically-induced weight loss may be associated with reduced ability to produce MaR1 and sustained inflammation.
Collapse
Affiliation(s)
- Fabian Schulte
- Forsyth Institute, Cambridge, MA, USA.,Department of Developmental Biology, Harvard School of Dental Medicine, Boston, MA, USA
| | - Abdul Aziz Asbeutah
- Division of Cardiology, Beth Israel Deaconess Medical Center, Harvard Medical School, 330 Brookline Avenue, SL 423, Boston, MA, 02215, USA.,Department of Medicine, University of Tennessee Health Science Center, Memphis, TN, USA
| | | | | | | | - Bruce R Bistrian
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Markus Hardt
- Forsyth Institute, Cambridge, MA, USA.,Department of Developmental Biology, Harvard School of Dental Medicine, Boston, MA, USA
| | - Francine K Welty
- Division of Cardiology, Beth Israel Deaconess Medical Center, Harvard Medical School, 330 Brookline Avenue, SL 423, Boston, MA, 02215, USA.
| |
Collapse
|
31
|
Dias IHK, Milic I, Heiss C, Ademowo OS, Polidori MC, Devitt A, Griffiths HR. Inflammation, Lipid (Per)oxidation, and Redox Regulation. Antioxid Redox Signal 2020; 33:166-190. [PMID: 31989835 DOI: 10.1089/ars.2020.8022] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Significance: Inflammation increases during the aging process. It is linked to mitochondrial dysfunction and increased reactive oxygen species (ROS) production. Mitochondrial macromolecules are critical targets of oxidative damage; they contribute to respiratory uncoupling with increased ROS production, redox stress, and a cycle of senescence, cytokine production, and impaired oxidative phosphorylation. Targeting the formation or accumulation of oxidized biomolecules, particularly oxidized lipids, in immune cells and mitochondria could be beneficial for age-related inflammation and comorbidities. Recent Advances: Inflammation is central to age-related decline in health and exhibits a complex relationship with mitochondrial redox state and metabolic function. Improvements in mass spectrometric methods have led to the identification of families of oxidized phospholipids (OxPLs), cholesterols, and fatty acids that increase during inflammation and which modulate nuclear factor erythroid 2-related factor 2 (Nrf2), peroxisome proliferator-activated receptor gamma (PPARγ), activator protein 1 (AP1), and NF-κB redox-sensitive transcription factor activity. Critical Issues: The kinetic and spatial resolution of the modified lipidome has profound and sometimes opposing effects on inflammation, promoting initiation at high concentration and resolution at low concentration of OxPLs. Future Directions: There is an emerging opportunity to prevent or delay age-related inflammation and vascular comorbidity through a resolving (oxy)lipidome that is dependent on improving mitochondrial quality control and restoring redox homeostasis.
Collapse
Affiliation(s)
- Irundika H K Dias
- Aston Medical Research Institute, Aston Medical School, Aston University, Birmingham, United Kingdom
| | - Ivana Milic
- Aston Research Center for Healthy Ageing, School of Life and Health Sciences, Aston University, Birmingham, United Kingdom
| | - Christian Heiss
- Faculty of Health and Medical Sciences, University of Surrey, Guildford, United Kingdom
| | - Opeyemi S Ademowo
- Aston Research Center for Healthy Ageing, School of Life and Health Sciences, Aston University, Birmingham, United Kingdom
| | - Maria Cristina Polidori
- Ageing Clinical Research, Department II of Internal Medicine and Cologne Center for Molecular Medicine Cologne, and CECAD, Faculty of Medicine, University of Cologne, Cologne, Germany
| | - Andrew Devitt
- Aston Research Center for Healthy Ageing, School of Life and Health Sciences, Aston University, Birmingham, United Kingdom
| | - Helen R Griffiths
- Aston Medical Research Institute, Aston Medical School, Aston University, Birmingham, United Kingdom.,Faculty of Health and Medical Sciences, University of Surrey, Guildford, United Kingdom
| |
Collapse
|
32
|
Tourki B, Kain V, Shaikh SR, Leroy X, Serhan CN, Halade GV. Deficit of resolution receptor magnifies inflammatory leukocyte directed cardiorenal and endothelial dysfunction with signs of cardiomyopathy of obesity. FASEB J 2020; 34:10560-10573. [PMID: 32543720 DOI: 10.1096/fj.202000495rr] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2020] [Revised: 05/22/2020] [Accepted: 05/27/2020] [Indexed: 12/13/2022]
Abstract
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.
Collapse
Affiliation(s)
- Bochra Tourki
- Division of Cardiovascular Sciences, Department of Medicine, University of South Florida, Tampa, FL, USA
| | - Vasundhara Kain
- Division of Cardiovascular Sciences, Department of Medicine, University of South Florida, Tampa, FL, USA
| | - Saame Raza Shaikh
- Department of Nutrition, Gillings School of Global Public Health and School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | | | - Charles N Serhan
- Center for Experimental Therapeutics and Reperfusion Injury, Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Ganesh V Halade
- Division of Cardiovascular Sciences, Department of Medicine, University of South Florida, Tampa, FL, USA
| |
Collapse
|
33
|
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.
