1
|
Wiggers H. Fatty acids in heart failure patients: friend or foe? Eur Heart J 2025:ehaf247. [PMID: 40277465 DOI: 10.1093/eurheartj/ehaf247] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 04/26/2025] Open
Affiliation(s)
- Henrik Wiggers
- Department of Cardiology, Aarhus University Hospital, Aarhus, Denmark
| |
Collapse
|
2
|
Tufail T, Bader Ul Ain H, Ashraf J, Mahmood S, Noreen S, Ijaz A, ikram A, Arshad MT, Abdullahi MA. Bioactive Compounds in Seafood: Implications for Health and Nutrition. Food Sci Nutr 2025; 13:e70181. [PMID: 40260061 PMCID: PMC12009756 DOI: 10.1002/fsn3.70181] [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: 09/22/2024] [Revised: 12/28/2024] [Accepted: 04/04/2025] [Indexed: 04/23/2025] Open
Abstract
The significance of Seafood as a reservoir of bioactive substances is increasing. With sea creatures making up approximately half of all living organisms on the planet, seas, and oceans present many innovative materials and are believed to hold the most substantial remaining reserve of beneficial natural compounds. Seafood provides a plentiful supply of essential nutrients, including high-quality protein, various fatty acids (such as omega-3s), and bioactive compounds like taurine, carotenoids, and phytosterols, all contributing to its numerous health advantages. Furthermore, seafood contains bio-lipopeptides, polysaccharides, and phenolic compounds, and it promotes health through its antioxidant and anti-inflammatory effects. Enzymes, vitamins, and minerals further enrich its nutritional profile, supporting various metabolic processes and overall well-being. This review emphasizes the health benefits of seafood consumption, encompassing its cardio-protective effects that bolster heart health, its antidiabetic properties that aid in regulating blood sugar levels and its anti-cancer effects that may lower the risk of specific cancers. Additionally, seafood contributes to anti-obesity effects, enhances brain health, delivers antioxidative activity to combat oxidative stress, and supports maternal care during pregnancy and lactation.
Collapse
Affiliation(s)
- Tabussam Tufail
- School of Food Science and EngineeringYangzhou UniversityYangzhouChina
- School of Food & Biological EngineeringJiangsu UniversityZhenjiangChina
| | - Huma Bader Ul Ain
- School of Food Science and EngineeringYangzhou UniversityYangzhouChina
- University Institute of Diet and Nutritional SciencesThe University of LahoreLahorePakistan
| | - Jawad Ashraf
- School of Food & Biological EngineeringJiangsu UniversityZhenjiangChina
| | - Sammina Mahmood
- Department of Botany, Division of Science and TechnologyUniversity of EducationLahorePakistan
| | - Sana Noreen
- University Institute of Diet and Nutritional SciencesThe University of LahoreLahorePakistan
| | - Aiman Ijaz
- University Institute of Diet and Nutritional SciencesThe University of LahoreLahorePakistan
| | - Ali ikram
- University Institute of Food Science and TechnologyThe University of LahoreLahorePakistan
| | - Muhammad Tayyab Arshad
- University Institute of Food Science and TechnologyThe University of LahoreLahorePakistan
| | - Muhammed Adem Abdullahi
- Department of Food Science and Postharvest TechnologyJimma University College of Agriculture and Veterinary Medicine, Jimma UniversityJimmaEthiopia
| |
Collapse
|
3
|
Kim DH, Lee MJ, Kang D, Lee JY, Park S, Khang AR, Bae JH, Kim JY, Kim SH, Kang YH, Yi D. Empagliflozin Attenuates High-Glucose-Induced Astrocyte Activation and Inflammation via NF-κB Pathway. Curr Issues Mol Biol 2024; 46:12417-12427. [PMID: 39590331 PMCID: PMC11592667 DOI: 10.3390/cimb46110737] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2024] [Revised: 10/28/2024] [Accepted: 10/31/2024] [Indexed: 11/28/2024] Open
Abstract
Sodium-glucose cotransporter-2 (SGLT2) inhibitors regulate blood glucose levels in patients with type 2 diabetes mellitus and may also exert anti-inflammatory and anti-atherosclerotic effects by promoting M2 macrophage polarization. Although SGLT2 is expressed in brain regions that influence glucose balance and cognitive function, its roles in the central nervous system are unclear. This study investigated the effects of empagliflozin (EMPA), an SGLT2 inhibitor, on hypothalamic inflammation associated with metabolic diseases. Mice were subjected to a high-fat diet (HFD) for varying durations (3 d, 3 weeks, and 16 weeks) and treated with EMPA for 3 weeks (NFD, NFD + EMPA, HFD, HFD + EMPA; n = 5/group). EMPA regulated the expression of astrocyte markers and pro-inflammatory cytokine mRNA in the hypothalamus of HFD-induced mice, which was linked to regulation of the NF-κB pathway. Under hyperglycemic conditions, EMPA may mitigate hypothalamic inflammation by modulating astrocyte activation via the NF-κB pathway. Our findings demonstrated that EMPA possesses therapeutic potential beyond merely lowering blood glucose levels, opening new avenues for addressing inflammation and providing neuroprotection in metabolic disease management.
Collapse
Affiliation(s)
- Dong Hee Kim
- Research Institute for Convergence of Biomedical Science and Technology, Pusan National University Yangsan Hospital, Yangsan 50621, Republic of Korea; (D.H.K.); (M.J.L.); (D.K.); (J.Y.L.); (S.P.); (A.R.K.); (J.H.B.); (J.Y.K.); (S.H.K.); (Y.H.K.)
| | - Min Jin Lee
- Research Institute for Convergence of Biomedical Science and Technology, Pusan National University Yangsan Hospital, Yangsan 50621, Republic of Korea; (D.H.K.); (M.J.L.); (D.K.); (J.Y.L.); (S.P.); (A.R.K.); (J.H.B.); (J.Y.K.); (S.H.K.); (Y.H.K.)
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Pusan National University Yangsan Hospital, Pusan National University School of Medicine, Yangsan 50621, Republic of Korea
| | - Dasol Kang
- Research Institute for Convergence of Biomedical Science and Technology, Pusan National University Yangsan Hospital, Yangsan 50621, Republic of Korea; (D.H.K.); (M.J.L.); (D.K.); (J.Y.L.); (S.P.); (A.R.K.); (J.H.B.); (J.Y.K.); (S.H.K.); (Y.H.K.)
| | - Ji Young Lee
- Research Institute for Convergence of Biomedical Science and Technology, Pusan National University Yangsan Hospital, Yangsan 50621, Republic of Korea; (D.H.K.); (M.J.L.); (D.K.); (J.Y.L.); (S.P.); (A.R.K.); (J.H.B.); (J.Y.K.); (S.H.K.); (Y.H.K.)
| | - Sujin Park
- Research Institute for Convergence of Biomedical Science and Technology, Pusan National University Yangsan Hospital, Yangsan 50621, Republic of Korea; (D.H.K.); (M.J.L.); (D.K.); (J.Y.L.); (S.P.); (A.R.K.); (J.H.B.); (J.Y.K.); (S.H.K.); (Y.H.K.)
| | - Ah Reum Khang
- Research Institute for Convergence of Biomedical Science and Technology, Pusan National University Yangsan Hospital, Yangsan 50621, Republic of Korea; (D.H.K.); (M.J.L.); (D.K.); (J.Y.L.); (S.P.); (A.R.K.); (J.H.B.); (J.Y.K.); (S.H.K.); (Y.H.K.)
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Pusan National University Yangsan Hospital, Pusan National University School of Medicine, Yangsan 50621, Republic of Korea
| | - Ji Hyun Bae
- Research Institute for Convergence of Biomedical Science and Technology, Pusan National University Yangsan Hospital, Yangsan 50621, Republic of Korea; (D.H.K.); (M.J.L.); (D.K.); (J.Y.L.); (S.P.); (A.R.K.); (J.H.B.); (J.Y.K.); (S.H.K.); (Y.H.K.)
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Pusan National University Yangsan Hospital, Pusan National University School of Medicine, Yangsan 50621, Republic of Korea
| | - Joo Yeon Kim
- Research Institute for Convergence of Biomedical Science and Technology, Pusan National University Yangsan Hospital, Yangsan 50621, Republic of Korea; (D.H.K.); (M.J.L.); (D.K.); (J.Y.L.); (S.P.); (A.R.K.); (J.H.B.); (J.Y.K.); (S.H.K.); (Y.H.K.)
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Pusan National University Yangsan Hospital, Pusan National University School of Medicine, Yangsan 50621, Republic of Korea
| | - Su Hyun Kim
- Research Institute for Convergence of Biomedical Science and Technology, Pusan National University Yangsan Hospital, Yangsan 50621, Republic of Korea; (D.H.K.); (M.J.L.); (D.K.); (J.Y.L.); (S.P.); (A.R.K.); (J.H.B.); (J.Y.K.); (S.H.K.); (Y.H.K.)
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Pusan National University Yangsan Hospital, Pusan National University School of Medicine, Yangsan 50621, Republic of Korea
| | - Yang Ho Kang
- Research Institute for Convergence of Biomedical Science and Technology, Pusan National University Yangsan Hospital, Yangsan 50621, Republic of Korea; (D.H.K.); (M.J.L.); (D.K.); (J.Y.L.); (S.P.); (A.R.K.); (J.H.B.); (J.Y.K.); (S.H.K.); (Y.H.K.)
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Pusan National University Yangsan Hospital, Pusan National University School of Medicine, Yangsan 50621, Republic of Korea
| | - Dongwon Yi
- Research Institute for Convergence of Biomedical Science and Technology, Pusan National University Yangsan Hospital, Yangsan 50621, Republic of Korea; (D.H.K.); (M.J.L.); (D.K.); (J.Y.L.); (S.P.); (A.R.K.); (J.H.B.); (J.Y.K.); (S.H.K.); (Y.H.K.)
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Pusan National University Yangsan Hospital, Pusan National University School of Medicine, Yangsan 50621, Republic of Korea
| |
Collapse
|
4
|
Tamir T, Nigussie J, Endawoke M. Bridging the gap between recommendation and reality: Improving dietary adherence of heart failure populations a cross-sectional study in Ethiopia. PLoS One 2024; 19:e0311663. [PMID: 39388426 PMCID: PMC11466410 DOI: 10.1371/journal.pone.0311663] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Accepted: 09/23/2024] [Indexed: 10/12/2024] Open
Abstract
BACKGROUND Heart failure (HF), a complex condition arising from impaired ventricular function, necessitates strict adherence to dietary recommendations for optimal patient management. However, information regarding adherence and its influencing factors remains limited. AIM This study aimed to assess dietary recommendation adherence and its associated factors among HF patients at Southern Ethiopia public hospitals. METHODS AND RESULTS A cross-sectional study involving 521 participants employed systematic random sampling. Data collection utilized pre-tested, interviewer-administered questionnaires and medical chart reviews. Data were entered and analyzed using Epi Data 3.1 and SPSS 20.0 software. Descriptive statistics were performed. Variables with p-values < 0.25 in binary logistic regression were included in multivariable logistic regression analyses. Statistical significance was set at p < 0.05 with a 95% confidence interval. Results are presented in text, tables, and figures. With a 97.4% response rate, adherence ranged from 20.3% (vegetables and fruits) to 60.3% (fat-free diet). Only 8.1% achieved good adherence across all parameters, with overall adherence at 33.4% (95% CI: 29-37). Multivariable analysis revealed that patients aged 41-60 years (AOR: 1.7), with a history of admission (AOR: 2.5), free from comorbidities (AOR: 0.58), and possessing a favorable attitude (AOR: 0.45) had statistically significant associations with good adherence. CONCLUSION Dietary adherence among HF patients remains a challenge. Healthcare providers, particularly those in chronic follow-up settings, should prioritize improving patient attitudes towards proper dietary practices. Tailored education programs targeting younger patients and those free from comorbidities should be implemented. Continuous monitoring, evaluation, and staff recognition for effective client counseling are crucial.
Collapse
Affiliation(s)
- Takla Tamir
- Department of Nursing, Dilla University College of Health Science and Medicine, Addis Ababa, Dilla, Ethiopia
| | - Jemberu Nigussie
- Department of Nursing, Dilla University College of Health Science and Medicine, Addis Ababa, Dilla, Ethiopia
| | - Migbaru Endawoke
- Department of Nursing, Dilla University College of Health Science and Medicine, Addis Ababa, Dilla, Ethiopia
| |
Collapse
|
5
|
Ataran A, Pompian A, Hajirezaei H, Lodhi R, Javaheri A. Fueling the Heart: What Are the Optimal Dietary Strategies in Heart Failure? Nutrients 2024; 16:3157. [PMID: 39339757 PMCID: PMC11434961 DOI: 10.3390/nu16183157] [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: 08/01/2024] [Revised: 09/10/2024] [Accepted: 09/15/2024] [Indexed: 09/30/2024] Open
Abstract
OBJECTIVES Heart failure (HF) is a global health concern with rising incidence and poor prognosis. While the essential role of nutritional and dietary strategies in HF patients is acknowledged in the existing scientific guidelines and clinical practice, there are no comprehensive nutritional recommendations for optimal dietary management of HF. METHODS In this review, we discuss results from recent studies on the obesity paradox and the effects of calorie restriction and weight loss, intermittent fasting, the Western diet, the Mediterranean diet, the ketogenic diet, and the DASH diet on HF progression. RESULTS Many of these strategies remain under clinical and basic investigation for their safety and efficacy, and there is considerable heterogeneity in the observed response, presumably because of heterogeneity in the pathogenesis of different types of HF. In addition, while specific aspects of cardiac metabolism, such as changes in ketone body utilization, might underlie the effects of certain dietary strategies on the heart, there is a critical divide between supplement strategies (i.e., with ketones) and dietary strategies that impact ketogenesis. CONCLUSION This review aims to highlight this gap by exploring emerging evidence supporting the importance of personalized dietary strategies in preventing progression and improving outcomes in the context of HF.
Collapse
Affiliation(s)
- Anahita Ataran
- Center for Cardiovascular Research, Washington University School of Medicine, St. Louis, MO 63110, USA; (A.A.); (A.P.); (H.H.); (R.L.)
| | - Alexander Pompian
- Center for Cardiovascular Research, Washington University School of Medicine, St. Louis, MO 63110, USA; (A.A.); (A.P.); (H.H.); (R.L.)
| | - Hamidreza Hajirezaei
- Center for Cardiovascular Research, Washington University School of Medicine, St. Louis, MO 63110, USA; (A.A.); (A.P.); (H.H.); (R.L.)
| | - Rehman Lodhi
- Center for Cardiovascular Research, Washington University School of Medicine, St. Louis, MO 63110, USA; (A.A.); (A.P.); (H.H.); (R.L.)
| | - Ali Javaheri
- Center for Cardiovascular Research, Washington University School of Medicine, St. Louis, MO 63110, USA; (A.A.); (A.P.); (H.H.); (R.L.)
- John Cochran VA Hospital, St. Louis, MO 63110, USA
| |
Collapse
|
6
|
Huang W, Yang X, Zhang N, Chen K, Xiao J, Qiu Z, You S, Gao Z, Ji J, Chen L. PDE5 inhibition mitigates heart failure in hyperlipidemia. Biomed Pharmacother 2024; 175:116710. [PMID: 38713942 DOI: 10.1016/j.biopha.2024.116710] [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: 01/30/2024] [Revised: 04/20/2024] [Accepted: 05/02/2024] [Indexed: 05/09/2024] Open
Abstract
PDE5 inhibitors was reported to play a protective role in both regulating lipid metabolism and reducing heart failure (HF). This study aimed to clarify the effectiveness of PDE5 inhibitors against hyperlipidemia-related HF by combining evidence from population-based study and animal models. The nationwide cohort study found that post-diagnostic use of PDE5 inhibitors was associated with a significantly lower risk of HF compared with patients who used alprostadil, especially among individuals with hyperlipidemia (adjusted HR = 0.56, 95% CI = 0.40-0.78). In animal models, sildenafil significantly recovered the cardiac structure and function induced by AAB surgery, as well as reversed liver dysfunction and ameliorated hyperlipidemia induced by HFD via reducing the level of ALT, AST and serum lipids. Lipidomic analysis identified four lipid metabolites involved in sildenafil administration, including FA 16:3, LPC O-18:1, DG24:0_18:0 and SE28:1/20:4. This study revealed the protective effect of PDE5 inhibitors against HF in hyperlipidemia, indicating the potential of being repurposed as an adjuvant for HF prevention in patients with hyperlipidemia if these findings can be further confirmed in clinical trials.
Collapse
Affiliation(s)
- Wuqing Huang
- Department of Epidemiology and Health Statistics, School of Public Health, Fujian Medical University, Fuzhou, Fujian, China; Key Laboratory of Cardio-Thoracic Surgery (Fujian Medical University), Fujian Province University, China; Department of Cardiovascular Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian, China.
| | - Xi Yang
- Key Laboratory of Cardio-Thoracic Surgery (Fujian Medical University), Fujian Province University, China; Department of Cardiovascular Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian, China
| | - Naiqi Zhang
- Center for Primary Health Care Research, Department of Clinical Sciences Malmö, Lund University, Sweden; Nutritional Epidemiology, Department of Clinical Sciences Malmö, Lund University, Malmö, Sweden
| | - Keyuan Chen
- Key Laboratory of Cardio-Thoracic Surgery (Fujian Medical University), Fujian Province University, China; Department of Cardiovascular Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian, China
| | - Jun Xiao
- Key Laboratory of Cardio-Thoracic Surgery (Fujian Medical University), Fujian Province University, China; Department of Cardiovascular Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian, China
| | - Zhihuang Qiu
- Key Laboratory of Cardio-Thoracic Surgery (Fujian Medical University), Fujian Province University, China; Department of Cardiovascular Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian, China
| | - Sujun You
- Department of Echocardiography, Fujian Medical University Union Hospital, Fuzhou, Fujian, China
| | - Ziting Gao
- Department of Epidemiology and Health Statistics, School of Public Health, Fujian Medical University, Fuzhou, Fujian, China
| | - Jianguang Ji
- Center for Primary Health Care Research, Department of Clinical Sciences Malmö, Lund University, Sweden.
| | - Liangwan Chen
- Key Laboratory of Cardio-Thoracic Surgery (Fujian Medical University), Fujian Province University, China; Department of Cardiovascular Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian, China.
| |
Collapse
|
7
|
Li X, Huang Z, Tian Y, Chen X, Wu H, Wang T. Association between plasma long-chain polyunsaturated n-3 fatty acids concentrations and cognitive function: findings from NHANES III. Front Psychol 2024; 15:1305570. [PMID: 38756498 PMCID: PMC11098013 DOI: 10.3389/fpsyg.2024.1305570] [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: 10/28/2023] [Accepted: 04/15/2024] [Indexed: 05/18/2024] Open
Abstract
Background With increased life expectancy, cognitive decline has emerged as a prevalent neurodegenerative disorder. Objective This study aimed to examine the correlation between concentrations of Plasma long-chain n-3 polyunsaturated fatty acids (LCPUFAs) and cognitive performance in elderly Americans. Methods Data were analyzed from older adults enrolled in two NHANES cycles. Participants completed four cognitive assessments, including the Immediate Recall Test (IRT), Delayed Recall Test (DRT), Animal Fluency Test (AFT), and Digit Symbol Substitution Test (DSST). Linear regression and restricted cubic spline modeling examined associations between plasma LCPUFAs levels and cognitive test outcomes. Results The cohort included 610 adults aged 69 years on average, 300 (49.2%) males and 310 (50.8%) females. The median LCPUFAs concentration was 309.4 μmol/L, with an interquartile range of 244.7-418.9 μmol/L. In unadjusted and adjusted generalized linear regression model analyses, circulating LCPUFAs exhibited significant positive correlations with DRT performance. No relationships were detected among those with chronic conditions (chronic heart failure, stroke, diabetes). A significant association between LCPUFAs levels and DRT scores was evident in males but not females. Conclusion Plasma LCPUFAs concentrations were significantly associated with DRT performance in males free of chronic illnesses, including heart failure, stroke, and diabetes.
Collapse
Affiliation(s)
- Xiaojing Li
- Department of Emergency, The Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen, China
| | - Zijie Huang
- Department of Emergency, The Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen, China
| | - Yueqin Tian
- Department of Rehabilitation Medicine, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Xing Chen
- Department of Emergency, The Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen, China
| | - Haidong Wu
- Department of Emergency, The Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen, China
| | - Tong Wang
- Department of Emergency, The Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen, China
| |
Collapse
|
8
|
Goyal P, Maurer MS, Roh J. Aging in Heart Failure: Embracing Biology Over Chronology: JACC Family Series. JACC. HEART FAILURE 2024; 12:795-809. [PMID: 38597865 PMCID: PMC11331491 DOI: 10.1016/j.jchf.2024.02.021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/26/2023] [Revised: 02/12/2024] [Accepted: 02/21/2024] [Indexed: 04/11/2024]
Abstract
Age is among the most potent risk factors for developing heart failure and is strongly associated with adverse outcomes. As the global population continues to age and the prevalence of heart failure rises, understanding the role of aging in the development and progression of this chronic disease is essential. Although chronologic age is on a fixed course, biological aging is more variable and potentially modifiable in patients with heart failure. This review describes the current knowledge on mechanisms of biological aging that contribute to the pathogenesis of heart failure. The discussion focuses on 3 hallmarks of aging-impaired proteostasis, mitochondrial dysfunction, and deregulated nutrient sensing-that are currently being targeted in therapeutic development for older adults with heart failure. In assessing existing and emerging therapeutic strategies, the review also enumerates the importance of incorporating geriatric conditions into the management of older adults with heart failure and in ongoing clinical trials.
Collapse
Affiliation(s)
- Parag Goyal
- Department of Medicine, Weill Cornell Medicine, New York, New York, USA
| | - Mathew S Maurer
- Department of Medicine, Columbia University Medical Center, New York, New York, USA.
| | - Jason Roh
- Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
9
|
Kovilakath A, Wohlford G, Cowart LA. Circulating sphingolipids in heart failure. Front Cardiovasc Med 2023; 10:1154447. [PMID: 37229233 PMCID: PMC10203217 DOI: 10.3389/fcvm.2023.1154447] [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: 01/30/2023] [Accepted: 04/04/2023] [Indexed: 05/27/2023] Open
Abstract
Lack of significant advancements in early detection and treatment of heart failure have precipitated the need for discovery of novel biomarkers and therapeutic targets. Over the past decade, circulating sphingolipids have elicited promising results as biomarkers that premonish adverse cardiac events. Additionally, compelling evidence directly ties sphingolipids to these events in patients with incident heart failure. This review aims to summarize the current literature on circulating sphingolipids in both human cohorts and animal models of heart failure. The goal is to provide direction and focus for future mechanistic studies in heart failure, as well as pave the way for the development of new sphingolipid biomarkers.
Collapse
Affiliation(s)
- Anna Kovilakath
- Department of Human and Molecular Genetics, Virginia Commonwealth University, Richmond, VA, United States
| | - George Wohlford
- Pauley Heart Center, Virginia Commonwealth University, Richmond, VA, United States
| | - L. Ashley Cowart
- Department of Biochemistry and Molecular Biology and the Massey Cancer Center, Virginia Commonwealth University, Richmond, VA, United States
- Richmond Veteran's Affairs Medical Center, Richmond, VA, United States
| |
Collapse
|
10
|
Schenkl C, Heyne E, Doenst T, Schulze PC, Nguyen TD. Targeting Mitochondrial Metabolism to Save the Failing Heart. Life (Basel) 2023; 13:life13041027. [PMID: 37109556 PMCID: PMC10143865 DOI: 10.3390/life13041027] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2023] [Revised: 03/28/2023] [Accepted: 04/11/2023] [Indexed: 04/29/2023] Open
Abstract
Despite considerable progress in treating cardiac disorders, the prevalence of heart failure (HF) keeps growing, making it a global medical and economic burden. HF is characterized by profound metabolic remodeling, which mostly occurs in the mitochondria. Although it is well established that the failing heart is energy-deficient, the role of mitochondria in the pathophysiology of HF extends beyond the energetic aspects. Changes in substrate oxidation, tricarboxylic acid cycle and the respiratory chain have emerged as key players in regulating myocardial energy homeostasis, Ca2+ handling, oxidative stress and inflammation. This work aims to highlight metabolic alterations in the mitochondria and their far-reaching effects on the pathophysiology of HF. Based on this knowledge, we will also discuss potential metabolic approaches to improve cardiac function.
Collapse
Affiliation(s)
- Christina Schenkl
- Department of Cardiothoracic Surgery, Jena University Hospital, Friedrich Schiller University Jena, Am Klinikum 1, 07747 Jena, Germany
| | - Estelle Heyne
- Department of Cardiothoracic Surgery, Jena University Hospital, Friedrich Schiller University Jena, Am Klinikum 1, 07747 Jena, Germany
| | - Torsten Doenst
- Department of Cardiothoracic Surgery, Jena University Hospital, Friedrich Schiller University Jena, Am Klinikum 1, 07747 Jena, Germany
| | - Paul Christian Schulze
- Department of Medicine I (Cardiology, Angiology, Critical Care Medicine), Jena University Hospital, Friedrich Schiller University Jena, Am Klinikum 1, 07747 Jena, Germany
| | - Tien Dung Nguyen
- Department of Medicine I (Cardiology, Angiology, Critical Care Medicine), Jena University Hospital, Friedrich Schiller University Jena, Am Klinikum 1, 07747 Jena, Germany
| |
Collapse
|
11
|
Abstract
Chronic kidney disease is associated with an increased risk for the development and progression of cardiovascular disorders including hypertension, dyslipidemia, and coronary artery disease. Chronic kidney disease may also affect the myocardium through complex systemic changes, resulting in structural remodeling such as hypertrophy and fibrosis, as well as impairments in both diastolic and systolic function. These cardiac changes in the setting of chronic kidney disease define a specific cardiomyopathic phenotype known as uremic cardiomyopathy. Cardiac function is tightly linked to its metabolism, and research over the past 3 decades has revealed significant metabolic remodeling in the myocardium during the development of heart failure. Because the concept of uremic cardiomyopathy has only been recognized in recent years, there are limited data on metabolism in the uremic heart. Nonetheless, recent findings suggest overlapping mechanisms with heart failure. This work reviews key features of metabolic remodeling in the failing heart in the general population and extends this to patients with chronic kidney disease. The knowledge of similarities and differences in cardiac metabolism between heart failure and uremic cardiomyopathy may help identify new targets for mechanistic and therapeutic research on uremic cardiomyopathy.
Collapse
Affiliation(s)
- T Dung Nguyen
- Department of Internal Medicine I, University Hospital Jena, Jena, Germany
| | | |
Collapse
|
12
|
Rocca C, De Bartolo A, Guzzi R, Crocco MC, Rago V, Romeo N, Perrotta I, De Francesco EM, Muoio MG, Granieri MC, Pasqua T, Mazza R, Boukhzar L, Lefranc B, Leprince J, Gallo Cantafio ME, Soda T, Amodio N, Anouar Y, Angelone T. Palmitate-Induced Cardiac Lipotoxicity Is Relieved by the Redox-Active Motif of SELENOT through Improving Mitochondrial Function and Regulating Metabolic State. Cells 2023; 12:cells12071042. [PMID: 37048116 PMCID: PMC10093731 DOI: 10.3390/cells12071042] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2023] [Revised: 03/22/2023] [Accepted: 03/27/2023] [Indexed: 04/14/2023] Open
Abstract
Cardiac lipotoxicity is an important contributor to cardiovascular complications during obesity. Given the fundamental role of the endoplasmic reticulum (ER)-resident Selenoprotein T (SELENOT) for cardiomyocyte differentiation and protection and for the regulation of glucose metabolism, we took advantage of a small peptide (PSELT), derived from the SELENOT redox-active motif, to uncover the mechanisms through which PSELT could protect cardiomyocytes against lipotoxicity. To this aim, we modeled cardiac lipotoxicity by exposing H9c2 cardiomyocytes to palmitate (PA). The results showed that PSELT counteracted PA-induced cell death, lactate dehydrogenase release, and the accumulation of intracellular lipid droplets, while an inert form of the peptide (I-PSELT) lacking selenocysteine was not active against PA-induced cardiomyocyte death. Mechanistically, PSELT counteracted PA-induced cytosolic and mitochondrial oxidative stress and rescued SELENOT expression that was downregulated by PA through FAT/CD36 (cluster of differentiation 36/fatty acid translocase), the main transporter of fatty acids in the heart. Immunofluorescence analysis indicated that PSELT also relieved the PA-dependent increase in CD36 expression, while in SELENOT-deficient cardiomyocytes, PA exacerbated cell death, which was not mitigated by exogenous PSELT. On the other hand, PSELT improved mitochondrial respiration during PA treatment and regulated mitochondrial biogenesis and dynamics, preventing the PA-provoked decrease in PGC1-α and increase in DRP-1 and OPA-1. These findings were corroborated by transmission electron microscopy (TEM), revealing that PSELT improved the cardiomyocyte and mitochondrial ultrastructures and restored the ER network. Spectroscopic characterization indicated that PSELT significantly attenuated infrared spectral-related macromolecular changes (i.e., content of lipids, proteins, nucleic acids, and carbohydrates) and also prevented the decrease in membrane fluidity induced by PA. Our findings further delineate the biological significance of SELENOT in cardiomyocytes and indicate the potential of its mimetic PSELT as a protective agent for counteracting cardiac lipotoxicity.
Collapse
Affiliation(s)
- Carmine Rocca
- Cellular and Molecular Cardiovascular Pathophysiology Laboratory, Department of Biology, Ecology and Earth Sciences (DiBEST), University of Calabria, 87036 Rende, Italy
| | - Anna De Bartolo
- Cellular and Molecular Cardiovascular Pathophysiology Laboratory, Department of Biology, Ecology and Earth Sciences (DiBEST), University of Calabria, 87036 Rende, Italy
- UNIROUEN, Inserm U1239, Neuroendocrine, Endocrine and Germinal Differentiation and Communication (NorDiC), Rouen Normandie University, 76000 Mont-Saint-Aignan, France
| | - Rita Guzzi
- Department of Physics, Molecular Biophysics Laboratory, University of Calabria, 87036 Rende, Italy
- CNR-NANOTEC, Department of Physics, University of Calabria, 87036 Rende, Italy
| | - Maria Caterina Crocco
- Department of Physics, Molecular Biophysics Laboratory, University of Calabria, 87036 Rende, Italy
- STAR Research Infrastructure, University of Calabria, Via Tito Flavio, 87036 Rende, Italy
| | - Vittoria Rago
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, 87036 Rende, Italy
| | - Naomi Romeo
- Cellular and Molecular Cardiovascular Pathophysiology Laboratory, Department of Biology, Ecology and Earth Sciences (DiBEST), University of Calabria, 87036 Rende, Italy
| | - Ida Perrotta
- Centre for Microscopy and Microanalysis (CM2), Department of Biology, Biology, Ecology and Earth Sciences (DiBEST), University of Calabria, 87036 Rende, Italy
| | - Ernestina Marianna De Francesco
- Endocrinology, Department of Clinical and Experimental Medicine, University of Catania, Garibaldi-Nesima Hospital, 95124 Catania, Italy
| | - Maria Grazia Muoio
- Endocrinology, Department of Clinical and Experimental Medicine, University of Catania, Garibaldi-Nesima Hospital, 95124 Catania, Italy
| | - Maria Concetta Granieri
- Cellular and Molecular Cardiovascular Pathophysiology Laboratory, Department of Biology, Ecology and Earth Sciences (DiBEST), University of Calabria, 87036 Rende, Italy
| | - Teresa Pasqua
- Department of Health Science, University Magna Graecia of Catanzaro, 88100 Catanzaro, Italy
| | - Rosa Mazza
- Cellular and Molecular Cardiovascular Pathophysiology Laboratory, Department of Biology, Ecology and Earth Sciences (DiBEST), University of Calabria, 87036 Rende, Italy
| | - Loubna Boukhzar
- UNIROUEN, Inserm U1239, Neuroendocrine, Endocrine and Germinal Differentiation and Communication (NorDiC), Rouen Normandie University, 76000 Mont-Saint-Aignan, France
| | - Benjamin Lefranc
- UNIROUEN, Inserm U1239, Neuroendocrine, Endocrine and Germinal Differentiation and Communication (NorDiC), Rouen Normandie University, 76000 Mont-Saint-Aignan, France
- UNIROUEN, UMS-UAR HERACLES, PRIMACEN, Cell Imaging Platform of Normandy, Institute for Research and Innovation in Biomedicine (IRIB), 76183 Rouen, France
| | - Jérôme Leprince
- UNIROUEN, Inserm U1239, Neuroendocrine, Endocrine and Germinal Differentiation and Communication (NorDiC), Rouen Normandie University, 76000 Mont-Saint-Aignan, France
- UNIROUEN, UMS-UAR HERACLES, PRIMACEN, Cell Imaging Platform of Normandy, Institute for Research and Innovation in Biomedicine (IRIB), 76183 Rouen, France
| | | | - Teresa Soda
- Department of Health Science, University Magna Graecia of Catanzaro, 88100 Catanzaro, Italy
| | - Nicola Amodio
- Department of Experimental and Clinical Medicine, Magna Graecia University, 88100 Catanzaro, Italy
| | - Youssef Anouar
- UNIROUEN, Inserm U1239, Neuroendocrine, Endocrine and Germinal Differentiation and Communication (NorDiC), Rouen Normandie University, 76000 Mont-Saint-Aignan, France
- UNIROUEN, UMS-UAR HERACLES, PRIMACEN, Cell Imaging Platform of Normandy, Institute for Research and Innovation in Biomedicine (IRIB), 76183 Rouen, France
| | - Tommaso Angelone
- Cellular and Molecular Cardiovascular Pathophysiology Laboratory, Department of Biology, Ecology and Earth Sciences (DiBEST), University of Calabria, 87036 Rende, Italy
- National Institute of Cardiovascular Research (INRC), 40126 Bologna, Italy
| |
Collapse
|
13
|
Zou L, Yang Y, Wang Z, Fu X, He X, Song J, Li T, Ma H, Yu T. Lysine Malonylation and Its Links to Metabolism and Diseases. Aging Dis 2023; 14:84-98. [PMID: 36818560 PMCID: PMC9937698 DOI: 10.14336/ad.2022.0711] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Accepted: 07/11/2022] [Indexed: 11/18/2022] Open
Abstract
Malonylation is a recently identified post-translational modification with malonyl-coenzyme A as the donor. It conserved both in prokaryotes and eukaryotes. Recent advances in the identification and quantification of lysine malonylation by bioinformatic analysis have improved our understanding of its role in the regulation of protein activity, interaction, and localization and have elucidated its involvement in many biological processes. Malonylation has been linked to diverse physiological processes, including metabolic disorders, inflammation, and immune regulation. This review discusses malonylation in theory, describes the underlying mechanism, and summarizes the recent progress in malonylation research. The latest findings point to novel functions of malonylation and highlight the mechanisms by which malonylation regulates a variety of cellular processes. Our review also marks the association between lysine malonylation, the enzymes involved, and various diseases, and discusses promising diagnostic and therapeutic biomolecular targets for future clinical applications.