Collapse
Affiliation(s)
- Amanda B Pullen
- Division of Cardiovascular Sciences Department of Medicine University of South Florida Tampa FL
| | - Vasundhara Kain
- Division of Cardiovascular Sciences Department of Medicine University of South Florida Tampa FL
| | - Charles N Serhan
- Center for Experimental Therapeutics and Reperfusion Injury Department of Anesthesiology, Perioperative and Pain Medicine Brigham and Women's Hospital Harvard Medical School, Boston MA
| | - Ganesh V Halade
- Division of Cardiovascular Sciences Department of Medicine University of South Florida Tampa FL
| |
Collapse
|
34
|
Mouton AJ, Li X, Hall ME, Hall JE. Obesity, Hypertension, and Cardiac Dysfunction: Novel Roles of Immunometabolism in Macrophage Activation and Inflammation. Circ Res 2020; 126:789-806. [PMID: 32163341 PMCID: PMC7255054 DOI: 10.1161/circresaha.119.312321] [Citation(s) in RCA: 352] [Impact Index Per Article: 70.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Obesity and hypertension, which often coexist, are major risk factors for heart failure and are characterized by chronic, low-grade inflammation, which promotes adverse cardiac remodeling. While macrophages play a key role in cardiac remodeling, dysregulation of macrophage polarization between the proinflammatory M1 and anti-inflammatory M2 phenotypes promotes excessive inflammation and cardiac injury. Metabolic shifting between glycolysis and mitochondrial oxidative phosphorylation has been implicated in macrophage polarization. M1 macrophages primarily rely on glycolysis, whereas M2 macrophages rely on the tricarboxylic acid cycle and oxidative phosphorylation; thus, factors that affect macrophage metabolism may disrupt M1/M2 homeostasis and exacerbate inflammation. The mechanisms by which obesity and hypertension may synergistically induce macrophage metabolic dysfunction, particularly during cardiac remodeling, are not fully understood. We propose that obesity and hypertension induce M1 macrophage polarization via mechanisms that directly target macrophage metabolism, including changes in circulating glucose and fatty acid substrates, lipotoxicity, and tissue hypoxia. We discuss canonical and novel proinflammatory roles of macrophages during obesity-hypertension-induced cardiac injury, including diastolic dysfunction and impaired calcium handling. Finally, we discuss the current status of potential therapies to target macrophage metabolism during heart failure, including antidiabetic therapies, anti-inflammatory therapies, and novel immunometabolic agents.
Collapse
Affiliation(s)
- Alan J. Mouton
- Department of Physiology and Biophysics, University of Mississippi Medical Center, 2500 North State Street; Jackson, MS, 39216-4505
- Mississippi Center for Obesity Research, University of Mississippi Medical Center, 2500 North State Street; Jackson, MS, 39216-4505
| | - Xuan Li
- Department of Physiology and Biophysics, University of Mississippi Medical Center, 2500 North State Street; Jackson, MS, 39216-4505
- Mississippi Center for Obesity Research, University of Mississippi Medical Center, 2500 North State Street; Jackson, MS, 39216-4505
| | - Michael E. Hall
- Department of Physiology and Biophysics, University of Mississippi Medical Center, 2500 North State Street; Jackson, MS, 39216-4505
- Department of Medicine, University of Mississippi Medical Center, 2500 North State Street; Jackson, MS, 39216-4505
- Mississippi Center for Obesity Research, University of Mississippi Medical Center, 2500 North State Street; Jackson, MS, 39216-4505
| | - John E. Hall
- Department of Physiology and Biophysics, University of Mississippi Medical Center, 2500 North State Street; Jackson, MS, 39216-4505
- Mississippi Center for Obesity Research, University of Mississippi Medical Center, 2500 North State Street; Jackson, MS, 39216-4505
| |
Collapse
|
35
|
Pullen AB, Jadapalli JK, Rhourri-Frih B, Halade GV. Re-evaluating the causes and consequences of non-resolving inflammation in chronic cardiovascular disease. Heart Fail Rev 2020; 25:381-391. [PMID: 31201605 PMCID: PMC6911017 DOI: 10.1007/s10741-019-09817-x] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
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.
Collapse
Affiliation(s)
- Amanda B Pullen
- Department of Medicine, Division of Cardiovascular Disease, The University of Alabama at Birmingham, Birmingham, AL, USA
| | - Jeevan Kumar Jadapalli
- Department of Medicine, Division of Cardiovascular Disease, The University of Alabama at Birmingham, Birmingham, AL, USA
| | - Boutayna Rhourri-Frih
- Chimie et Biologie des Membranes et Nanoobjets, University of Bordeaux, CNRS UMR 5248, 146, rue Léo Saignat, 33076, Bordeaux, France
| | - Ganesh V Halade
- Department of Medicine, Division of Cardiovascular Disease, The University of Alabama at Birmingham, Birmingham, AL, USA.
| |
Collapse
|
36
|
Costa-Santos K, Damasceno K, Portela RD, Santos FL, Araújo GC, Martins-Filho EF, Silva LP, Barral TD, Santos SA, Estrela-Lima A. Lipid and metabolic profiles in female dogs with mammary carcinoma receiving dietary fish oil supplementation. BMC Vet Res 2019; 15:401. [PMID: 31703601 PMCID: PMC6839264 DOI: 10.1186/s12917-019-2151-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2019] [Accepted: 10/22/2019] [Indexed: 02/08/2023] Open
Abstract
Background Dyslipidemias induce angiogenesis and accelerate the development and in vitro growth of breast tumors. The aim of this study was to assess the lipid and metabolic profile of female dogs with mammary carcinomas and their correlations with body condition score and degree of tumor malignancy, as well as to study the effect of dietary fish oil supplementation on these animals. Results Overweight or obese dogs had more aggressive carcinomas and higher triglyceride (p = 0.0363), VLDL (p = 0.0181), albumin (p = 0.0188), globulin (p = 0.0145) and lactate (p = 0.0255) concentrations. There was no change in the lipid profile after supplementation with fish oil at any concentration. However, in relation to the metabolic profile, glucose (p = 0.0067), total protein (p = 0.0002) and globulin (p = 0.0002) concentrations were increased when 90% omega-3 fish oil was used as a dietary supplement. Conclusion Obese dogs showed altered lipid and metabolic profiles and more aggressive tumors, suggesting an important relationship between dyslipidemia and tumor aggressiveness. Supplementation with fish oil, rich in omega-3 fatty acids, may alter metabolic parameters in cancer patients.
Collapse
Affiliation(s)
- Keidylania Costa-Santos
- Escola de Medicina Veterinária e Zootecnia, Universidade Federal da Bahia, Salvador, Bahia, 40170-110, Brazil.,Instituto Federal de Educação, Ciência e Tecnologia do Sertão Pernambucano, Santa Maria da Boa Vista, Pernambuco, 56380-000, Brazil
| | - Karine Damasceno
- Instituto Gonçalo Moniz, Fundação Oswaldo Cruz, Salvador, Bahia, Brazil
| | - Ricardo Dias Portela
- Instituto de Ciências da Saúde, Universidade Federal da Bahia, Salvador, Bahia, 40110-100, Brazil
| | - Ferlando Lima Santos
- Universidade Federal do Recôncavo da Bahia, Santo Antônio de Jesus, Bahia, 44570-000, Brazil
| | | | | | - Laís Pereira Silva
- Escola de Medicina Veterinária e Zootecnia, Universidade Federal da Bahia, Salvador, Bahia, 40170-110, Brazil
| | - Thiago Doria Barral
- Instituto de Ciências da Saúde, Universidade Federal da Bahia, Salvador, Bahia, 40110-100, Brazil
| | - Stefanie Alvarenga Santos
- Escola de Medicina Veterinária e Zootecnia, Universidade Federal da Bahia, Salvador, Bahia, 40170-110, Brazil
| | - Alessandra Estrela-Lima
- Escola de Medicina Veterinária e Zootecnia, Universidade Federal da Bahia, Salvador, Bahia, 40170-110, Brazil.
| |
Collapse
|
37
|
Navarro-Hortal MD, Ramírez-Tortosa CL, Varela-López A, Romero-Márquez JM, Ochoa JJ, Ramírez-Tortosa MC, Forbes-Hernández TY, Granados-Principal S, Battino M, Quiles JL. Heart Histopathology and Mitochondrial Ultrastructure in Aged Rats Fed for 24 Months on Different Unsaturated Fats (Virgin Olive Oil, Sunflower Oil or Fish Oil) and Affected by Different Longevity. Nutrients 2019; 11:E2390. [PMID: 31591312 PMCID: PMC6835383 DOI: 10.3390/nu11102390] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2019] [Revised: 09/27/2019] [Accepted: 10/01/2019] [Indexed: 12/18/2022] Open
Abstract
Diet plays a decisive role in heart physiology, with lipids having especial importance in pathology prevention and development. This study aimed to investigate how dietary lipids varying in lipid profile (virgin olive oil, sunflower oil or fish oil) affected the heart of rats during aging. Heart histopathology, mitochondrial morphometry, and oxidative status were assessed. Typical histopathological features associated with aging, such as valvular lesions, endomyocardical hyperplasia, or papillary muscle calcification, were found at a low extent in all the experimental groups. The most relevant finding was that inflammation registered by fish oil group was lower compared to the other treatments. At the ultrastructural level, heart mitochondrial area, perimeter, and aspect ratio were higher in fish oil-fed rats than in those fed on sunflower oil. Concerning oxidative stress markers, there were differences only in coenzyme Q levels and catalase activity, lower in sunflower oil-fed animals compared with those fed on fish oil. In summary, dietary intake for a long period on dietary fats with different fatty acids profile led to differences in some aspects associated with the aging process at the heart. Fish oil seems to be the fat most protective of heart during aging.