Collapse
Affiliation(s)
- Lu Zou
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, Qingdao, China.
| | - Yanyan Yang
- Department of Immunology, Basic Medicine School, Qingdao University, Qingdao, China.
| | - Zhibin Wang
- Department of Cardiac Ultrasound, The Affiliated Hospital of Qingdao University, Qingdao, China.
| | - Xiuxiu Fu
- Department of Cardiac Ultrasound, The Affiliated Hospital of Qingdao University, Qingdao, China.
| | - Xiangqin He
- Department of Cardiac Ultrasound, The Affiliated Hospital of Qingdao University, Qingdao, China.
| | - Jiayi Song
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, Qingdao, China.
| | - Tianxiang Li
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, Qingdao, China.
| | - Huibo Ma
- Department of Vascular Surgery, The Affiliated Hospital of Qingdao University, Qingdao, China.
| | - Tao Yu
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, Qingdao, China.,Department of Cardiac Ultrasound, The Affiliated Hospital of Qingdao University, Qingdao, China.,Correspondence should be addressed to: Dr. Tao Yu, Center for Regenerative Medicine, Institute for Translational Medicine, the Affiliated Hospital of Qingdao University, Qingdao, China.
| |
Collapse
|
14
|
Semeraro MD, Beltrami AP, Kharrat F, Almer G, Sedej S, Renner W, Gruber HJ, Curcio F, Herrmann M. The impact of moderate endurance exercise on cardiac telomeres and cardiovascular remodeling in obese rats. Front Cardiovasc Med 2023; 9:1080077. [PMID: 36684585 PMCID: PMC9853517 DOI: 10.3389/fcvm.2022.1080077] [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: 10/25/2022] [Accepted: 12/13/2022] [Indexed: 01/07/2023] Open
Abstract
Introduction Hypercaloric nutrition and physical inactivity cause obesity, a potential driver of myocardial apoptosis and senescence that may accelerate cardiac aging. Although physical activity reduces mortality, its impact on myocardial aging is insufficiently understood. Here we investigated the effects of a hypercaloric high-fat diet (HFD) and regular exercise training on cardiac cells telomeres and histomorphometric indices of cardiac aging. Methods Ninety-six 4-months old female Sprague-Dawley rats were fed for 10 months normal (ND) or a HFD diet. Half of the animals in each group performed 30 min treadmill-running sessions on 5 consecutive days per week. At study end, cardiomyocyte cross-sectional area (CSA), interstitial collagen content, vascular density, apoptotic and senescent cells, relative telomere length (RTL), and expression of telomerase-reverse transcriptase (Tert) as marker of telomere-related senescence and apoptosis were analyzed. Results Compared to ND, the HFD group developed obesity, higher CSA, lower capillary density and tended to have more apoptotic cardiomyocytes and interstitials cells. Myocardial RTL and the expression of Terf-1 and Terf-2 were comparable in sedentary HFD and ND animals. In the HFD group, regular moderate endurance exercise improved myocardial vascularization, but had no effect on CSA or apoptosis. Notably, the combination of exercise and HFD increased senescence when compared to sedentary ND or HFD, and reduced RTL when compared to exercise ND animals. Exercising HFD animals also showed a trend toward higher Tert expression compared to all other groups. In addition, exercise reduced Terf-1 expression regardless of diet. Conclusion HFD-induced obesity showed no effects on myocardial telomeres and induced only mild morphologic alterations. Summarized, long-term moderate endurance exercise partially reverses HFD-induced effects but may even trigger cardiac remodeling in the context of obesity.
Collapse
Affiliation(s)
- Maria Donatella Semeraro
- Clinical Institute of Medical and Chemical Laboratory Diagnostics, Medical University of Graz, Graz, Austria
| | - Antonio Paolo Beltrami
- Department of Medicine (DAME), University of Udine, Udine, Italy,*Correspondence: Antonio Paolo Beltrami ✉
| | - Feras Kharrat
- Department of Medicine (DAME), University of Udine, Udine, Italy
| | - Gunter Almer
- Clinical Institute of Medical and Chemical Laboratory Diagnostics, Medical University of Graz, Graz, Austria
| | - Simon Sedej
- Department of Cardiology, Medical University of Graz, Graz, Austria,BTM BioTechMed-Graz, Graz, Austria,Faculty of Medicine, University of Maribor, Maribor, Slovenia
| | - Wilfried Renner
- Clinical Institute of Medical and Chemical Laboratory Diagnostics, Medical University of Graz, Graz, Austria
| | - Hans-Jürgen Gruber
- Clinical Institute of Medical and Chemical Laboratory Diagnostics, Medical University of Graz, Graz, Austria
| | - Francesco Curcio
- Department of Medicine (DAME), University of Udine, Udine, Italy
| | - Markus Herrmann
- Clinical Institute of Medical and Chemical Laboratory Diagnostics, Medical University of Graz, Graz, Austria
| |
Collapse
|
15
|
Sztolsztener K, Bzdęga W, Hodun K, Chabowski A. N-Acetylcysteine Decreases Myocardial Content of Inflammatory Mediators Preventing the Development of Inflammation State and Oxidative Stress in Rats Subjected to a High-Fat Diet. Int J Inflam 2023; 2023:5480199. [PMID: 36941865 PMCID: PMC10024630 DOI: 10.1155/2023/5480199] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Revised: 02/15/2023] [Accepted: 03/06/2023] [Indexed: 03/13/2023] Open
Abstract
Arachidonic acid (AA) is a key precursor for proinflammatory and anti-inflammatory derivatives that regulate the inflammatory response. The modulation of AA metabolism is a target for searching a therapeutic agent with potent anti-inflammatory action in cardiovascular disorders. Therefore, our study aims to determine the potential preventive impact of N-acetylcysteine (NAC) supplementation on myocardial inflammation and the occurrence of oxidative stress in obesity induced by high-fat feeding. The experiment was conducted for eight weeks on male Wistar rats fed a standard chow or a high-fat diet (HFD) with intragastric NAC supplementation. The Gas-Liquid Chromatography (GLC) method was used to quantify the plasma and myocardial AA levels in the selected lipid fraction. The expression of proteins included in the inflammation pathway was measured by the Western blot technique. The concentrations of arachidonic acid derivatives, cytokines and chemokines, and oxidative stress parameters were determined by the ELISA, colorimetric, and multiplex immunoassay kits. We established that in the left ventricle tissue NAC reduced AA concentration, especially in the phospholipid fraction. NAC administration ameliorated the COX-2 and 5-LOX expression, leading to a decrease in the PGE2 and LTC4 contents, respectively, and augmented the 12/15-LOX expression, increasing the LXA4 content. In obese rats, NAC ameliorated NF-κB expression, inhibiting the secretion of proinflammatory cytokines. NAC also affected the antioxidant levels in HFD rats through an increase in GSH and CAT contents with a simultaneous decrease in the levels of 4-HNE and MDA. We concluded that NAC treatment weakens the NF-κB signaling pathway, limiting the development of myocardial low-grade inflammation, and increasing the antioxidant content that may protect against the development of oxidative stress in rats with obesity induced by an HFD.
Collapse
Affiliation(s)
- Klaudia Sztolsztener
- Department of Physiology, Medical University of Bialystok, 15-089 Bialystok, Poland
| | - Wiktor Bzdęga
- Department of Physiology, Medical University of Bialystok, 15-089 Bialystok, Poland
| | - Katarzyna Hodun
- Department of Physiology, Medical University of Bialystok, 15-089 Bialystok, Poland
| | - Adrian Chabowski
- Department of Physiology, Medical University of Bialystok, 15-089 Bialystok, Poland
| |
Collapse
|
16
|
Influence of cardiac function on intermittent hypoxia in rats fed with high-fat diet. Biochem Biophys Rep 2022; 32:101393. [DOI: 10.1016/j.bbrep.2022.101393] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2022] [Revised: 11/15/2022] [Accepted: 11/18/2022] [Indexed: 11/24/2022] Open
|
17
|
Kuhn AR, van Bilsen M. Oncometabolism: A Paradigm for the Metabolic Remodeling of the Failing Heart. Int J Mol Sci 2022; 23:ijms232213902. [PMID: 36430377 PMCID: PMC9699042 DOI: 10.3390/ijms232213902] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2022] [Revised: 11/03/2022] [Accepted: 11/07/2022] [Indexed: 11/16/2022] Open
Abstract
Heart failure is associated with profound alterations in cardiac intermediary metabolism. One of the prevailing hypotheses is that metabolic remodeling leads to a mismatch between cardiac energy (ATP) production and demand, thereby impairing cardiac function. However, even after decades of research, the relevance of metabolic remodeling in the pathogenesis of heart failure has remained elusive. Here we propose that cardiac metabolic remodeling should be looked upon from more perspectives than the mere production of ATP needed for cardiac contraction and relaxation. Recently, advances in cancer research have revealed that the metabolic rewiring of cancer cells, often coined as oncometabolism, directly impacts cellular phenotype and function. Accordingly, it is well feasible that the rewiring of cardiac cellular metabolism during the development of heart failure serves similar functions. In this review, we reflect on the influence of principal metabolic pathways on cellular phenotype as originally described in cancer cells and discuss their potential relevance for cardiac pathogenesis. We discuss current knowledge of metabolism-driven phenotypical alterations in the different cell types of the heart and evaluate their impact on cardiac pathogenesis and therapy.
Collapse
|
18
|
Singh RB, Fedacko J, Pella D, Fatima G, Elkilany G, Moshiri M, Hristova K, Jakabcin P, Vaňova N. High Exogenous Antioxidant, Restorative Treatment (Heart) for Prevention of the Six Stages of Heart Failure: The Heart Diet. Antioxidants (Basel) 2022; 11:1464. [PMID: 36009183 PMCID: PMC9404840 DOI: 10.3390/antiox11081464] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2022] [Revised: 07/08/2022] [Accepted: 07/19/2022] [Indexed: 01/06/2023] Open
Abstract
The exact pathophysiology of heart failure (HF) is not yet known. Western diet, characterized by highly sweetened foods, as well as being rich in fat, fried foods, red meat and processed meat, eggs, and sweet beverages, may cause inflammation, leading to oxidative dysfunction in the cardiac ultra-structure. Oxidative function of the myocardium and how oxidative dysfunction causes physio-pathological remodeling, leading to HF, is not well known. Antioxidants, such as polyphenolics and flavonoids, omega-3 fatty acids, and other micronutrients that are rich in Indo-Mediterranean-type diets, could be protective in sustaining the oxidative functions of the heart. The cardiomyocytes use glucose and fatty acids for the physiological functions depending upon the metabolic requirements of the heart. Apart from toxicity due to glucose, lipotoxicity also adversely affects the cardiomyocytes, which worsen in the presence of deficiency of endogenous antioxidants and deficiency of exogenous antioxidant nutrients in the diet. The high-sugar-and-high-fat-induced production of ceramide, advanced glycation end products (AGE) and triamino-methyl-N-oxide (TMAO) can predispose individuals to oxidative dysfunction and Ca-overloading. The alteration in the biology may start with normal cardiac cell remodeling to biological remodeling due to inflammation. An increase in the fat content of a diet in combination with inducible nitric oxide synthase (NOSi) via N-arginine methyl ester has been found to preserve the ejection fraction in HF. It is proposed that a greater intake of high exogenous antioxidant restorative treatment (HEART) diet, polyphenolics and flavonoids, as well as cessation of red meat intake and egg, can cause improvement in the oxidative function of the heart, by inhibiting oxidative damage to lipids, proteins and DNA in the cell, resulting in beneficial effects in the early stage of the Six Stages of HF. There is an unmet need to conduct cohort studies and randomized, controlled studies to demonstrate the role of the HEART diet in the treatment of HF.
Collapse
Affiliation(s)
- Ram B. Singh
- Halberg Hospital and Research Institute, Moradabad 244001, India;
| | - Jan Fedacko
- Department of Gerontology and Geriatric, Medipark, University Research Park, PJ Safarik University, 040-11 Kosice, Slovakia
| | - Dominik Pella
- Department of Cardiology, Faculty of Medicine and East Slovak, Institute for Cardiovascular Disease, PJ Safarik University, 040-11 Kosice, Slovakia;
| | - Ghizal Fatima
- Department of Biotechnology, Era University, Lucknow 226001, India;
| | - Galal Elkilany
- International College of Cardiology, Laplace, LA 90001, USA;
| | - Mahmood Moshiri
- International College of Cardiology, Richmond Hill, ON LL-9955, Canada;
| | - Krasimira Hristova
- Department of Cardiology, National University Hospital, 1000 Sofia, Bulgaria;
| | - Patrik Jakabcin
- Department of Social and Clinical Pharmacy, Faculty of Pharmacy in Hradec Králové, Charles University, 10000 Prague, Czech Republic;
| | - Natalia Vaňova
- Department of Internal Medicine UPJS MF and AGEL Hospital, Research Park, PJ Safaric University, 040-11 Kosice, Slovakia;
| |
Collapse
|
19
|
Zhou H, Li J, Su H, Li J, Lydic TA, Young ME, Chen W. BSCL2/Seipin deficiency in hearts causes cardiac energy deficit and dysfunction via inducing excessive lipid catabolism. Clin Transl Med 2022; 12:e736. [PMID: 35384404 PMCID: PMC8982503 DOI: 10.1002/ctm2.736] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2021] [Revised: 01/26/2022] [Accepted: 01/29/2022] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND Heart failure (HF) is one of the leading causes of death worldwide and is associated with cardiac metabolic perturbations. Human Type 2 Berardinelli-Seip Congenital Lipodystrophy (BSCL2) disease is caused by mutations in the BSCL2 gene. Global lipodystrophic Bscl2-/- mice exhibit hypertrophic cardiomyopathy with reduced cardiac steatosis. Whether BSCL2 plays a direct role in regulating cardiac substrate metabolism and/or contractile function remains unknown. METHODS We generated mice with cardiomyocyte-specific deletion of Bscl2 (Bscl2cKO ) and studied their cardiac substrate utilisation, bioenergetics, lipidomics and contractile function under baseline or after either a treatment regimen using fatty acid oxidation (FAO) inhibitor trimetazidine (TMZ) or a prevention regimen with high-fat diet (HFD) feeding. Mice with partial ATGL deletion and cardiac-specific deletion of Bscl2 were also generated followed by cardiac phenotyping. RESULTS Different from hypertrophic cardiomyopathy in Bscl2-/- mice, mice with cardiac-specific deletion of Bscl2 developed systolic dysfunction with dilation. Myocardial BSCL2 deletion led to elevated ATGL expression and FAO along with reduced cardiac lipid contents. Cardiac dysfunction in Bscl2cKO mice was independent of mitochondrial dysfunction and oxidative stress, but associated with decreased metabolic reserve and ATP levels. Importantly, cardiac dysfunction in Bscl2cKO mice could be partially reversed by FAO inhibitor TMZ, or prevented by genetic abolishment of one ATGL allele or HFD feeding. Lipidomic analysis further identified markedly reduced glycerolipids, glycerophospholipids, NEFA and acylcarnitines in Bscl2cKO hearts, which were partially normalised by TMZ or HFD. CONCLUSIONS We identified a new form of cardiac dysfunction with excessive lipid utilisation which ultimately causes cardiac substrate depletion and bioenergetics failure. Our findings also uncover a crucial role of BSCL2 in controlling cardiac lipid catabolism and contractile function and provide novel insights into metabolically treating energy-starved HF using FAO inhibitor or HFD.
Collapse
Affiliation(s)
- Hongyi Zhou
- Department of PhysiologyMedical College of Georgia at Augusta UniversityAugustaGeorgiaUSA
| | - Jie Li
- Vascular Biology CenterMedical College of Georgia at Augusta UniversityAugustaGeorgiaUSA
| | - Huabo Su
- Vascular Biology CenterMedical College of Georgia at Augusta UniversityAugustaGeorgiaUSA
| | - Ji Li
- Department of SurgeryMorsani College of MedicineUniversity of South FloridaTampaFloridaUSA
| | - Todd A. Lydic
- Department of PhysiologyMichigan State UniversityEast LansingMichiganUSA
| | - Martin E Young
- Department of MedicineUniversity of Alabama at BirminghamBirminghamAlabamaUSA
| | - Weiqin Chen
- Department of PhysiologyMedical College of Georgia at Augusta UniversityAugustaGeorgiaUSA
| |
Collapse
|
20
|
Song S, Tien CL, Cui H, Basil P, Zhu N, Gong Y, Li W, Li H, Fan Q, Min Choi J, Luo W, Xue Y, Cao R, Zhou W, Ortiz AR, Stork B, Mundra V, Putluri N, York B, Chu M, Chang J, Yun Jung S, Xie L, Song J, Zhang L, Sun Z. Myocardial Rev-erb-Mediated Diurnal Metabolic Rhythm and Obesity Paradox. Circulation 2022; 145:448-464. [PMID: 35034472 PMCID: PMC8812427 DOI: 10.1161/circulationaha.121.056076] [Citation(s) in RCA: 47] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/11/2021] [Accepted: 12/02/2021] [Indexed: 11/16/2022]
Abstract
BACKGROUND The nuclear receptor Rev-erbα/β, a key component of the circadian clock, emerges as a drug target for heart diseases, but the function of cardiac Rev-erb has not been studied in vivo. Circadian disruption is implicated in heart diseases, but it is unknown whether cardiac molecular clock dysfunction is associated with the progression of any naturally occurring human heart diseases. Obesity paradox refers to the seemingly protective role of obesity for heart failure, but the mechanism is unclear. METHODS We generated mouse lines with cardiac-specific Rev-erbα/β knockout (KO), characterized cardiac phenotype, conducted multi-omics (RNA-sequencing, chromatin immunoprecipitation sequencing, proteomics, and metabolomics) analyses, and performed dietary and pharmacological rescue experiments to assess the time-of-the-day effects. We compared the temporal pattern of cardiac clock gene expression with the cardiac dilation severity in failing human hearts. RESULTS KO mice display progressive dilated cardiomyopathy and lethal heart failure. Inducible ablation of Rev-erbα/β in adult hearts causes similar phenotypes. Impaired fatty acid oxidation in the KO myocardium, in particular, in the light cycle, precedes contractile dysfunctions with a reciprocal overreliance on carbohydrate utilization, in particular, in the dark cycle. Increasing dietary lipid or sugar supply in the dark cycle does not affect cardiac dysfunctions in KO mice. However, obesity coupled with systemic insulin resistance paradoxically ameliorates cardiac dysfunctions in KO mice, associated with rescued expression of lipid oxidation genes only in the light cycle in phase with increased fatty acid availability from adipose lipolysis. Inhibition of glycolysis in the light cycle and lipid oxidation in the dark cycle, but not vice versa, ameliorate cardiac dysfunctions in KO mice. Altered temporal patterns of cardiac Rev-erb gene expression correlate with the cardiac dilation severity in human hearts with dilated cardiomyopathy. CONCLUSIONS The study delineates temporal coordination between clock-mediated anticipation and nutrient-induced response in myocardial metabolism at multi-omics levels. The obesity paradox is attributable to increased cardiac lipid supply from adipose lipolysis in the fasting cycle due to systemic insulin resistance and adiposity. Cardiac molecular chronotypes may be involved in human dilated cardiomyopathy. Myocardial bioenergetics downstream of Rev-erb may be a chronotherapy target in treating heart failure and dilated cardiomyopathy.