Collapse
Affiliation(s)
- María D Navarro-Hortal
- Department of Physiology, Institute of Nutrition and Food Technology "José Mataix Verdú", Biomedical Research Center, University of Granada, Avda del Conocimiento sn., 18100 Armilla, Granada, Spain.
| | - César L Ramírez-Tortosa
- UGC de Anatomía Patológica, Hospital San Cecilio de Granada, Avda, Conocimiento s/n, 18100 Granada, Spain.
| | - Alfonso Varela-López
- Department of Physiology, Institute of Nutrition and Food Technology "José Mataix Verdú", Biomedical Research Center, University of Granada, Avda del Conocimiento sn., 18100 Armilla, Granada, Spain.
| | - José M Romero-Márquez
- Department of Physiology, Institute of Nutrition and Food Technology "José Mataix Verdú", Biomedical Research Center, University of Granada, Avda del Conocimiento sn., 18100 Armilla, Granada, Spain.
| | - Julio J Ochoa
- Department of Physiology, Institute of Nutrition and Food Technology "José Mataix Verdú", Biomedical Research Center, University of Granada, Avda del Conocimiento sn., 18100 Armilla, Granada, Spain.
| | - MCarmen Ramírez-Tortosa
- Department of Biochemistry and Molecular Biology II, Institute of Nutrition and Food Technology "José Mataix Verdú", Biomedical Research Center, University of Granada, Avda del Conocimiento sn., 18100 Armilla, Granada, Spain.
| | - Tamara Y Forbes-Hernández
- Nutrition and Food Science Group, Department of Analytical and Food Chemistry, CITACA, CACTI, University of Vigo, 36310 Vigo, Spain.
| | - Sergio Granados-Principal
- UGC de Oncología Médica, Hospital Universitario de Jaén, Avenida del Ejército Español 10, 23007 Jaén, Spain.
- Genyo, Centre for Genomics and Oncological Research, Pfizer/University of Granada/Andalusian Regional Government, PTS Granada-Avenida de la Ilustración 114, 18016 Granada, Spain.
| | - Maurizio Battino
- Nutrition and Food Science Group, Department of Analytical and Food Chemistry, CITACA, CACTI, University of Vigo, 36310 Vigo, Spain.
- Dipartimento di Scienze Cliniche Specialistiche ed Odontostomatologiche-Sez. Biochimica, Università Politecnica delle Marche, Ancona, 60131 Ancona, Italy.
- International Research Center for Food Nutrition and Safety, Jiangsu University, 212013 Zhenjiang, China.
| | - José L Quiles
- Department of Physiology, Institute of Nutrition and Food Technology "José Mataix Verdú", Biomedical Research Center, University of Granada, Avda del Conocimiento sn., 18100 Armilla, Granada, Spain.
| |
Collapse
|
38
|
Halade GV, Kain V, Tourki B, Jadapalli JK. Lipoxygenase drives lipidomic and metabolic reprogramming in ischemic heart failure. Metabolism 2019; 96:22-32. [PMID: 30999004 DOI: 10.1016/j.metabol.2019.04.011] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/23/2019] [Revised: 04/02/2019] [Accepted: 04/11/2019] [Indexed: 12/31/2022]
Abstract
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.
Collapse
Affiliation(s)
- Ganesh V Halade
- Division of Cardiovascular Disease, Department of Medicine, The University of Alabama at Birmingham, AL, United States.
| | - Vasundhara Kain
- Division of Cardiovascular Disease, Department of Medicine, The University of Alabama at Birmingham, AL, United States
| | - Bochra Tourki
- Division of Cardiovascular Disease, Department of Medicine, The University of Alabama at Birmingham, AL, United States
| | - Jeevan Kumar Jadapalli
- Division of Cardiovascular Disease, Department of Medicine, The University of Alabama at Birmingham, AL, United States
| |
Collapse
|
39
|
Kain V, Van Der Pol W, Mariappan N, Ahmad A, Eipers P, Gibson DL, Gladine C, Vigor C, Durand T, Morrow C, Halade GV. Obesogenic diet in aging mice disrupts gut microbe composition and alters neutrophil:lymphocyte ratio, leading to inflamed milieu in acute heart failure. FASEB J 2019; 33:6456-6469. [PMID: 30768364 PMCID: PMC6463911 DOI: 10.1096/fj.201802477r] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2018] [Accepted: 01/22/2019] [Indexed: 12/16/2022]
Abstract
Calorie-dense obesogenic diet (OBD) is a prime risk factor for cardiovascular disease in aging. However, increasing age coupled with changes in the diet can affect the interaction of intestinal microbiota influencing the immune system, which can lead to chronic inflammation. How age and calorie-enriched OBD interact with microbial flora and impact leukocyte profiling is currently under investigated. Here, we tested the interorgan hypothesis to determine whether OBD in young and aging mice alters the gut microbe composition and the splenic leukocyte profile in acute heart failure (HF). Young (2-mo-old) and aging (18-mo-old) mice were supplemented with standard diet (STD, ∼4% safflower oil diet) and OBD (10% safflower oil) for 2 mo and then subjected to coronary artery ligation to induce myocardial infarction. Fecal samples were collected pre- and post-diet intervention, and the microbial flora were analyzed using 16S variable region 4 rRNA gene DNA sequencing and Quantitative Insights Into Microbial Ecology informatics. The STD and OBD in aging mice resulted in an expansion of the genus Allobaculum in the fecal microbiota. However, we found a pathologic change in the neutrophil:lymphocyte ratio in aging mice in comparison with their young counterparts. Thus, calorie-enriched OBD dysregulated splenic leukocytes by decreasing immune-responsive F4/80+ and CD169+ macrophages in aging mice. OBD programmed neutrophil swarming with an increase in isoprostanoid levels, with dysregulation of lipoxygenases, cytokines, and metabolite-sensing receptor expression. In summary, calorie-dense OBD in aging mice disrupted the composition of the gut microbiome, which correlates with the development of integrative and system-wide nonresolving inflammation in acute HF.-Kain, V., Van Der Pol, W., Mariappan, N., Ahmad, A., Eipers, P., Gibson, D. L., Gladine, C., Vigor, C., Durand, T., Morrow, C., Halade, G. V. Obesogenic diet in aging mice disrupts gut microbe composition and alters neutrophil:lymphocyte ratio, leading to inflamed milieu in acute heart failure.
Collapse
Affiliation(s)
- Vasundhara Kain
- Division of Cardiovascular Disease, Department of Medicine, The University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - William Van Der Pol
- Biomedical Informatics, Center for Clinical and Translational Sciences, The University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Nithya Mariappan
- Department of Anesthesiology and Perioperative Medicine, The University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Aftab Ahmad
- Department of Anesthesiology and Perioperative Medicine, The University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Peter Eipers
- Department of Cell, Developmental, and Integrative Biology, The University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Deanna L. Gibson
- Department of Biology, University of British Columbia Okanagan, Kelowna, British Columbia, Canada
| | - Cecile Gladine
- Unité de Nutrition Humaine (UNH), Institut National de la Recherche Agronomique (INRA), Centre de Recherche en Nutrition Humaine (CRNH) Auvergne, Université Clermont Auvergne, Clermont-Ferrand, France
| | - Claire Vigor
- Unité Mixte de Recherche (UMR) 247, Institut des Biomolécules Max Mousseron (IBMM), Centre National de la Recherche Scientifique (CNRS), Ecole Nationale Supérieure de Chimie de Montpellier (ENSCM), University of Montpellier, Montpellier, France
| | - Thierry Durand
- Unité Mixte de Recherche (UMR) 247, Institut des Biomolécules Max Mousseron (IBMM), Centre National de la Recherche Scientifique (CNRS), Ecole Nationale Supérieure de Chimie de Montpellier (ENSCM), University of Montpellier, Montpellier, France
| | - Casey Morrow
- Department of Cell, Developmental, and Integrative Biology, The University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Ganesh V. Halade
- Division of Cardiovascular Disease, Department of Medicine, The University of Alabama at Birmingham, Birmingham, Alabama, USA
| |
Collapse
|
40
|
Cardiovascular Risks Associated with Gender and Aging. J Cardiovasc Dev Dis 2019; 6:jcdd6020019. [PMID: 31035613 PMCID: PMC6616540 DOI: 10.3390/jcdd6020019] [Citation(s) in RCA: 496] [Impact Index Per Article: 82.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2019] [Revised: 04/19/2019] [Accepted: 04/23/2019] [Indexed: 12/12/2022] Open
Abstract
The aging and elderly population are particularly susceptible to cardiovascular disease. Age is an independent risk factor for cardiovascular disease (CVD) in adults, but these risks are compounded by additional factors, including frailty, obesity, and diabetes. These factors are known to complicate and enhance cardiac risk factors that are associated with the onset of advanced age. Sex is another potential risk factor in aging adults, given that older females are reported to be at a greater risk for CVD than age-matched men. However, in both men and women, the risks associated with CVD increase with age, and these correspond to an overall decline in sex hormones, primarily of estrogen and testosterone. Despite this, hormone replacement therapies are largely shown to not improve outcomes in older patients and may also increase the risks of cardiac events in older adults. This review discusses current findings regarding the impacts of age and gender on heart disease.
Collapse
|
41
|
Tippens KM, Erlandsen A, Hanes DA, Graybill R, Jackson C, Briley J, Zwickey H. Impact of a Short-Term Naturopathic Whole-Foods-Based Nutrition Education Intervention on Dietary Behavior and Diabetes Risk Markers: A Pilot Study. J Altern Complement Med 2018; 25:234-240. [PMID: 30312107 DOI: 10.1089/acm.2018.0025] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/28/2022] Open
Abstract
OBJECTIVE To assess the feasibility of measuring the health impacts of a 12-week naturopathic whole foods nutrition education course among adults with or at risk for prediabetes. DESIGN A pilot nonrandomized pre-post intervention design investigated physiological changes associated with participation in a 12-week structured naturopathic nutrition education course. Follow-up measures were assessed at 6 and 12 months from baseline. SETTINGS/LOCATION Three community-based kitchens in Oregon. SUBJECTS Forty-five adults with or at risk for prediabetes. INTERVENTION Twelve weekly 90-min workshops emphasizing the health benefits of a naturopathic whole foods diet and including collective meal preparation and communal dining. OUTCOME MEASURES Changes in biomarkers of diabetes and cardiovascular disease risk, including high-sensitivity C-reactive protein (hs-CRP), hemoglobin A1c, insulin, and lipids, and changes in dietary behaviors at baseline, postintervention (12 weeks), and 6- and 12-month follow-up. Changes in biomarkers and food consumption patterns were assessed using linear mixed models with random intercept, including data from all participants who completed the end-of-intervention assessment. RESULTS Pre-post intervention results for 45 participants showed decreases in blood glucose and hs-CRP. Food frequency questionnaire data showed decreases in daily servings of grain, dairy, and fat postintervention. Decreases in blood glucose, hs-CRP, triglycerides, cholesterol, low-density lipoprotein, very-low-density lipoprotein, and HbA1c from baseline were observed at 12-month follow-up. Both insulin and high-density lipoprotein were decreased from baseline and 12-week levels at the 6-month follow-up but increased from all earlier levels at 12 months (all p < 0.0001). Daily servings of meat, dairy, and fat remained decreased at 6-month follow-up. Reduced grain consumption observed at 12-week and at 6-month follow-up was not sustained, and increased at 12 months, although still decreased from baseline. CONCLUSIONS Results suggest that this naturopathic nutrition education series may promote dietary behavior change with associated changes in clinical biomarkers. Larger controlled studies are needed to confirm these results.