Collapse
Affiliation(s)
- Shiyang Song
- Department of Medicine, Division of Diabetes, Endocrinology, and Metabolism (S.S., P.B., N.Z., Y.G., W. Li, Y.X., R.C., W.Z., V.M., Z.S.), Baylor College of Medicine, Houston, TX
- Children’s Heart Center, Institute of Cardiovascular Development and Translational Medicine, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, China (S.S., M.C.)
| | - Chih-Liang Tien
- Department of Molecular and Human Genetics (C.-L.T., H.L., L.Z.), Baylor College of Medicine, Houston, TX
| | - Hao Cui
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences (CAMS) and Peking Union Medical College (PUMC), Beijing, China (H.C., J.S.)
| | - Paul Basil
- Department of Critical Care, Division of Anesthesiology, Critical Care, and Pain Medicine, University of Texas MD Anderson Cancer Center, Houston, TX (P.B.)
| | - Ningxia Zhu
- Department of Pathology and Physiopathology, Guilin Medical University, Guilin, China (N.Z.)
| | - Yingyun Gong
- Department of Medicine, Division of Diabetes, Endocrinology, and Metabolism (S.S., P.B., N.Z., Y.G., W. Li, Y.X., R.C., W.Z., V.M., Z.S.), Baylor College of Medicine, Houston, TX
| | - Wenbo Li
- Department of Medicine, Division of Diabetes, Endocrinology, and Metabolism (S.S., P.B., N.Z., Y.G., W. Li, Y.X., R.C., W.Z., V.M., Z.S.), Baylor College of Medicine, Houston, TX
| | - Hui Li
- Department of Molecular and Human Genetics (C.-L.T., H.L., L.Z.), Baylor College of Medicine, Houston, TX
| | - Qiying Fan
- Department of Medicine, Division of Atherosclerosis and Vascular Medicine, Cardiovascular Research Institute (CVRI), Houston, TX (Q.F., L.X.)
| | - Jong Min Choi
- Department of Molecular and Cellular Biology (J.M.C., A.R.O., B.S., N.P., B.Y., S.Y.J.), Baylor College of Medicine, Houston, TX
| | - Weijia Luo
- Center for Genomic and Precision Medicine, Texas A&M University, Institute of Biosciences and Technology, Houston (W. Luo, J.C., L.Z.)
| | - Yanfeng Xue
- Department of Medicine, Division of Diabetes, Endocrinology, and Metabolism (S.S., P.B., N.Z., Y.G., W. Li, Y.X., R.C., W.Z., V.M., Z.S.), Baylor College of Medicine, Houston, TX
| | - Rui Cao
- Department of Medicine, Division of Diabetes, Endocrinology, and Metabolism (S.S., P.B., N.Z., Y.G., W. Li, Y.X., R.C., W.Z., V.M., Z.S.), Baylor College of Medicine, Houston, TX
| | - Wenjun Zhou
- Department of Medicine, Division of Diabetes, Endocrinology, and Metabolism (S.S., P.B., N.Z., Y.G., W. Li, Y.X., R.C., W.Z., V.M., Z.S.), Baylor College of Medicine, Houston, TX
| | - Andrea R. Ortiz
- Department of Molecular and Cellular Biology (J.M.C., A.R.O., B.S., N.P., B.Y., S.Y.J.), Baylor College of Medicine, Houston, TX
| | - Brittany Stork
- Department of Molecular and Cellular Biology (J.M.C., A.R.O., B.S., N.P., B.Y., S.Y.J.), Baylor College of Medicine, Houston, TX
| | - Vatsala Mundra
- Department of Medicine, Division of Diabetes, Endocrinology, and Metabolism (S.S., P.B., N.Z., Y.G., W. Li, Y.X., R.C., W.Z., V.M., Z.S.), Baylor College of Medicine, Houston, TX
| | - Nagireddy Putluri
- Department of Molecular and Cellular Biology (J.M.C., A.R.O., B.S., N.P., B.Y., S.Y.J.), Baylor College of Medicine, Houston, TX
| | - Brian York
- Department of Molecular and Cellular Biology (J.M.C., A.R.O., B.S., N.P., B.Y., S.Y.J.), Baylor College of Medicine, Houston, TX
| | - Maoping Chu
- Children’s Heart Center, Institute of Cardiovascular Development and Translational Medicine, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, China (S.S., M.C.)
| | - Jiang Chang
- Center for Genomic and Precision Medicine, Texas A&M University, Institute of Biosciences and Technology, Houston (W. Luo, J.C., L.Z.)
| | - Sung Yun Jung
- Department of Molecular and Cellular Biology (J.M.C., A.R.O., B.S., N.P., B.Y., S.Y.J.), Baylor College of Medicine, Houston, TX
| | - Liang Xie
- Department of Medicine, Division of Diabetes, Endocrinology, and Metabolism (S.S., P.B., N.Z., Y.G., W. Li, Y.X., R.C., W.Z., V.M., Z.S.), Baylor College of Medicine, Houston, TX
| | - Jiangping Song
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences (CAMS) and Peking Union Medical College (PUMC), Beijing, China (H.C., J.S.)
| | - Lilei Zhang
- Department of Molecular and Human Genetics (C.-L.T., H.L., L.Z.), Baylor College of Medicine, Houston, TX
- Center for Genomic and Precision Medicine, Texas A&M University, Institute of Biosciences and Technology, Houston (W. Luo, J.C., L.Z.)
| | - Zheng Sun
- Department of Medicine, Division of Diabetes, Endocrinology, and Metabolism (S.S., P.B., N.Z., Y.G., W. Li, Y.X., R.C., W.Z., V.M., Z.S.), Baylor College of Medicine, Houston, TX
| |
Collapse
|
21
|
Sheashea M, Xiao J, Farag MA. MUFA in metabolic syndrome and associated risk factors: is MUFA the opposite side of the PUFA coin? Food Funct 2021; 12:12221-12234. [PMID: 34779464 DOI: 10.1039/d1fo00979f] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Omega-9 fatty acids represent some of the main mono-unsaturated fatty acids (MUFA) found in plant and animal sources. They can be synthesized endogenously in the human body, but they do not fully provide all the body's requirements. Consequently, they are considered as partially essential fatty acids. MUFA represent a healthier alternative to saturated animal fats and have several health benefits, including the prevention of metabolic syndrome (MetS) and its complications. This review concentrates on the major MUFA pharmacological activities in the context of MetS management, including alleviating cardiovascular disease (CVD) and dyslipidemia, central obesity, non-alcoholic fatty liver disease (NAFLD), and type 2 diabetes mellitus (T2DM). The beneficial effects of MUFA for CVD were found to be consistent with those of polyunsaturated fatty acids (PUFA) for the alleviation of systolic and diastolic blood pressure and high low density lipoprotein cholesterol (LDLc) and triacylglcerol (TAG) levels, albeit MUFA had a more favorable effect on decreasing night systolic blood pressure (SBP). To reduce the obesity profile, the use of MUFA was found to induce a higher oxidation rate with a higher energy expenditure, compared with PUFA. For NAFLD, PUFA was found to be a better potential drug candidate for the improvement of liver steatosis in children than MUFA. Any advantageous outcomes from using MUFA for diabetes and insulin resistance (IR) compared to using PUFA were found to be either non-significant or resulted from a small number of meta-analyses. Such an increase in the number of studies of the mechanisms of action require more clinical and epidemiological studies to confirm the beneficial outcomes, especially over a long-term treatment period.
Collapse
Affiliation(s)
- Mohamed Sheashea
- Aromatic and Medicinal Plants Department, Desert Research Center, Cairo, Egypt
| | - Jianbo Xiao
- Nutrition and Bromatology Group, Department of Analytical Chemistry and Food Science, Faculty of Food Science and Technology, University of Vigo - Ourense Campus, E-32004 Ourense, Spain.
- College of Food Science and Technology, Guangdong Ocean University, Zhanjiang 524088, China
- International Research Center for Food Nutrition and Safety, Jiangsu University, Zhenjiang 212013, China
| | - Mohamed A Farag
- Pharmacognosy Department, College of Pharmacy, Cairo University, Kasr el Aini St., P.B. 11562, Cairo, Egypt.
- Chemistry Department, School of Sciences & Engineering, The American University in Cairo, New Cairo 11835, Egypt
| |
Collapse
|
22
|
Schütte T, Kedziora SM, Haase N, Herse F, Alenina N, Müller DN, Bader M, Schupp M, Dechend R, Golic M, Kräker K. Diabetic pregnancy as a novel risk factor for cardiac dysfunction in the offspring-the heart as a target for fetal programming in rats. Diabetologia 2021; 64:2829-2842. [PMID: 34537857 PMCID: PMC8563640 DOI: 10.1007/s00125-021-05566-5] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/15/2021] [Accepted: 05/28/2021] [Indexed: 11/02/2022]
Abstract
AIMS/HYPOTHESIS The impact of diabetic pregnancy has been investigated extensively regarding offspring metabolism; however, little is known about the influence on the heart. We aimed to characterise the effects of a diabetic pregnancy on male adult offspring cardiac health after feeding a high-fat diet in an established transgenic rat model. METHODS We applied our rat model for maternal type 2 diabetes characterised by maternal insulin resistance with hyperglycaemia and hyperinsulinaemia. Diabetes was induced preconceptionally via doxycycline-induced knock down of the insulin receptor in transgenic rats. Male wild-type offspring of diabetic and normoglycaemic pregnancies were raised by foster mothers, followed up into adulthood and subgroups were challenged by a high-fat diet. Cardiac phenotype was assessed by innovative speckle tracking echocardiography, circulating factors, immunohistochemistry and gene expression in the heart. RESULTS When feeding normal chow, we did not observe differences in cardiac function, gene expression and plasma brain natriuretic peptide between adult diabetic or normoglycaemic offspring. Interestingly, when being fed a high-fat diet, adult offspring of diabetic pregnancy demonstrated decreased global longitudinal (-14.82 ± 0.59 vs -16.60 ± 0.48%) and circumferential strain (-23.40 ± 0.57 vs -26.74 ± 0.34%), increased relative wall thickness (0.53 ± 0.06 vs 0.37 ± 0.02), altered cardiac gene expression, enlarged cardiomyocytes (106.60 ± 4.14 vs 87.94 ± 1.67 μm), an accumulation of immune cells in the heart (10.27 ± 0.30 vs 6.48 ± 0.48 per fov) and higher plasma brain natriuretic peptide levels (0.50 ± 0.12 vs 0.12 ± 0.03 ng/ml) compared with normoglycaemic offspring on a high-fat diet. Blood pressure, urinary albumin, blood glucose and body weight were unaltered between groups on a high-fat diet. CONCLUSIONS/INTERPRETATION Diabetic pregnancy in rats induces cardiac dysfunction, left ventricular hypertrophy and altered proinflammatory status in adult offspring only after a high-fat diet. A diabetic pregnancy itself was not sufficient to impair myocardial function and gene expression in male offspring later in life. This suggests that a postnatal high-fat diet is important for the development of cardiac dysfunction in rat offspring after diabetic pregnancy. Our data provide evidence that a diabetic pregnancy is a novel cardiac risk factor that becomes relevant when other challenges, such as a high-fat diet, are present.
Collapse
Affiliation(s)
- Till Schütte
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Institute of Pharmacology, Berlin, Germany
- Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Berlin, Germany
- DZHK (German Center for Cardiovascular Research), Partner Site Berlin, Berlin, Germany
| | - Sarah M Kedziora
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Berlin, Germany
- DZHK (German Center for Cardiovascular Research), Partner Site Berlin, Berlin, Germany
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
- Experimental and Clinical Research Center - a joint cooperation between the Max Delbrück Center for Molecular Medicine and the Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Nadine Haase
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Berlin, Germany
- DZHK (German Center for Cardiovascular Research), Partner Site Berlin, Berlin, Germany
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
- Experimental and Clinical Research Center - a joint cooperation between the Max Delbrück Center for Molecular Medicine and the Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Florian Herse
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Berlin, Germany
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
- Experimental and Clinical Research Center - a joint cooperation between the Max Delbrück Center for Molecular Medicine and the Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Natalia Alenina
- Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Berlin, Germany
- DZHK (German Center for Cardiovascular Research), Partner Site Berlin, Berlin, Germany
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
| | - Dominik N Müller
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Berlin, Germany
- DZHK (German Center for Cardiovascular Research), Partner Site Berlin, Berlin, Germany
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
- Experimental and Clinical Research Center - a joint cooperation between the Max Delbrück Center for Molecular Medicine and the Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Michael Bader
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Berlin, Germany
- DZHK (German Center for Cardiovascular Research), Partner Site Berlin, Berlin, Germany
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
- Institute for Biology, University of Lübeck, Lübeck, Germany
| | - Michael Schupp
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Institute of Pharmacology, Berlin, Germany
- Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Ralf Dechend
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Berlin, Germany
- DZHK (German Center for Cardiovascular Research), Partner Site Berlin, Berlin, Germany
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
- Experimental and Clinical Research Center - a joint cooperation between the Max Delbrück Center for Molecular Medicine and the Charité - Universitätsmedizin Berlin, Berlin, Germany
- HELIOS-Klinikum, Department of Cardiology and Nephrology, Berlin, Germany
| | - Michaela Golic
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Berlin, Germany
- DZHK (German Center for Cardiovascular Research), Partner Site Berlin, Berlin, Germany
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
- Experimental and Clinical Research Center - a joint cooperation between the Max Delbrück Center for Molecular Medicine and the Charité - Universitätsmedizin Berlin, Berlin, Germany
- HSD Hochschule Döpfer, University of Applied Sciences, Cologne, Germany
| | - Kristin Kräker
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany.
- Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Berlin, Germany.
- DZHK (German Center for Cardiovascular Research), Partner Site Berlin, Berlin, Germany.
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany.
- Experimental and Clinical Research Center - a joint cooperation between the Max Delbrück Center for Molecular Medicine and the Charité - Universitätsmedizin Berlin, Berlin, Germany.
| |
Collapse
|
23
|
Influence of Hyperglycemia and Diabetes on Cardioprotection by Humoral Factors Released after Remote Ischemic Preconditioning (RIPC). Int J Mol Sci 2021; 22:ijms22168880. [PMID: 34445586 PMCID: PMC8396298 DOI: 10.3390/ijms22168880] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2021] [Revised: 08/11/2021] [Accepted: 08/16/2021] [Indexed: 02/07/2023] Open
Abstract
Remote ischemic preconditioning (RIPC) protects hearts from ischemia-reperfusion (I/R) injury in experimental studies; however, clinical RIPC trials were unsatisfactory. This discrepancy could be caused by a loss of cardioprotection due to comorbidities in patients, including diabetes mellitus (DM) and hyperglycemia (HG). RIPC is discussed to confer protective properties by release of different humoral factors activating cardioprotective signaling cascades. Therefore, we investigated whether DM type 1 and/or HG (1) inhibit the release of humoral factors after RIPC and/or (2) block the cardioprotective effect directly at the myocardium. Experiments were performed on male Wistar rats. Animals in part 1 of the study were either healthy normoglycemic (NG), type 1 diabetic (DM1), or hyperglycemic (HG). RIPC was implemented by four cycles of 5 min bilateral hind-limb ischemia/reperfusion. Control (Con) animals were not treated. Blood plasma taken in vivo was further investigated in isolated rat hearts in vitro. Plasma from diseased animals (DM1 or HG) was administered onto healthy (NG) hearts for 10 min before 33 min of global ischemia and 60 min of reperfusion. Part 2 of the study was performed vice versa-plasma taken in vivo, with or without RIPC, from healthy rats was transferred to DM1 and HG hearts in vitro. Infarct size was determined by TTC staining. Part 1: RIPC plasma from NG (NG Con: 49 ± 8% vs. NG RIPC 29 ± 6%; p < 0.05) and DM1 animals (DM1 Con: 47 ± 7% vs. DM1 RIPC: 38 ± 7%; p < 0.05) reduced infarct size. Interestingly, transfer of HG plasma showed comparable infarct sizes independent of prior treatment (HG Con: 34 ± 9% vs. HG RIPC 35 ± 9%; ns). Part 2: No infarct size reduction was detectable when transferring RIPC plasma from healthy rats to DM1 (DM1 Con: 54 ± 13% vs. DM1 RIPC 53 ± 10%; ns) or HG hearts (HG Con: 60 ± 16% vs. HG RIPC 53 ± 14%; ns). These results suggest that: (1) RIPC under NG and DM1 induces the release of humoral factors with cardioprotective impact, (2) HG plasma might own cardioprotective properties, and (3) RIPC does not confer cardioprotection in DM1 and HG myocardium.