Collapse
Affiliation(s)
- Kimberly M Tippens
- 1 National University of Natural Medicine, Portland, OR.,2 Helfgott Research Institute, Portland, OR
| | - Andrew Erlandsen
- 1 National University of Natural Medicine, Portland, OR.,2 Helfgott Research Institute, Portland, OR.,3 Food As Medicine Institute, Portland, OR
| | - Douglas A Hanes
- 1 National University of Natural Medicine, Portland, OR.,2 Helfgott Research Institute, Portland, OR
| | - Robert Graybill
- 1 National University of Natural Medicine, Portland, OR.,2 Helfgott Research Institute, Portland, OR
| | - Courtney Jackson
- 1 National University of Natural Medicine, Portland, OR.,3 Food As Medicine Institute, Portland, OR
| | - Julie Briley
- 1 National University of Natural Medicine, Portland, OR.,3 Food As Medicine Institute, Portland, OR
| | - Heather Zwickey
- 1 National University of Natural Medicine, Portland, OR.,2 Helfgott Research Institute, Portland, OR
| |
Collapse
|
42
|
Doyle R, Sadlier DM, Godson C. Pro-resolving lipid mediators: Agents of anti-ageing? Semin Immunol 2018; 40:36-48. [PMID: 30293857 DOI: 10.1016/j.smim.2018.09.002] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/22/2018] [Revised: 09/14/2018] [Accepted: 09/24/2018] [Indexed: 12/11/2022]
Abstract
Inflammation is an essential response to injury and its timely and adequate resolution permits tissue repair and avoidance of chronic inflammation. Ageing is associated with increased inflammation, sub-optimal resolution and these act as drivers for a number of ageing-associated pathologies. We describe the role played by specialised proresolving lipid mediators (SPMs) in the resolution of inflammation and how insufficient levels of these mediators, or compromised responsiveness may play a role in the pathogenesis of many ageing-associated pathologies, e.g. Alzheimer's Disease, atherosclerosis, obesity, diabetes and kidney disease. Detailed examination of the resolution phase of inflammation highlights the potential to harness these lipid mediators and or mimetics of their bioactions, in particular, their synthetic analogues to promote effective resolution of inflammation, without compromising the host immune system.
Collapse
Affiliation(s)
- Ross Doyle
- School of Medicine, University College Dublin, Belfield, Dublin 4, Ireland; Mater Misericordiae University Hospital, Eccles St., Inns Quay, Dublin 7, Ireland.
| | - Denise M Sadlier
- School of Medicine, University College Dublin, Belfield, Dublin 4, Ireland; Mater Misericordiae University Hospital, Eccles St., Inns Quay, Dublin 7, Ireland
| | - Catherine Godson
- School of Medicine, University College Dublin, Belfield, Dublin 4, Ireland
| |
Collapse
|
43
|
Kain V, Halade GV. Immune responsive resolvin D1 programs peritoneal macrophages and cardiac fibroblast phenotypes in diversified metabolic microenvironment. J Cell Physiol 2018; 234:3910-3920. [PMID: 30191990 DOI: 10.1002/jcp.27165] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2017] [Accepted: 07/11/2018] [Indexed: 12/20/2022]
Abstract
Bioactive lipid mediators derived from n-3 and n-6 fatty acids are known to modulate leukocytes. Metabolic transformation of essential fatty acids to endogenous bioactive molecules plays a major role in human health. Here we tested the potential of substrates; linoleic acid (LA) and docosahexaenoic acid (DHA) and their bioactive products; resolvin D1 (RvD1) and 12- S-hydroxyeicosatetraenoic acids (HETE) to modulate macrophage plasticity and cardiac fibroblast phenotype in presence or absence of lipid metabolizing enzyme 12/15-lipoxygenase (LOX). Peritoneal macrophages and cardiac fibroblasts were isolated from wild-type (C57BL/6J) and 12/15LOX -/- mice and treated with DHA, LA, 12(S)-HETE, and RvD1 for 4, 8, 12, and 24 hr. LA, DHA, 12(S)-HETE, and RvD1 elicited mRNA expression of proinflammatory markers; tumor necrosis factor-α ( Tnf-α), interleukin 6 ( IL-6), chemokine (C-C motif) ligand 2 (Ccl2), and IL-1β in wild type (WT) and in 12/15LOX -/- macrophages at early time point (4 hr). Bioactive immunoresolvent RvD1 lowered the levels of Tnf-α, IL-6, and IL-1β at 24 hr time point. Both DHA and RvD1 stimulated the proresolving markers such as arginase 1 ( Arg-1), chitinase-like protein 3 ( Ym-1), and mannose receptor C-type 1 in WT macrophage. RvD1 induced proresolving phenotype Arg-1 expression in both WT 12/15LOX -/- macrophages even in presence of 12(S)-HETE. RvD1 peaked 5LOX expression in both WT and 12/15LOX -/- at 24 hr time point compared with DHA. RvD1 diminished cyclooxygenase-2 but upregulated 5LOX expression in fibroblast compared with DHA. In summary, the feed-forward enzymatic interaction with fatty acids substrates and direct mediators (RvD1 and 12(S)-HETE) are responsive in determining macrophages phenotype and cardiac fibroblast plasticity. Particularly, macrophages and fibroblast phenotypes are responsive to milieu and RvD1 governs the milieu-dependent chemokine signaling in presence or absence of 12/15LOX enzyme to resolve inflammation.
Collapse
Affiliation(s)
- Vasundhara Kain
- Division of Cardiovascular Disease, Department of Medicine, The University of Alabama at Birmingham, Birmingham, Alabama
| | - Ganesh V Halade
- Division of Cardiovascular Disease, Department of Medicine, The University of Alabama at Birmingham, Birmingham, Alabama
| |
Collapse
|
44
|
Sanches JM, Giraldo PC, Amaral R, Eberlin MN, Marques LA, Migliorini I, Nakahira M, Bieleveld MJM, Discacciati MG. Vaginal lipidomics of women with vulvovaginal candidiasis and cytolytic vaginosis: A non-targeted LC-MS pilot study. PLoS One 2018; 13:e0202401. [PMID: 30133508 PMCID: PMC6105002 DOI: 10.1371/journal.pone.0202401] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2017] [Accepted: 08/02/2018] [Indexed: 02/06/2023] Open
Abstract
Objective To characterize the lipid profile in vaginal discharge of women with vulvovaginal candidiasis, cytolytic vaginosis, or no vaginal infection or dysbiosis. Design Cross-sectional study. Setting Genital Infections Ambulatory, Department of Tocogynecology, University of Campinas, Campinas, São Paulo–Brazil. Sample Twenty-four women were included in this study: eight with vulvovaginal candidiasis, eight with cytolytic vaginosis and eight with no vaginal infections or dysbiosis (control group). Methods The lipid profile in vaginal discharge of the different study groups was determined by liquid chromatography-mass spectrometry and further analyzed with MetaboAnalyst 3.0 platform. Main outcome measures Vaginal lipids concentration and its correlation with vulvovaginal candidiasis and cytolytic vaginosis. Results PCA, PLS-DA and hierarchical clustering analyses indicated 38 potential lipid biomarkers for the different groups, correlating with oxidative stress, inflammation, apoptosis and integrity of the vaginal epithelial tissue. Among these, greater concentrations were found for Glycochenodeoxycholic acid-7-sulfate, O-adipoylcarnitine, 1-eicosyl-2-heptadecanoyl-glycero-3-phosphoserine, undecanoic acid, formyl dodecanoate and lipoic acid in the vulvovaginal candidiasis group; N–(tetradecanoyl)-sphinganine, DL-PPMP, 1-oleoyl-cyclic phosphatidic, palmitic acid and 5-aminopentanoic acid in the cytolytic vaginosis group; and 1-nonadecanoyl-glycero-3-phosphate, eicosadienoic acid, 1-stearoyl-cyclic-phosphatidic acid, 1-(9Z,12Z-heptadecadienoyl)-glycero-3-phosphate, formyl 9Z-tetradecenoate and 7Z,10Z-hexadecadienoic acid in the control group. Conclusions Lipids related to oxidative stress and apoptosis were found in higher concentrations in women with vulvovaginal candidiasis and cytolytic vaginosis, while lipids related to epithelial tissue integrity were more pronounced in the control group. Furthermore, in women with cytolytic vaginosis, we observed higher concentrations of lipids related to bacterial overgrowth.