Collapse
|
24
|
Xiong CF, Zhu QF, Chen YY, He DX, Feng YQ. Screening and Identification of Epoxy/Dihydroxy-Oxylipins by Chemical Labeling-Assisted Ultrahigh-Performance Liquid Chromatography Coupled with High-Resolution Mass Spectrometry. Anal Chem 2021; 93:9904-9911. [PMID: 34227808 DOI: 10.1021/acs.analchem.1c02058] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Epoxy/dihydroxy-oxylipins are important biologically active compounds that are mainly formed from polyunsaturated fatty acids (PUFAs) in the reactions catalyzed by the cytochrome P450 (CYP 450) enzyme. The analysis of epoxy/dihydroxy-oxylipins would be helpful to gain insights into their landscape in living organisms and provide a reference for the biological studies of these compounds. In this work, we employed chemical labeling-assisted liquid chromatography (LC) coupled with high-resolution mass spectrometry (CL-LC-HRMS) to establish a highly sensitive and specific method for screening and annotating epoxy/dihydroxy-oxylipins in biological samples. The isotope reagents 2-dimethylaminoethylamine (DMED) and DMED-d4 were employed to label epoxy/dihydroxy-oxylipins containing carboxyl groups so as to improve the analysis selectivity and MS detection sensitivity of epoxy/dihydroxy-oxylipins. Based on a pair of diagnostic ions with a mass-to-charge ratio (m/z) difference of 15.995 originating from the fragmentation of derivatives via high-energy collision dissociation (HCD), the potential epoxy/dihydroxy-oxylipins were rapidly screened from the complex matrix. Furthermore, the epoxy/dihydroxy groups could be readily localized by the diagnostic ion pairs, which enabled us to accurately annotate the epoxy/dihydroxy-oxylipins detected in biological samples. The applicability of our method was demonstrated by profiling epoxy/dihydroxy-oxylipins in human serum and heart samples from mice with high-fat diet (HFD). By the proposed method, a total of 32 and 62 potential epoxy/dihydroxy-oxylipins including 42 unreported ones were detected from human serum and the mice heart sample, respectively. Moreover, the relative quantitative results showed that most of the potential epoxy/dihydroxy-oxylipins, especially the oxidation products of linoleic acid (LA) or α-linolenic acid (ALA), were significantly decreased in the heart of mice with HFD. Our developed method is of high specificity and sensitivity and thus is a promising tool for the identification of novel epoxy/dihydroxy-oxylipins in biological samples.
Collapse
Affiliation(s)
- Cai-Feng Xiong
- Department of Chemistry, Wuhan University, Wuhan 430072, P. R. China
| | - Quan-Fei Zhu
- Department of Chemistry, Wuhan University, Wuhan 430072, P. R. China
| | - Yao-Yu Chen
- Department of Chemistry, Wuhan University, Wuhan 430072, P. R. China
| | - Dong-Xiao He
- Department of Chemistry, Wuhan University, Wuhan 430072, P. R. China
| | - Yu-Qi Feng
- Department of Chemistry, Wuhan University, Wuhan 430072, P. R. China.,Frontier Science Center for Immunology and Metabolism, Wuhan University, Wuhan 430072, P. R. China
| |
Collapse
|
25
|
Aravani D, Kassi E, Chatzigeorgiou A, Vakrou S. Cardiometabolic Syndrome: An Update on Available Mouse Models. Thromb Haemost 2021; 121:703-715. [PMID: 33280078 DOI: 10.1055/s-0040-1721388] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Cardiometabolic syndrome (CMS), a disease entity characterized by abdominal obesity, insulin resistance (IR), hypertension, and hyperlipidemia, is a global epidemic with approximately 25% prevalence in adults globally. CMS is associated with increased risk for cardiovascular disease (CVD) and development of diabetes. Due to its multifactorial etiology, the development of several animal models to simulate CMS has contributed significantly to the elucidation of the disease pathophysiology and the design of therapies. In this review we aimed to present the most common mouse models used in the research of CMS. We found that CMS can be induced either by genetic manipulation, leading to dyslipidemia, lipodystrophy, obesity and IR, or obesity and hypertension, or by administration of specific diets and drugs. In the last decade, the ob/ob and db/db mice were the most common obesity and IR models, whereas Ldlr-/- and Apoe-/- were widely used to induce hyperlipidemia. These mice have been used either as a single transgenic or combined with a different background with or without diet treatment. High-fat diet with modifications is the preferred protocol, generally leading to increased body weight, hyperlipidemia, and IR. A plethora of genetically engineered mouse models, diets, drugs, or synthetic compounds that are available have advanced the understanding of CMS. However, each researcher should carefully select the most appropriate model and validate its consistency. It is important to consider the differences between strains of the same animal species, different animals, and most importantly differences to human when translating results.
Collapse
Affiliation(s)
- Dimitra Aravani
- Department of Physiology, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Eva Kassi
- Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Antonios Chatzigeorgiou
- Department of Physiology, Medical School, National and Kapodistrian University of Athens, Athens, Greece
- Institute for Clinical Chemistry and Laboratory Medicine, University Hospital and Faculty of Medicine Carl Gustav Carus of TU Dresden, Dresden, Germany
| | - Styliani Vakrou
- Department of Physiology, Medical School, National and Kapodistrian University of Athens, Athens, Greece
- Department of Cardiology, "Laiko" General Hospital, Athens, Greece
| |
Collapse
|
26
|
Impact of Obesity-Induced Inflammation on Cardiovascular Diseases (CVD). Int J Mol Sci 2021; 22:ijms22094798. [PMID: 33946540 PMCID: PMC8125716 DOI: 10.3390/ijms22094798] [Citation(s) in RCA: 102] [Impact Index Per Article: 25.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2021] [Revised: 04/23/2021] [Accepted: 04/28/2021] [Indexed: 12/22/2022] Open
Abstract
Overweight and obesity are key risk factors of cardiovascular disease (CVD). Obesity is currently presented as a pro-inflammatory state with an expansion in the outflow of inflammatory cytokines, such as interleukin-6 (IL-6) and tumor necrosis factor-alpha (TNF-α), alongside the expanded emission of leptin. The present review aimed to evaluate the relationship between obesity and inflammation and their impacts on the development of cardiovascular disease. A literature search was conducted by employing three academic databases, namely PubMed (Medline), Scopus (EMBASE), and the Cumulative Index to Nursing and Allied Health Literature (CINAHL). The search presented 786 items, and by inclusion and exclusion filterers, 59 works were considered for final review. The Newcastle–Ottawa Scale (NOS) method was adopted to conduct quality assessment; 19 papers were further selected based on the quality score. Obesity-related inflammation leads to a low-grade inflammatory state in organisms by upregulating pro-inflammatory markers and downregulating anti-inflammatory cytokines, thereby contributing to cardiovascular disease pathogenesis. Because of inflammatory and infectious symptoms, adipocytes appear to instigate articulation and discharge a few intense stage reactants and carriers of inflammation. Obesity and inflammatory markers are strongly associated, and are important factors in the development of CVD. Hence, weight management can help prevent cardiovascular risks and poor outcomes by inhibiting inflammatory mechanisms.
Collapse
|
27
|
Tan Y, Li M, Wu G, Lou J, Feng M, Xu J, Zhou J, Zhang P, Yang H, Dong L, Li J, Zhang X, Gao F. Short-term but not long-term high fat diet feeding protects against pressure overload-induced heart failure through activation of mitophagy. Life Sci 2021; 272:119242. [PMID: 33607155 DOI: 10.1016/j.lfs.2021.119242] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2020] [Revised: 02/01/2021] [Accepted: 02/08/2021] [Indexed: 01/04/2023]
Abstract
AIMS Recent studies have shown that enhancement of fatty acid utilization through feeding animals a high fat diet (HFD) attenuated cardiac dysfunction in heart failure (HF). Here, we aimed to examine the temporal effects of HFD feeding on cardiac function in mice with heart failure and its underlying mechanism. MAIN METHODS Pressure overload-induced HF was established via transverse aortic constriction (TAC) surgery. After surgery, the mice were fed on either normal diet or HFD for 8 or 16 weeks. KEY FINDINGS HFD feeding exerted opposite effects on cardiac function at different time points post-surgery. Short-term HFD feeding (8 wk) protected the heart against pressure overload, inhibiting cardiac hypertrophy and improving cardiac function, while long-term HFD feeding (16 wk) aggravated cardiac dysfunction in TAC mice. Short-term HFD feeding elevated cardiac fatty acid utilization, while long-term HFD feeding showed no significant effects on cardiac fatty acid utilization in TAC mice. Specifically, an increase in cardiac fatty acid utilization was accompanied with activated mitophagy and improved mitochondrial function. Palmitic acid treatment (400 μM, 2 h) stimulated fatty acid oxidation and mitophagy in neonatal myocytes. Mechanistically, fatty acid utilization stimulated mitophagy through upregulation of Parkin. Cardiac-specific knockdown of Parkin abolished the protective effects of short-term HFD feeding on cardiac function in TAC mice. SIGNIFICANCES These results suggested that short-term but not long-term HFD feeding protects against pressure overload-induced heart failure through activation of mitophagy, and dietary fat intake should be used with caution in treatment of heart failure.
Collapse
Affiliation(s)
- Yanzhen Tan
- School of Aerospace Medicine, Fourth Military Medical University, Xi'an 710032, China; Department of Cardiovascular Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, China
| | - Min Li
- School of Aerospace Medicine, Fourth Military Medical University, Xi'an 710032, China
| | - Guiling Wu
- School of Aerospace Medicine, Fourth Military Medical University, Xi'an 710032, China
| | - Jing Lou
- School of Aerospace Medicine, Fourth Military Medical University, Xi'an 710032, China; School of Life Science and Technology and Frontier Institute of Science and Technology, Xi'an Jiaotong University, Xi'an 710049, China
| | - Mengya Feng
- School of Aerospace Medicine, Fourth Military Medical University, Xi'an 710032, China; School of Life Science and Technology and Frontier Institute of Science and Technology, Xi'an Jiaotong University, Xi'an 710049, China
| | - Jie Xu
- Department of Cardiology, 986(th) Hospital, Fourth Military Medical University, Xi'an 710032, China
| | - Jiaheng Zhou
- School of Aerospace Medicine, Fourth Military Medical University, Xi'an 710032, China
| | - Pengfei Zhang
- School of Aerospace Medicine, Fourth Military Medical University, Xi'an 710032, China
| | - Hongyan Yang
- School of Aerospace Medicine, Fourth Military Medical University, Xi'an 710032, China
| | - Ling Dong
- School of Aerospace Medicine, Fourth Military Medical University, Xi'an 710032, China
| | - Jia Li
- School of Aerospace Medicine, Fourth Military Medical University, Xi'an 710032, China
| | - Xing Zhang
- School of Aerospace Medicine, Fourth Military Medical University, Xi'an 710032, China.
| | - Feng Gao
- School of Aerospace Medicine, Fourth Military Medical University, Xi'an 710032, China
| |
Collapse
|
28
|
Hussain T, Tan B, Murtaza G, Metwally E, Yang H, Kalhoro MS, Kalhoro DH, Chughtai MI, Yin Y. Role of Dietary Amino Acids and Nutrient Sensing System in Pregnancy Associated Disorders. Front Pharmacol 2020; 11:586979. [PMID: 33414718 PMCID: PMC7783402 DOI: 10.3389/fphar.2020.586979] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2020] [Accepted: 11/23/2020] [Indexed: 12/16/2022] Open
Abstract
Defective implantation is related to pregnancy-associated disorders such as spontaneous miscarriage, intrauterine fetal growth restriction and others. Several factors proclaimed to be involved such as physiological, nutritional, environmental and managemental that leads to cause oxidative stress. Overloading of free radicals promotes oxidative stress, and the internal body system could not combat its ability to encounter the damaging effects and subsequently leading to pregnancy-related disorders. During pregnancy, essential amino acids display important role for optimum fetal growth and other necessary functions for continuing fruitful pregnancy. In this context, dietary amino acids have received much attention regarding the nutritional concerns during pregnancy. Arginine, glutamine, tryptophan and taurine play a crucial role in fetal growth, development and survival while ornithine and proline are important players for the regulation of gene expression, protein synthesis and angiogenesis. Moreover, amino acids also stimulate the mammalian target of rapamycin (mTOR) signaling pathway which plays a central role in the synthesis of proteins in placenta, uterus and fetus. This review article explores the significances of dietary amino acids in pregnancy development, regulation of nutrient-sensing pathways such as mTOR, peroxisome proliferator-activated receptors (PPARs), insulin/insulin-like growth factor signaling pathway (IIS) and 5' adenosine monophosphate-activated protein kinase (AMPK) which exhibit important role in reproduction and its related problems. In addition, the antioxidant function of dietary amino acids against oxidative stress triggering pregnancy disorders and their possible outcomes will also be enlightened. Dietary supplementation of amino acids during pregnancy could help mitigate reproductive disorders and thereby improving fertility in animals as well as humans.
Collapse
Affiliation(s)
- Tarique Hussain
- College of Animal Science and Technology, Hunan Agricultural University, Changsha, China
- Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, China
- Animal Sciences Division, Nuclear Institute for Agriculture and Biology College, Pakistan Institute of Engineering and Applied Sciences (NIAB-C,PIEAS), Faisalabad, Pakistan
| | - Bie Tan
- College of Animal Science and Technology, Hunan Agricultural University, Changsha, China
| | - Ghulam Murtaza
- Department of Animal Reproduction, Faculty of Animal Husbandry and Veterinary Sciences, Sindh Agriculture University, Sindh, Pakistan
| | - Elsayed Metwally
- Department of Cytology & Histology, Faculty of Veterinary Medicine, Suez Canal University, Ismailia, Egypt
| | - Huansheng Yang
- Hunan International Joint laboratory of Animal Intestinal Ecology and Health, Laboratory of Animal Nutrition and Human Health, College of Life Sciences, Hunan Normal University, Changsha, China
| | - Muhammad Saleem Kalhoro
- Department of Animal Products Technology, Faculty of Animal Husbandry and Veterinary Sciences, Sindh Agriculture University, Sindh, Pakistan
| | - Dildar Hussain Kalhoro
- Department of Veterinary Microbiology, Faculty of Animal Husbandry and Veterinary Sciences, Sindh Agriculture University, Sindh, Pakistan
| | - Muhammad Ismail Chughtai
- Animal Sciences Division, Nuclear Institute for Agriculture and Biology College, Pakistan Institute of Engineering and Applied Sciences (NIAB-C,PIEAS), Faisalabad, Pakistan
| | - Yulong Yin
- College of Animal Science and Technology, Hunan Agricultural University, Changsha, China
| |
Collapse
|
29
|
Penna C, Andreadou I, Aragno M, Beauloye C, Bertrand L, Lazou A, Falcão‐Pires I, Bell R, Zuurbier CJ, Pagliaro P, Hausenloy DJ. Effect of hyperglycaemia and diabetes on acute myocardial ischaemia-reperfusion injury and cardioprotection by ischaemic conditioning protocols. Br J Pharmacol 2020; 177:5312-5335. [PMID: 31985828 PMCID: PMC7680002 DOI: 10.1111/bph.14993] [Citation(s) in RCA: 69] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2019] [Revised: 11/19/2019] [Accepted: 01/09/2020] [Indexed: 12/12/2022] Open
Abstract
Diabetic patients are at increased risk of developing coronary artery disease and experience worse clinical outcomes following acute myocardial infarction. Novel therapeutic strategies are required to protect the myocardium against the effects of acute ischaemia-reperfusion injury (IRI). These include one or more brief cycles of non-lethal ischaemia and reperfusion prior to the ischaemic event (ischaemic preconditioning [IPC]) or at the onset of reperfusion (ischaemic postconditioning [IPost]) either to the heart or to extracardiac organs (remote ischaemic conditioning [RIC]). Studies suggest that the diabetic heart is resistant to cardioprotective strategies, although clinical evidence is lacking. We overview the available animal models of diabetes, investigating acute myocardial IRI and cardioprotection, experiments investigating the effects of hyperglycaemia on susceptibility to acute myocardial IRI, the response of the diabetic heart to cardioprotective strategies e.g. IPC, IPost and RIC. Finally we highlight the effects of anti-hyperglycaemic agents on susceptibility to acute myocardial IRI and cardioprotection. LINKED ARTICLES: This article is part of a themed issue on Risk factors, comorbidities, and comedications in cardioprotection. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v177.23/issuetoc.