Collapse
Affiliation(s)
- José Marcos Sanches
- Campinas State University, Department of Tocoginecology, Campinas, São Paulo, Brazil
- * E-mail:
| | - Paulo César Giraldo
- Campinas State University, Department of Tocoginecology, Campinas, São Paulo, Brazil
| | - Rose Amaral
- Campinas State University, Department of Tocoginecology, Campinas, São Paulo, Brazil
| | | | | | - Isabel Migliorini
- Campinas State University, Department of Tocoginecology, Campinas, São Paulo, Brazil
| | - Marcel Nakahira
- Campinas State University, Institute of Chemistry, Campinas, São Paulo, Brazil
| | | | | |
Collapse
|
45
|
Ruggiero SA, Huber JS, Murrant CL, Brunt KR, Simpson JA. Splenic blood-flow response following myocardial infarction in rat. Can J Physiol Pharmacol 2018; 96:1060-1068. [PMID: 30102865 DOI: 10.1139/cjpp-2018-0134] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
During physiological stress (e.g., exercise, hypoxia), blood flow is shunted to specific anatomical regions to protect critical organs; yet, splenic blood flow in these circumstances remains to be investigated. Despite being classically viewed as a non-critical organ, recent experimental and epidemiological evidence suggests the spleen plays a significant role in cardiovascular pathophysiology. We hypothesized that splenic blood flow is prioritized in the development of heart failure (i.e., chronic state of reduced cardiac output). Five-week-old male Wistar rats were randomized for either myocardial infarction (MI; n = 58) or sham (n = 56) surgery. At 2, 5, and 9 weeks post-surgery, Doppler ultrasound measurements of the splenic, left renal, left common carotid, and left femoral arteries were performed. Cardiac function was assessed at all time points using echocardiography and at 9 weeks post-surgery using invasive hemodynamic analysis. Splenic and cerebral blood flow was preferentially maintained at 9 weeks post-MI, whereas blood flow to the lower limb and kidney were reduced. Spleen size increased by 5 weeks post-MI and remained elevated. Splenic blood flow was maintained in conditions of decreased cardiac output, when other tissues showed decreased blood flow. The maintenance of blood flow in the face of decreased cardiac output indicates that splenic function is being prioritized during heart failure.
Collapse
Affiliation(s)
- Sara A Ruggiero
- a Department of Human Health and Nutritional Science, College of Biological Sciences, University of Guelph, Guelph, ON N1G 2W1, Canada
| | - Jason S Huber
- a Department of Human Health and Nutritional Science, College of Biological Sciences, University of Guelph, Guelph, ON N1G 2W1, Canada.,b IMPART team Canada Investigator Network
| | - Coral L Murrant
- a Department of Human Health and Nutritional Science, College of Biological Sciences, University of Guelph, Guelph, ON N1G 2W1, Canada
| | - Keith R Brunt
- b IMPART team Canada Investigator Network.,c Department of Pharmacology, Dalhousie Medicine New Brunswick, Dalhousie University, Saint John, NB E2K 5E2, Canada
| | - Jeremy A Simpson
- a Department of Human Health and Nutritional Science, College of Biological Sciences, University of Guelph, Guelph, ON N1G 2W1, Canada.,b IMPART team Canada Investigator Network
| |
Collapse
|
46
|
Ahn SH, Lim SJ, Ryu YM, Park HR, Suh HJ, Han SH. Absorption rate of krill oil and fish oil in blood and brain of rats. Lipids Health Dis 2018; 17:162. [PMID: 30021606 PMCID: PMC6052518 DOI: 10.1186/s12944-018-0812-7] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2018] [Accepted: 07/04/2018] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Krill (Euphausia superba) is a small marine crustacean with a lipid content. The mechanism of Krill oil function is not clear yet and research reports on the absorption rate of the phospholipids of krill oil in the blood and brain are very poor. METHODS We studied the effect of oral short-term and long-term administration of Krill oils (KOs) on bioavailability in the blood and brain of rats. For short-term testing of fish and KO bioavailability, rats were divided into four groups: normal, fish oil (FO), Krill oil 1 (KO), and Krill oil 2 (CKO). The blood and brain were collected at 2, 4, 8, 12, 24, and 48 h after oral administration (1000 mg/rat). Five hundred milligrams of FO, KO, and CKO were orally administered daily for 2 weeks for long-term administration, and then the brain and blood were collected. RESULTS Two types of KOs showed high content of eicosapentaenoic acid (EPA) and docosahexaenoic acid (DHA) in the PL. The EPA content of CKO and KO were 41.13 and 32.49%, respectively. After short-term KO administration, KO showed a higher EPA content than CKO in the blood after 2 h. KO showed higher content of DHA than CKO even after 2 h. FO increased until 8 h, but then decreased rapidly until 12 h. Although the total unsaturated fatty acid (UFA) content of KOs was lower than the total UFS content in FO, the remaining UFS content in the brain was higher than that in FO over time. Following oral administration of FO, KO, and CKO for 1 and 2 weeks, triglycerides (TG) and PL contents in the blood for KOs were slightly higher than for FO. EPA and DHA levels in the brain were slightly higher in KOs following long-term administration, but the difference was not significant. CONCLUSIONS Base on these findings, KOs have functional potential for the brain and vascular diseases, and can be utilized as a multi-functional material composed mainly of functional ingredients.
Collapse
Affiliation(s)
- So Hyun Ahn
- Department of Food and Nutrition, Korea University, Seoul, 07249, Republic of Korea
| | - Su Jin Lim
- Alpha B&H, Seoul, 06705, Republic of Korea
| | | | - Hye-Ryung Park
- Department of Food Science and Biotechnology, Kyonggi University, Suwon, 16227, Republic of Korea
| | - Hyung Joo Suh
- BK21Plus, College of Health Science, Korea University, Seoul, 02841, Republic of Korea
| | - Sung Hee Han
- BK21Plus, College of Health Science, Korea University, Seoul, 02841, Republic of Korea.
| |
Collapse
|