Collapse
Affiliation(s)
- Claudia Penna
- Department of Clinical and Biological SciencesUniversity of TurinTurinItaly
| | - Ioanna Andreadou
- Laboratory of Pharmacology, Faculty of PharmacyNational and Kapodistrian University of AthensAthensGreece
| | - Manuela Aragno
- Department of Clinical and Biological SciencesUniversity of TurinTurinItaly
| | | | - Luc Bertrand
- Division of CardiologyCliniques Universitaires Saint‐LucBrusselsBelgium
- Pole of Cardiovascular Research, Institut de Recherche Experimetnale et CliniqueUCLouvainBrusselsBelgium
| | - Antigone Lazou
- School of BiologyAristotle University of ThessalonikiThessalonikiGreece
| | - Ines Falcão‐Pires
- Unidade de Investigação Cardiovascular, Departamento de Cirurgia e Fisiologia, Faculdade de MedicinaUniversidade do PortoPortoPortugal
| | - Robert Bell
- The Hatter Cardiovascular InstituteUniversity College LondonLondonUK
| | - Coert J. Zuurbier
- Laboratory of Experimental Intensive Care and Anesthesiology (L.E.I.C.A.), Department of Anesthesiology, Amsterdam UMCUniversity of Amsterdam, Cardiovascular SciencesAmsterdamThe Netherlands
| | - Pasquale Pagliaro
- Department of Clinical and Biological SciencesUniversity of TurinTurinItaly
| | - Derek J. Hausenloy
- The Hatter Cardiovascular InstituteUniversity College LondonLondonUK
- Cardiovascular and Metabolic Disorders ProgramDuke–NUS Medical SchoolSingapore
- National Heart Research Institute SingaporeNational Heart Centre SingaporeSingapore
- Yong Loo Lin School of MedicineNational University of SingaporeSingapore
- Cardiovascular Research Center, College of Medical and Health SciencesAsia UniversityTaiwan
| |
Collapse
|
30
|
Intralipid postconditioning in patients of cardiac surgery undergoing cardiopulmonary bypass (iCPB): study protocol for a randomized controlled trial. Trials 2020; 21:953. [PMID: 33228739 PMCID: PMC7686691 DOI: 10.1186/s13063-020-04854-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2020] [Accepted: 11/01/2020] [Indexed: 02/08/2023] Open
Abstract
Background Intralipid is a necessary fatty acid carrier that has been safely used as an energy supplier in the clinic. It has played an important role in rescuing the cardiac arrest caused by local anesthetic toxicity. In recent years, experimental studies have shown that intralipid postconditioning (ILPC) could reduce myocardial ischemic/reperfusion (I/R) injuries. Our research group has innovatively conducted a pilot randomized controlled trial (RCT), and the results showed that ILPC could reduce the release of cTnT and CK-MB, biomarkers of myocardial I/R injury, in valve replacement surgery. However, the potential effects of ILPC on the clinical outcome of adult cardiac surgery patients are unclear. Intralipid postconditioning in patients of cardiac surgery undergoing cardiopulmonary bypass (iCPB) trial is aimed to further study whether ILPC could improve short-term and long-term clinical outcome, as well as cardiac function in adult cardiac surgery patients. Methods The iCPB trial is an ongoing, single-center, prospective, double-blinded, large sample RCT. In total, 1000 adults undergoing cardiac surgery will be randomly allocated to either the ILPC group or the control group. The intervention group received an intravenous infusion of 2 mL/kg of 20% intralipid (medium-chain and long-chain fat emulsion injection C6~C24, Pharmaceutical) within 10 min before aortic cross-unclamping, and the control group received an equivalent volume of normal saline. The primary endpoints are complex morbidity of major complications during hospitalization and all-cause mortality within 30 days after surgery. The secondary endpoints include (1) all-cause mortality 6 months and 1 year postoperatively; (2) the quality of life within 1 year after surgery, using the QoR-15 questionnaire; (3) the postoperative cardiac function evaluated by LVEF, LVEDS, and LVEDD, and the myocardial injury evaluated by CK-MB, cTnT, and BNP; and (4) short-term clinical outcomes during hospitalization and total cost are also detailed evaluated. Discussion The iCPB trial is the first to explore ILPC on the clinical outcome of adult cardiac surgery patients. The results are expected to provide potential evidences about whether ILPC could reduce the morbidity and mortality and improve the cardiac function and quality of life. Therefore, the results will provide a rationale for the evaluation of the potentially clinically relevant benefit of intralipid therapy. Trial registration Chictr.org.cn ChiCTR1900024387. Prospectively registered on 9 July 2019.
Collapse
|
31
|
Liakh I, Sledzinski T, Kaska L, Mozolewska P, Mika A. Sample Preparation Methods for Lipidomics Approaches Used in Studies of Obesity. Molecules 2020; 25:E5307. [PMID: 33203044 PMCID: PMC7696154 DOI: 10.3390/molecules25225307] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Revised: 11/11/2020] [Accepted: 11/12/2020] [Indexed: 02/06/2023] Open
Abstract
Obesity is associated with alterations in the composition and amounts of lipids. Lipids have over 1.7 million representatives. Most lipid groups differ in composition, properties and chemical structure. These small molecules control various metabolic pathways, determine the metabolism of other compounds and are substrates for the syntheses of different derivatives. Recently, lipidomics has become an important branch of medical/clinical sciences similar to proteomics and genomics. Due to the much higher lipid accumulation in obese patients and many alterations in the compositions of various groups of lipids, the methods used for sample preparations for lipidomic studies of samples from obese subjects sometimes have to be modified. Appropriate sample preparation methods allow for the identification of a wide range of analytes by advanced analytical methods, including mass spectrometry. This is especially the case in studies with obese subjects, as the amounts of some lipids are much higher, others are present in trace amounts, and obese subjects have some specific alterations of the lipid profile. As a result, it is best to use a method previously tested on samples from obese subjects. However, most of these methods can be also used in healthy, nonobese subjects or patients with other dyslipidemias. This review is an overview of sample preparation methods for analysis as one of the major critical steps in the overall analytical procedure.
Collapse
Affiliation(s)
- Ivan Liakh
- Department of Pharmaceutical Biochemistry, Medical University of Gdansk, Debinki 1, 80-211 Gdansk, Poland; (I.L.); (T.S.); (P.M.)
- Department of Toxicology, Medical University of Gdańsk, Al. Gen. Hallera 107, 80-416 Gdańsk, Poland
| | - Tomasz Sledzinski
- Department of Pharmaceutical Biochemistry, Medical University of Gdansk, Debinki 1, 80-211 Gdansk, Poland; (I.L.); (T.S.); (P.M.)
| | - Lukasz Kaska
- Department of General, Endocrine and Transplant Surgery, Faculty of Medicine, Medical University of Gdansk, Smoluchowskiego 17, 80-214 Gdansk, Poland;
| | - Paulina Mozolewska
- Department of Pharmaceutical Biochemistry, Medical University of Gdansk, Debinki 1, 80-211 Gdansk, Poland; (I.L.); (T.S.); (P.M.)
| | - Adriana Mika
- Department of Pharmaceutical Biochemistry, Medical University of Gdansk, Debinki 1, 80-211 Gdansk, Poland; (I.L.); (T.S.); (P.M.)
- Department of Environmental Analysis, Faculty of Chemistry, University of Gdansk, Wita Stwosza 63, 80-308 Gdansk, Poland
| |
Collapse
|
32
|
Han L, Wu G, Feng C, Yang Y, Li B, Ge Y, Jiang Y, Shi Y, Le G. Dietary methionine restriction improves the impairment of cardiac function in middle-aged obese mice. Food Funct 2020; 11:1764-1778. [PMID: 32044910 DOI: 10.1039/c9fo02819f] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Dietary methionine restriction (MR) has been reported to extend lifespan, reduce obesity and decrease oxidative damage to mtDNA in the heart of rats, and increase endogenous hydrogen sulfide (H2S) production in the liver and blood. H2S has many potential benefits in the pathophysiology of the cardiovascular system. MR also increases the level of homocysteine (Hcy) in the liver and plasma, but elevated plasma Hcy is a risk factor for cardiovascular disease. Therefore, this study aimed to determine the effect of MR on cardiac function and metabolic status in obese middle-aged mice and its possible mechanisms. C57BL/6J mice (aged approximately 28 weeks) were divided into six dietary groups: CON (0.86% methionine + 4% fat), CMR40 (0.52% methionine + 4% fat), CMR80 (0.17% methionine + 4% fat), HFD (0.86% methionine + 24% fat), HMR40 (0.52% methionine + 24% fat) and HMR80 (0.17% methionine + 24% fat) for 15 consecutive weeks. Our results showed that 80% MR improves systolic dysfunction in middle-aged obese mice and enhances myocardial energy metabolism. 80% MR also reduces myocardial oxidative stress and improves inflammatory response. In addition, 80% MR increased mice Hcy levels and activated remethylation and transsulfur pathways of Hcy and promoted endogenous H2S production in the heart. 40% MR has the same trend, but is not significant. Moreover 40% MR at variance with 80% MR, did not decrease the body weight in both control and high-fat diet mice. These findings suggest that MR can improve myocardial energy metabolism, reduce heart inflammation and oxidative stress by increasing cardiac H2S production, and improve cardiac dysfunction in middle-aged obese mice.
Collapse
Affiliation(s)
- Le Han
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, China. and Center for Food Nutrition and Functional Food Engineering, School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, China
| | - Guoqin Wu
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, China. and Center for Food Nutrition and Functional Food Engineering, School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, China
| | - Chuanxin Feng
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, China. and Center for Food Nutrition and Functional Food Engineering, School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, China
| | - Yuhui Yang
- College of Food Science and Technology, Henan University of Technology, Zhengzhou, Henan 450001, China
| | - Bowen Li
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, China. and Center for Food Nutrition and Functional Food Engineering, School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, China
| | - Yueting Ge
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, China. and Center for Food Nutrition and Functional Food Engineering, School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, China
| | - Yuge Jiang
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, China. and Center for Food Nutrition and Functional Food Engineering, School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, China
| | - Yonghui Shi
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, China. and Center for Food Nutrition and Functional Food Engineering, School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, China
| | - Guowei Le
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, China. and Center for Food Nutrition and Functional Food Engineering, School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, China
| |
Collapse
|
33
|
Nguyen TD, Schulze PC. Lipid in the midst of metabolic remodeling - Therapeutic implications for the failing heart. Adv Drug Deliv Rev 2020; 159:120-132. [PMID: 32791076 DOI: 10.1016/j.addr.2020.08.004] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2020] [Revised: 08/07/2020] [Accepted: 08/07/2020] [Indexed: 02/07/2023]
Abstract
A healthy heart relies on an intact cardiac lipid metabolism. Fatty acids represent the major source for ATP production in the heart. Not less importantly, lipids are directly involved in critical processes such as cell growth, proliferation, and cell death by functioning as building blocks or signaling molecules. In the development of heart failure, perturbations in fatty acid utilization impair cardiac energetics. Furthermore, they may affect glucose and amino acid metabolism and induce the synthesis of several lipid intermediates, whose biological functions are still poorly understood. This work outlines the pivotal role of lipid metabolism in the heart and provides a lipocentric view of metabolic remodeling in heart failure. We will also critically revisit therapeutic attempts targeting cardiac lipid metabolism in heart failure and propose specific strategies for future investigations in this regard.
Collapse
|
34
|
Bianchi VE. Caloric restriction in heart failure: A systematic review. Clin Nutr ESPEN 2020; 38:50-60. [PMID: 32690177 DOI: 10.1016/j.clnesp.2020.04.009] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2020] [Accepted: 04/17/2020] [Indexed: 12/28/2022]
Abstract
BACKGROUND AND AIMS Nutrition exerts a determinant role in maintaining cardiac function, regulating insulin and mitochondrial efficiency, that are essential to support energy production for contractility. In patients with heart failure (HF), myocardial tissue efficiency is reduced because of decreased mitochondrial oxidative capacity. In HF conditions, cardiomyocytes shift toward glucose and a reduction in fatty acid utilization. Calorie restriction induces weight loss in obese patients and can be beneficial in some HF patients, although this has generated some controversy. This study aims to evaluate the impact of the CR diet on myocardial efficiency in HF patients. METHODS On Pubmed and Embase, articles related to the keywords: "chronic heart failure" with "diet," "nutrition," "insulin resistance," and "caloric restriction" have been searched, Studies, including exercise or food supplementation, were excluded. RESULTS The retrieved articles showed that weight loss, through the activation of insulin and various kinase pathways, regulates the efficiency of myocardial tissue. In contrast, insulin resistance represents a strong cardiovascular risk factor that reduces myocardial function. CONCLUSION CR diet represents the first therapy in overweight HF patients, both with preserved ejection fraction (HFpEF) and with reduced ejection fraction (HFrHF) because reducing body fat, the myocardial function increased. Insulin activity is the critical hormone that regulates mitochondrial function and cardiac efficiency. However, a severely restricted diet may represent a severe risk factor correlated with all-cause mortality, particularly in underweight HF patients. Long-term studies conducted on large populations are necessary to evaluate the effects of CR on myocardial function in HF patients.
Collapse
|
35
|
Heyne E, Schrepper A, Doenst T, Schenkl C, Kreuzer K, Schwarzer M. High-fat diet affects skeletal muscle mitochondria comparable to pressure overload-induced heart failure. J Cell Mol Med 2020; 24:6741-6749. [PMID: 32363733 PMCID: PMC7299710 DOI: 10.1111/jcmm.15325] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2020] [Revised: 04/02/2020] [Accepted: 04/03/2020] [Indexed: 01/01/2023] Open
Abstract
In heart failure, high-fat diet (HFD) may exert beneficial effects on cardiac mitochondria and contractility. Skeletal muscle mitochondrial dysfunction in heart failure is associated with myopathy. However, it is not clear if HFD affects skeletal muscle mitochondria in heart failure as well. To induce heart failure, we used pressure overload (PO) in rats fed normal chow or HFD. Interfibrillar mitochondria (IFM) and subsarcolemmal mitochondria (SSM) from gastrocnemius were isolated and functionally characterized. With PO heart failure, maximal respiratory capacity was impaired in IFM but increased in SSM of gastrocnemius. Unexpectedly, HFD affected mitochondria comparably to PO. In combination, PO and HFD showed additive effects on mitochondrial subpopulations which were reflected by isolated complex activities. While PO impaired diastolic as well as systolic cardiac function and increased glucose tolerance, HFD did not affect cardiac function but decreased glucose tolerance. We conclude that HFD and PO heart failure have comparable effects leading to more severe impairment of IFM. Glucose tolerance seems not causally related to skeletal muscle mitochondrial dysfunction. The additive effects of HFD and PO may suggest accelerated skeletal muscle mitochondrial dysfunction when heart failure is accompanied with a diet containing high fat.
Collapse
Affiliation(s)
- Estelle Heyne
- Department of Cardiothoracic Surgery, Jena University Hospital - Friedrich Schiller University of Jena, Jena, Germany
| | - Andrea Schrepper
- Department of Cardiothoracic Surgery, Jena University Hospital - Friedrich Schiller University of Jena, Jena, Germany
| | - Torsten Doenst
- Department of Cardiothoracic Surgery, Jena University Hospital - Friedrich Schiller University of Jena, Jena, Germany
| | - Christina Schenkl
- Department of Cardiothoracic Surgery, Jena University Hospital - Friedrich Schiller University of Jena, Jena, Germany
| | - Katrin Kreuzer
- Department of Cardiothoracic Surgery, Jena University Hospital - Friedrich Schiller University of Jena, Jena, Germany
| | - Michael Schwarzer
- Department of Cardiothoracic Surgery, Jena University Hospital - Friedrich Schiller University of Jena, Jena, Germany
| |
Collapse
|
36
|
Glatz JFC, Nabben M, Young ME, Schulze PC, Taegtmeyer H, Luiken JJFP. Re-balancing cellular energy substrate metabolism to mend the failing heart. Biochim Biophys Acta Mol Basis Dis 2020; 1866:165579. [PMID: 31678200 PMCID: PMC7586321 DOI: 10.1016/j.bbadis.2019.165579] [Citation(s) in RCA: 60] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2019] [Revised: 09/16/2019] [Accepted: 10/04/2019] [Indexed: 12/13/2022]
Abstract
Fatty acids and glucose are the main substrates for myocardial energy provision. Under physiologic conditions, there is a distinct and finely tuned balance between the utilization of these substrates. Using the non-ischemic heart as an example, we discuss that upon stress this substrate balance is upset resulting in an over-reliance on either fatty acids or glucose, and that chronic fuel shifts towards a single type of substrate appear to be linked with cardiac dysfunction. These observations suggest that interventions aimed at re-balancing a tilted substrate preference towards an appropriate mix of substrates may result in restoration of cardiac contractile performance. Examples of manipulating cellular substrate uptake as a means to re-balance fuel supply, being associated with mended cardiac function underscore this concept. We also address the molecular mechanisms underlying the apparent need for a fatty acid-glucose fuel balance. We propose that re-balancing cellular fuel supply, in particular with respect to fatty acids and glucose, may be an effective strategy to treat the failing heart.
Collapse
Affiliation(s)
- Jan F C Glatz
- Department of Genetics & Cell Biology, Faculty of Health, Medicine and Life Sciences (FHML), Maastricht University, Maastricht, the Netherlands.
| | - Miranda Nabben
- Department of Genetics & Cell Biology, Faculty of Health, Medicine and Life Sciences (FHML), Maastricht University, Maastricht, the Netherlands
| | - Martin E Young
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
| | - P Christian Schulze
- Department of Internal Medicine I, Division of Cardiology, Angiology, Pneumology and Intensive Medical Care, University Hospital Jena, Friedrich Schiller University Jena, Jena, Germany
| | - Heinrich Taegtmeyer
- Department of Internal Medicine, Division of Cardiology, McGovern Medical School at The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Joost J F P Luiken
- Department of Genetics & Cell Biology, Faculty of Health, Medicine and Life Sciences (FHML), Maastricht University, Maastricht, the Netherlands
| |
Collapse
|
37
|
Pakiet A, Jakubiak A, Mierzejewska P, Zwara A, Liakh I, Sledzinski T, Mika A. The Effect of a High-Fat Diet on the Fatty Acid Composition in the Hearts of Mice. Nutrients 2020; 12:nu12030824. [PMID: 32245049 PMCID: PMC7146498 DOI: 10.3390/nu12030824] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2020] [Revised: 03/17/2020] [Accepted: 03/18/2020] [Indexed: 12/11/2022] Open
Abstract
The Western diet can lead to alterations in cardiac function and increase cardiovascular risk, which can be reproduced in animal models by implementing a high-fat diet (HFD). However, the mechanism of these alterations is not fully understood and may be dependent on alterations in heart lipid composition. The aim of this study was to evaluate the effect of an HFD on the fatty acid (FA) composition of total lipids, as well as of various lipid fractions in the heart, and on heart function. C57BL/6 mice were fed an HFD or standard laboratory diet. The FA composition of chow, serum, heart and skeletal muscle tissues was measured by gas chromatography–mass spectrometry. Cardiac function was evaluated by ultrasonography. Our results showed an unexpected increase in polyunsaturated FAs (PUFAs) and a significant decrease in monounsaturated FAs (MUFAs) in the heart tissue of mice fed the HFD. For comparison, no such effects were observed in skeletal muscle or serum samples. Furthermore, we found that the largest increase in PUFAs was in the sphingolipid fraction, whereas the largest decrease in MUFAs was in the phospholipid and sphingomyelin fractions. The hearts of mice fed an HFD had an increased content of triacylglycerols. Moreover, the HFD treatment altered aortic flow pattern. We did not find significant changes in heart mass or oxidative stress markers between mice fed the HFD and standard diet. The above results suggest that alterations in FA composition in the heart may contribute to deterioration of heart function. A possible mechanism of this phenomenon is the alteration of sphingolipids and phospholipids in the fatty acid profile, which may change the physical properties of these lipids. Since phospho- and sphingolipids are the major components of cell membranes, alterations in their structures in heart cells can result in changes in cell membrane properties.
Collapse
Affiliation(s)
- Alicja Pakiet
- Department of Environmental Analysis, Faculty of Chemistry, University of Gdansk, Wita Stwosza 63, 80-308 Gdansk, Poland; (A.P.); (A.Z.)
| | - Agnieszka Jakubiak
- Tri-City Academic Laboratory Animal Centre - Research & Services Centre, Medical University of Gdansk, 80-210 Gdansk, Poland;
| | - Paulina Mierzejewska
- Department of Biochemistry, Faculty of Medicine, Medical University of Gdansk, Debinki 1, 80-211 Gdansk, Poland;
| | - Agata Zwara
- Department of Environmental Analysis, Faculty of Chemistry, University of Gdansk, Wita Stwosza 63, 80-308 Gdansk, Poland; (A.P.); (A.Z.)
| | - Ivan Liakh
- Department of Pharmaceutical Biochemistry, Faculty of Pharmacy, Medical University of Gdansk, Debinki 1, 80-211 Gdansk, Poland; (I.L.); (T.S.)
| | - Tomasz Sledzinski
- Department of Pharmaceutical Biochemistry, Faculty of Pharmacy, Medical University of Gdansk, Debinki 1, 80-211 Gdansk, Poland; (I.L.); (T.S.)
| | - Adriana Mika
- Department of Environmental Analysis, Faculty of Chemistry, University of Gdansk, Wita Stwosza 63, 80-308 Gdansk, Poland; (A.P.); (A.Z.)
- Department of Pharmaceutical Biochemistry, Faculty of Pharmacy, Medical University of Gdansk, Debinki 1, 80-211 Gdansk, Poland; (I.L.); (T.S.)
- Correspondence: ; Tel.: +48-585-230-810
| |
Collapse
|
38
|
Abstract
Patients with diabetes mellitus have >2× the risk for developing heart failure (HF; HF with reduced ejection fraction and HF with preserved ejection fraction). Cardiovascular outcomes, hospitalization, and prognosis are worse for patients with diabetes mellitus relative to those without. Beyond the structural and functional changes that characterize diabetic cardiomyopathy, a complex underlying, and interrelated pathophysiology exists. Despite the success of many commonly used antihyperglycemic therapies to lower hyperglycemia in type 2 diabetes mellitus the high prevalence of HF persists. This, therefore, raises the possibility that additional factors beyond glycemia might contribute to the increased HF risk in diabetes mellitus. This review summarizes the state of knowledge about the impact of existing antihyperglycemic therapies on HF and discusses potential mechanisms for beneficial or deleterious effects. Second, we review currently approved pharmacological therapies for HF and review evidence that addresses their efficacy in the context of diabetes mellitus. Dysregulation of many cellular mechanisms in multiple models of diabetic cardiomyopathy and in human hearts have been described. These include oxidative stress, inflammation, endoplasmic reticulum stress, aberrant insulin signaling, accumulation of advanced glycated end-products, altered autophagy, changes in myocardial substrate metabolism and mitochondrial bioenergetics, lipotoxicity, and altered signal transduction such as GRK (g-protein receptor kinase) signaling, renin angiotensin aldosterone signaling and β-2 adrenergic receptor signaling. These pathophysiological pathways might be amenable to pharmacological therapy to reduce the risk of HF in the context of type 2 diabetes mellitus. Successful targeting of these pathways could alter the prognosis and risk of HF beyond what is currently achieved using existing antihyperglycemic and HF therapeutics.
Collapse
Affiliation(s)
- Helena C Kenny
- From the Fraternal Order of Eagles Diabetes Research Center, and Division of Endocrinology and Metabolism, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City
| | - E Dale Abel
- From the Fraternal Order of Eagles Diabetes Research Center, and Division of Endocrinology and Metabolism, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City
| |
Collapse
|
39
|
Emelyanova L, Boukatina A, Myers C, Oyarzo J, Lustgarten J, Shi Y, Jahangir A. High calories but not fat content of lard-based diet contribute to impaired mitochondrial oxidative phosphorylation in C57BL/6J mice heart. PLoS One 2019; 14:e0217045. [PMID: 31265457 PMCID: PMC6605645 DOI: 10.1371/journal.pone.0217045] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2018] [Accepted: 05/05/2019] [Indexed: 11/24/2022] Open
Abstract
Purpose High calorie intake leads to obesity, a global socio-economic and health problem, reaching epidemic proportion in children and adolescents. Saturated and monounsaturated fatty acids from animal (lard) fat are major components of the western-pattern diet and its regular consumption leads to obesity, a risk factor for cardiovascular disease. However, no clear evidence exists whether consumption of diet rich in saturated (SFAs) and monounsaturated (MUFAs) fatty acids has detrimental effects on cardiac structure and energetics primarily due to excessive calories. We, therefore, sought to determine the impact of high calories versus fat content in diet on cardiac structure and mitochondrial energetics. Methods Six-week-old C57BL/6J mice were fed with high calorie, high lard fat-based diet (60% fat, HFD), high-calorie and low lard fat-based diet (10% fat, LFD), and lower-calorie and fat diet (standard chow, 12% fat, SCD) for 10 weeks. Results The HFD- and LFD-fed mice had higher body weight, ventricular mass and thickness of posterior and septal wall with increased cardiomyocytes diameter compared to the SCD-fed mice. These changes were associated with a reduction in the mitochondrial oxidative phosphorylation (OXPHOS) complexes I and III activity compared to the SCD-fed mice without significant differences between the HFD- and LFD-fed animals. The HFD-fed animals had higher level of malondialdehyde (MDA) than LFD and SCD-fed mice. Conclusions We assume that changes in cardiac morphology and selective reduction of the OXPHOS complexes activity observed in the HFD- and LFD-fed mice might be related to excessive calories with additional effect of fat content on oxidative stress.
Collapse
Affiliation(s)
- Larisa Emelyanova
- Center for Integrative Research on Cardiovascular Aging, Aurora St. Luke's Medical Center, Aurora Health Care, Milwaukee, Wisconsin, United States of America
| | - Anna Boukatina
- Mayo Clinic, Scottsdale, Arizona, United States of America
| | - Cheryl Myers
- Mayo Clinic, Scottsdale, Arizona, United States of America
| | - Janice Oyarzo
- Mayo Clinic, Scottsdale, Arizona, United States of America
| | | | - Yang Shi
- Center for Integrative Research on Cardiovascular Aging, Aurora St. Luke's Medical Center, Aurora Health Care, Milwaukee, Wisconsin, United States of America
| | - Arshad Jahangir
- Aurora Cardiovascular Services, Aurora Health Care, Milwaukee, Wisconsin, United States of America
| |
Collapse
|
40
|
Smithline HA, Donnino M, Blank FSJ, Barus R, Coute RA, Knee AB, Visintainer P. Supplemental thiamine for the treatment of acute heart failure syndrome: a randomized controlled trial. BMC COMPLEMENTARY AND ALTERNATIVE MEDICINE 2019; 19:96. [PMID: 31060559 PMCID: PMC6501378 DOI: 10.1186/s12906-019-2506-8] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/29/2018] [Accepted: 04/16/2019] [Indexed: 11/10/2022]
Abstract
Background The purpose of this pilot study was to determine if a definitive clinical trial of thiamine supplementation was warranted in patients with acute heart failure. We hypothesized that thiamine, when added to standard of care, would improve dyspnea (primary outcome) in hospitalized patients with acute heart failure. Peak expiratory flow rate, type B natriuretic peptide, free fatty acids, glucose, hospital length of stay, as well as 30-day rehospitalization and mortality were pre-planned secondary outcome measures. Methods This was a blinded experimental study at two urban academic hospitals. Consecutive patients admitted from the Emergency Department with a primary diagnosis of acute heart failure were recruited over 2 years. Patients on a daily dietary supplement were excluded. Randomization was stratified by type B natriuretic peptide and diabetes medication categories. Subjects received study drug (100 mg thiamine or placebo) in the evening of their first and second day. Outcome measures were obtained 8 h after study drug infusion. Dyspnea was measured on a 100-mm visual analog scale sitting up on oxygen, sitting up off oxygen, and lying supine off oxygen with 0 indicating no dyspnea. Data were analyzed using mixed-models as well as linear, negative binomial and logistic regression models to assess the impact of group on outcome measures. Results Of 130 subjects randomized, 118 had evaluable data (55 in the control and 63 in the treatment groups), 89% in both groups were adjudicated to have primarily AHF. Thiamine values increased significantly in the treatment group and were unchanged in the control group. One patient had thiamine deficiency. Only dyspnea measured sitting upright on oxygen differed significantly by group over time. No change was found for the other measures of dyspnea and all of the secondary measures. Conclusions In mild-moderate acute heart failure patients without thiamine deficiency, a standard dosing regimen of thiamine did not improve dyspnea, biomarkers, or other clinical parameters. Trial registration ClinicalTrials.gov: NCT00680706, May 20, 2008 (retrospectively registered). Electronic supplementary material The online version of this article (10.1186/s12906-019-2506-8) contains supplementary material, which is available to authorized users.
Collapse
|
41
|
Nakamura M, Sadoshima J. Cardiomyopathy in obesity, insulin resistance and diabetes. J Physiol 2019; 598:2977-2993. [PMID: 30869158 DOI: 10.1113/jp276747] [Citation(s) in RCA: 172] [Impact Index Per Article: 28.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2019] [Accepted: 02/25/2019] [Indexed: 12/17/2022] Open
Abstract
The prevalence of obesity, insulin resistance and diabetes is increasing rapidly. Most patients with these disorders have hypertriglyceridaemia and increased plasma levels of fatty acids, which are taken up and stored in lipid droplets in the heart. Intramyocardial lipids that exceed the capacity for storage and oxidation can be lipotoxic and induce non-ischaemic and non-hypertensive cardiomyopathy, termed diabetic or lipotoxic cardiomyopathy. The clinical features of diabetic cardiomyopathy are cardiac hypertrophy and diastolic dysfunction, which lead to heart failure, especially heart failure with preserved ejection fraction. Although the pathogenesis of the cardiomyopathy is multifactorial, diabetic dyslipidaemia and intramyocardial lipid accumulation are the key pathological features, triggering cellular signalling and modifications of proteins and lipids via generation of toxic metabolic intermediates. Most clinical studies have shown no beneficial effect of anti-diabetic agents and statins on outcomes in heart failure patients without atherosclerotic diseases, indicating the importance of identifying underlying mechanisms and early interventions for diabetic cardiomyopathy. Here, we summarize the molecular mechanisms of diabetic cardiomyopathy, with a special emphasis on cardiac lipotoxicity, and discuss the role of peroxisome proliferator-activated receptor α and dysregulated fatty acid metabolism as potential therapeutic targets.
Collapse
Affiliation(s)
- Michinari Nakamura
- Department of Cell Biology and Molecular Medicine, Cardiovascular Research Institute, Rutgers New Jersey Medical School, 185 South Orange Ave, Newark, NJ, 07103, USA
| | - Junichi Sadoshima
- Department of Cell Biology and Molecular Medicine, Cardiovascular Research Institute, Rutgers New Jersey Medical School, 185 South Orange Ave, Newark, NJ, 07103, USA
| |
Collapse
|
42
|
The effect of diet, lifestyle and psychological factors in the prognosis of ischemic heart failure. Metabol Open 2019; 1:11-18. [PMID: 32812917 PMCID: PMC7424785 DOI: 10.1016/j.metop.2019.03.001] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2019] [Revised: 03/02/2019] [Accepted: 03/04/2019] [Indexed: 12/28/2022] Open
Abstract
Background/Objective Dietary patterns may play an important role in the prognosis of heart failure. Methods Dietary habits, sleeping habits, physical activity and anxiety and depression status were recorded in 326 patients (90 females, mean age 73.45 ± 10.9 years) with ischemic heart failure prospectively followed for 30 months. Results Lower ΗADS-depression scores (p = 0.03), a low-fat meat diet (p = 0.035) and moderate coffee consumption (p = 0.005) were associated with better prognosis. Non-significant differences were recorded in outcomes with regard to consumption of other dietary categories. Conclusions A balanced diet as well as emphasis on the treatment of depression may improve outcomes in ischemic heart failure. Coffee consumption is associated with better outcomes in ischemic heart failure. A low fat meat diet is associated with better outcomes in ischemic heart failure. High ΗADS depression score is related to worst prognosis in ischemic heart failure. Sedentary lifestyle is related to worst outcomes in ischemic heart failure.
Collapse
|
43
|
Chen D, Li X, Zhang L, Zhu M, Gao L. A high-fat diet impairs mitochondrial biogenesis, mitochondrial dynamics, and the respiratory chain complex in rat myocardial tissues. J Cell Biochem 2018; 119:9602. [PMID: 30171706 PMCID: PMC6220867 DOI: 10.1002/jcb.27068] [Citation(s) in RCA: 87] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2017] [Accepted: 04/23/2018] [Indexed: 12/15/2022]
Abstract
A high‐fat diet (HFD) has been associated with heart failure and arrhythmias; however, the molecular mechanisms underlying these associations are poorly understood. The mitochondria play an essential role in optimal heart performance, most of the energy for which is obtained from the oxidation of fatty acids. As such, chronic exposure to excess fatty acids may cause mitochondrial dysfunction and heart failure. To investigate the effects of a HFD on the mitochondrial function in the myocardium, 40 male rats were randomly divided into two groups and fed with either a normal diet or a HFD for 28 weeks. The myocardial lipid content, cardiac parameters and function, and mitochondrial morphology and function were evaluated. The expression of a number of genes involved in mitochondrial dynamics was measured using quantitative polymerase chain reaction and Western blot analyses. Proteomic analysis was also performed to identify the proteins affected by HFD treatment. Significant fat deposition in the myocardia, cardiac hypertrophy, and cardiac dysfunction were all observed in HFD‐treated rats. Electron microscopy showed abnormal mitochondrial density and morphology. In addition, abnormal expression of genes involved in mitochondrial dynamics, decreased mitochondrial DNA copy numbers, reduced complex I‐III and citrate synthase activities, and decreased mitochondrial respiration were observed in HFD‐treated rats. High performance liquid chromatography showed downregulated adenosine triphosphate (ATP) and adenosine diphosphate levels and an increased adenosine monophosphate (AMP)/ATP ratio. Proteomic analysis confirmed the alteration of mitochondrial function and impaired expression of proteins involved in mitochondrial dynamics in HFD‐treated rats. Mitochondrial dysfunction and impaired mitochondrial dynamics play an important role in heart dysfunction induced by a HFD, thus presenting a potential therapeutic target for the treatment of heart disease.
Collapse
Affiliation(s)
- Dan Chen
- Department of Endocrinology, Shandong Provincial Hospital Affiliated to Shandong University, Shandong Provincial Key Laboratory of Endocrinology and Lipid Metabolism, Institute of Endocrinology and Metabolism, Shandong Academy of Clinical Medicine, Jinan, China.,Department of Electrocardiographic, Shandong Provincial Hospital affiliated to Shandong University, Jinan, China
| | - Xia Li
- Department of Ultrasound, Shandong Provincial Hospital affiliated to Shandong University, Jinan, China
| | - LiTing Zhang
- Department of Electrocardiographic, Shandong Provincial Hospital affiliated to Shandong University, Jinan, China
| | - Mei Zhu
- Department of Ultrasound, Shandong Provincial Hospital affiliated to Shandong University, Jinan, China
| | - Ling Gao
- Department of Endocrinology, Shandong Provincial Hospital Affiliated to Shandong University, Shandong Provincial Key Laboratory of Endocrinology and Lipid Metabolism, Institute of Endocrinology and Metabolism, Shandong Academy of Clinical Medicine, Jinan, China.,Department of Electrocardiographic, Shandong Provincial Hospital affiliated to Shandong University, Jinan, China
| |
Collapse
|
44
|
Guo Y, Wang Z, Qin X, Xu J, Hou Z, Yang H, Mao X, Xing W, Li X, Zhang X, Gao F. Enhancing fatty acid utilization ameliorates mitochondrial fragmentation and cardiac dysfunction via rebalancing optic atrophy 1 processing in the failing heart. Cardiovasc Res 2018; 114:979-991. [PMID: 29490017 DOI: 10.1093/cvr/cvy052] [Citation(s) in RCA: 50] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/01/2017] [Accepted: 02/24/2018] [Indexed: 09/12/2024] Open
Abstract
AIMS Heart failure (HF) is characterized by reduced fatty acid (FA) utilization associated with mitochondrial dysfunction. Recent evidence has shown that enhancing FA utilization may provide cardioprotection against HF. Our aim was to investigate the effects and the underlying mechanisms of cardiac FA utilization on cardiac function in response to pressure overload. METHODS AND RESULTS Transverse aortic constriction (TAC) was used in C57 mice to establish pressure overload-induced HF. TAC mice fed on a high fat diet (HFD) exhibited increased cardiac FA utilization and improved cardiac function and survival compared with those on control diet. Such cardioprotection could also be provided by cardiac-specific overexpression of CD36. Notably, both HFD and CD36 overexpression attenuated mitochondrial fragmentation and improved mitochondrial function in the failing heart. Pressure overload decreased ATP-dependent metalloprotease (YME1L) expression and induced the proteolytic cleavage of the dynamin-like guanosine triphosphatase OPA1 as a result of suppressed FA utilization. Enhancing FA utilization upregulated YME1L expression and subsequently rebalanced OPA1 processing, resulting in restoration of mitochondrial morphology in the failing heart. In addition, cardiac-specific overexpression of YME1L exerted similar cardioprotective effects against HF to those provided by HFD or CD36 overexpression. CONCLUSIONS These findings demonstrate that enhancing FA utilization ameliorates mitochondrial fragmentation and cardiac dysfunction via rebalancing OPA1 processing in pressure overload-induced HF, suggesting a unique metabolic intervention approach to improving cardiac functions in HF.
Collapse
MESH Headings
- Animals
- CD36 Antigens/genetics
- CD36 Antigens/metabolism
- Cells, Cultured
- Diet, High-Fat
- Disease Models, Animal
- Energy Metabolism
- Fatty Acids/metabolism
- GTP Phosphohydrolases/genetics
- GTP Phosphohydrolases/metabolism
- Heart Failure/diet therapy
- Heart Failure/metabolism
- Heart Failure/pathology
- Heart Failure/physiopathology
- Male
- Metalloendopeptidases/genetics
- Metalloendopeptidases/metabolism
- Mice, Inbred C57BL
- Mitochondria, Heart/metabolism
- Mitochondria, Heart/pathology
- Myocytes, Cardiac/metabolism
- Myocytes, Cardiac/pathology
- Proteolysis
- Rats, Sprague-Dawley
- Ventricular Function, Left
Collapse
Affiliation(s)
- Yongzheng Guo
- School of Aerospace Medicine, Fourth Military Medical University, 17 Changlexi Road, Xi'an 710032, China and
| | - Zhen Wang
- School of Aerospace Medicine, Fourth Military Medical University, 17 Changlexi Road, Xi'an 710032, China and
| | - Xinghua Qin
- School of Aerospace Medicine, Fourth Military Medical University, 17 Changlexi Road, Xi'an 710032, China and
| | - Jie Xu
- School of Aerospace Medicine, Fourth Military Medical University, 17 Changlexi Road, Xi'an 710032, China and
| | - Zuoxu Hou
- School of Aerospace Medicine, Fourth Military Medical University, 17 Changlexi Road, Xi'an 710032, China and
| | - Hongyan Yang
- School of Aerospace Medicine, Fourth Military Medical University, 17 Changlexi Road, Xi'an 710032, China and
| | - Xuechao Mao
- School of Aerospace Medicine, Fourth Military Medical University, 17 Changlexi Road, Xi'an 710032, China and
| | - Wenjuan Xing
- School of Aerospace Medicine, Fourth Military Medical University, 17 Changlexi Road, Xi'an 710032, China and
| | - Xiaoliang Li
- Department of Emergency Medicine, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, China
| | - Xing Zhang
- School of Aerospace Medicine, Fourth Military Medical University, 17 Changlexi Road, Xi'an 710032, China and
| | - Feng Gao
- School of Aerospace Medicine, Fourth Military Medical University, 17 Changlexi Road, Xi'an 710032, China and
| |
Collapse
|
45
|
Ribeiro Junior RF, Dabkowski ER, Shekar KC, O Connell KA, Hecker PA, Murphy MP. MitoQ improves mitochondrial dysfunction in heart failure induced by pressure overload. Free Radic Biol Med 2018; 117:18-29. [PMID: 29421236 PMCID: PMC5866124 DOI: 10.1016/j.freeradbiomed.2018.01.012] [Citation(s) in RCA: 118] [Impact Index Per Article: 16.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/31/2017] [Revised: 01/03/2018] [Accepted: 01/10/2018] [Indexed: 01/01/2023]
Abstract
Heart failure remains a major public-health problem with an increase in the number of patients worsening from this disease. Despite current medical therapy, the condition still has a poor prognosis. Heart failure is complex but mitochondrial dysfunction seems to be an important target to improve cardiac function directly. Our goal was to analyze the effects of MitoQ (100 µM in drinking water) on the development and progression of heart failure induced by pressure overload after 14 weeks. The main findings are that pressure overload-induced heart failure in rats decreased cardiac function in vivo that was not altered by MitoQ. However, we observed a reduction in right ventricular hypertrophy and lung congestion in heart failure animals treated with MitoQ. Heart failure also decreased total mitochondrial protein content, mitochondrial membrane potential in the intermyofibrillar mitochondria. MitoQ restored membrane potential in IFM but did not restore mitochondrial protein content. These alterations are associated with the impairment of basal and stimulated mitochondrial respiration in IFM and SSM induced by heart failure. Moreover, MitoQ restored mitochondrial respiration in heart failure induced by pressure overload. We also detected higher levels of hydrogen peroxide production in heart failure and MitoQ restored the increase in ROS production. MitoQ was also able to improve mitochondrial calcium retention capacity, mainly in the SSM whereas in the IFM we observed a small alteration. In summary, MitoQ improves mitochondrial dysfunction in heart failure induced by pressure overload, by decreasing hydrogen peroxide formation, improving mitochondrial respiration and improving mPTP opening.
Collapse
Affiliation(s)
- Rogério Faustino Ribeiro Junior
- Division of Cardiology, Department of Medicine, University of Maryland, Baltimore, MD, USA; Department of Physiological Sciences, Federal University of Espirito Santo, Vitoria, ES, Brazil.
| | - Erinne Rose Dabkowski
- Division of Cardiology, Department of Medicine, University of Maryland, Baltimore, MD, USA
| | | | - Kelly A O Connell
- Division of Cardiology, Department of Medicine, University of Maryland, Baltimore, MD, USA
| | - Peter A Hecker
- Division of Cardiology, Department of Medicine, University of Maryland, Baltimore, MD, USA
| | - Michael P Murphy
- Medical Research Council Mitochondrial Biology Unit, Cambridge BioMedical Campus, Cambridge, UK
| |
Collapse
|
46
|
Dasagrandhi D, R ASK, Muthuswamy A, Lennox AM, Jayavelu T, Devanathan V, Kesavan Swaminathan J. Ischemia/reperfusion injury in male guinea pigs: An efficient model to investigate myocardial damage in cardiovascular complications. Biomed Pharmacother 2018; 99:469-479. [PMID: 29665648 DOI: 10.1016/j.biopha.2018.01.087] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2017] [Revised: 12/20/2017] [Accepted: 01/12/2018] [Indexed: 12/28/2022] Open
Abstract
Myocardial ischemia/reperfusion (I/R) injury is the major problem that aggravates cardiac damage. Several established animal models fail to explain the similarity in disease mechanism and progression as seen in humans; whereas guinea pig shows high similarity in cardiovascular parameters. Hence, current study is aimed to develop an animal model using guinea pigs that may best correlate with disease mechanism of human myocardial I/R injury. Male guinea pigs were randomized into three groups: normal diet (ND), high fat diet (HFD) and sham; fed with respective diets for 90 days. Myocardial infarction (MI) was induced by ligating left anterior descending artery (LAD) for 30 min followed by 24 h and 7 days of reperfusion in ND and HFD groups. Electrocardiogram (ECG) showed the alterations in electrical conduction during myocardial I/R injury. Elevated levels of lactate dehydrogenase (LDH) and creatine kinase-MB ((CK-MB)) were higher in HFD compared to ND. Inflammatory markers such as tumor necrosis factor-α (TNF-α) and interleukin-6 (IL-6) were up-regulated in I/R injury animals compared to sham. Fold change of these protein expression levels were higher in HFD compared to ND. Elevated lipid profile and increased aortic wall thickness in HFD animals depicts the risk of developing cardiovascular complications. ECG analysis strongly confirmed MI through changes in sinus rhythm that are reflected in infarcted tissue as verified through TTC staining. Thus the combination of HFD followed by I/R injury proved to be an efficient model to study pathophysiology of myocardial I/R injury with minimal tissue damage and surgical mortality.
Collapse
Affiliation(s)
- Divya Dasagrandhi
- Drug Discovery and Molecular Cardiology Laboratory, Department of Bioinformatics, Bharathidasan University, Tiruchirappalli, 620024, India
| | - Arul Salomee Kamalabai R
- Drug Discovery and Molecular Cardiology Laboratory, Department of Bioinformatics, Bharathidasan University, Tiruchirappalli, 620024, India
| | - Anusuyadevi Muthuswamy
- Molecular Gerontology Laboratory, Department of Biochemistry, Bharathidasan University, Tiruchirappalli, 620024, India
| | - Angela Marie Lennox
- Department of Veterinary Clinical Sciences, Purdue University College of Veterinary Medicine, West Lafayette, IN 47907, United States
| | | | - Vasudharani Devanathan
- Department of Biological Sciences, Indian Institute of Science Education and Research, Tirupati, 517507, India
| | - Jayachandran Kesavan Swaminathan
- Drug Discovery and Molecular Cardiology Laboratory, Department of Bioinformatics, Bharathidasan University, Tiruchirappalli, 620024, India.
| |
Collapse
|
47
|
SIRT3: A New Regulator of Cardiovascular Diseases. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2018; 2018:7293861. [PMID: 29643974 PMCID: PMC5831850 DOI: 10.1155/2018/7293861] [Citation(s) in RCA: 125] [Impact Index Per Article: 17.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 11/01/2017] [Revised: 12/20/2017] [Accepted: 01/04/2018] [Indexed: 01/13/2023]
Abstract
Cardiovascular diseases (CVDs) are the leading causes of death worldwide, and defects in mitochondrial function contribute largely to the occurrence of CVDs. Recent studies suggest that sirtuin 3 (SIRT3), the mitochondrial NAD+-dependent deacetylase, may regulate mitochondrial function and biosynthetic pathways such as glucose and fatty acid metabolism and the tricarboxylic acid (TCA) cycle, oxidative stress, and apoptosis by reversible protein lysine deacetylation. SIRT3 regulates glucose and lipid metabolism and maintains myocardial ATP levels, which protects the heart from metabolic disturbances. SIRT3 can also protect cardiomyocytes from oxidative stress-mediated cell damage and block the development of cardiac hypertrophy. Recent reports show that SIRT3 is involved in the protection of several heart diseases. This review discusses the progress in SIRT3-related research and the role of SIRT3 in the prevention and treatment of CVDs.
Collapse
|
48
|
Carpentier AC. Abnormal Myocardial Dietary Fatty Acid Metabolism and Diabetic Cardiomyopathy. Can J Cardiol 2018; 34:605-614. [PMID: 29627307 DOI: 10.1016/j.cjca.2017.12.029] [Citation(s) in RCA: 51] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2017] [Revised: 12/08/2017] [Accepted: 12/19/2017] [Indexed: 12/13/2022] Open
Abstract
Patients with diabetes are at very high risk of hospitalization and death from heart failure. Increased prevalence of coronary heart disease, hypertension, autonomic neuropathy, and kidney failure all play a role in this increased risk. However, cardiac metabolic abnormalities are now recognized to play a role in this increased risk. Increased reliance on fatty acids to produce energy might predispose the diabetic heart to oxidative stress and ischemic damage. Intramyocellular accumulation of toxic lipid metabolites leads to a number of cellular abnormalities that might also contribute to cardiac remodelling and cardiac dysfunction. However, fatty acid availability from circulation and from intracellular lipid droplets to fuel the heart is critical to maintain its function. Fatty acids delivery to the heart is very complex and includes plasma nonesterified fatty acid flux as well as triglyceride-rich lipoprotein-mediated transport. Although many studies have shown a cross-sectional association between enhanced fatty acid delivery to the heart and reduction in left ventricular function in subjects with prediabetes and diabetes, these mechanisms change very rapidly during type 2 diabetes treatment. The present review focuses on the role of fatty acids in cardiac function, with particular emphasis on the possible role of early abnormalities of dietary fatty acid metabolism in the development of diabetic cardiomyopathy.
Collapse
Affiliation(s)
- André C Carpentier
- Division of Endocrinology, Department of Medicine, Centre de recherche du CHUS, Université de Sherbrooke, Sherbrooke, Québec, Canada.
| |
Collapse
|
49
|
Ribeiro Júnior RF, Ronconi KS, Jesus ICG, Almeida PWM, Forechi L, Vassallo DV, Guatimosim S, Stefanon I, Fernandes AA. Testosterone deficiency prevents left ventricular contractility dysfunction after myocardial infarction. Mol Cell Endocrinol 2018; 460:14-23. [PMID: 28606867 DOI: 10.1016/j.mce.2017.06.011] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/06/2016] [Revised: 05/17/2017] [Accepted: 06/08/2017] [Indexed: 11/30/2022]
Abstract
Testosterone may affect myocardial contractility since its deficiency decreases the contraction and relaxation of the heart. Meanwhile, testosterone replacement therapy has raised concerns because it may worsen cardiac dysfunction and remodeling after myocardial infarction (MI). In this study, we evaluate cardiac contractility 60 days after MI in rats with suppressed testosterone. Male Wistar rats underwent bilateral orchidectomy one week before the ligation of the anterior descending left coronary artery. The animals were divided into orchidectomized (OCT); MI; orchidectomized + MI (OCT + MI); orchidectomized + MI + testosterone (OCT + MI + T) and control (Sham) groups. Eight weeks after MI, papillary muscle contractility was analyzed under increasing calcium (0.62, 1.25, 2.5 and 3.75 mM) and isoproterenol (10-8 to 10-2 M) concentrations. Ventricular myocytes were isolated for intracellular calcium measurements and assessment of Ca2+ handling proteins. Contractility was preserved in the orchidectomized animals after myocardial infarction and was reduced when testosterone was replaced (Ca2+ 3.75 mM: Sham: 608 ± 70 (n = 11); OCT: 590 ± 37 (n = 16); MI: 311 ± 33* (n = 9); OCT + MI: 594 ± 76 (n = 7); OCT + MI + T: 433 ± 38* (n=4), g/g *p < 0.05 vs Sham). Orchidectomy also increased the Ca2+ transient amplitude of the ventricular myocytes and SERCA-2a protein expression levels. PLB phosphorylation levels at Thr17 were not different in the orchidectomized animals compared to the Sham animals but were reduced after testosterone replacement. CAMKII phosphorylation and protein nitrosylation increased in the orchidectomized animals. Our results support the view that testosterone deficiency prevents MI contractility dysfunction by altering the key proteins involved in Ca2+ handling.
Collapse
Affiliation(s)
- R F Ribeiro Júnior
- Department of Physiological Sciences, Universidade Federal do Espírito Santo, Vitória, ES, Brazil.
| | - K S Ronconi
- Department of Physiological Sciences, Universidade Federal do Espírito Santo, Vitória, ES, Brazil
| | - I C G Jesus
- Department of Physiology and Biophysics, Universidade Federal de Minas Gerais, Minas Gerais, MG, Brazil
| | - P W M Almeida
- Department of Physiology and Biophysics, Universidade Federal de Minas Gerais, Minas Gerais, MG, Brazil
| | - L Forechi
- Department of Physiological Sciences, Universidade Federal do Espírito Santo, Vitória, ES, Brazil
| | - D V Vassallo
- Department of Physiological Sciences, Universidade Federal do Espírito Santo, Vitória, ES, Brazil
| | - S Guatimosim
- Department of Physiology and Biophysics, Universidade Federal de Minas Gerais, Minas Gerais, MG, Brazil
| | - I Stefanon
- Department of Physiological Sciences, Universidade Federal do Espírito Santo, Vitória, ES, Brazil
| | - A A Fernandes
- Department of Physiological Sciences, Universidade Federal do Espírito Santo, Vitória, ES, Brazil
| |
Collapse
|
50
|
Boudia D, Domergue V, Mateo P, Fazal L, Prud'homme M, Prigent H, Delcayre C, Cohen-Solal A, Garnier A, Ventura-Clapier R, Samuel JL. Beneficial effects of exercise training in heart failure are lost in male diabetic rats. J Appl Physiol (1985) 2017; 123:1579-1591. [PMID: 28883044 DOI: 10.1152/japplphysiol.00117.2017] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Exercise training has been demonstrated to have beneficial effects in patients with heart failure (HF) or diabetes. However, it is unknown whether diabetic patients with HF will benefit from exercise training. Male Wistar rats were fed either a standard (Sham, n = 53) or high-fat, high-sucrose diet ( n = 66) for 6 mo. After 2 mo of diet, the rats were already diabetic. Rats were then randomly subjected to either myocardial infarction by coronary artery ligation (MI) or sham operation. Two months later, heart failure was documented by echocardiography and animals were randomly subjected to exercise training with treadmill for an additional 8 wk or remained sedentary. At the end, rats were euthanized and tissues were assayed by RT-PCR, immunoblotting, spectrophotometry, and immunohistology. MI induced a similar decrease in ejection fraction in diabetic and lean animals but a higher premature mortality in the diabetic group. Exercise for 8 wk resulted in a higher working power developed by MI animals with diabetes and improved glycaemia but not ejection fraction or pathological phenotype. In contrast, exercise improved the ejection fraction and increased adaptive hypertrophy after MI in the lean group. Trained diabetic rats with MI were nevertheless able to develop cardiomyocyte hypertrophy but without angiogenic responses. Exercise improved stress markers and cardiac energy metabolism in lean but not diabetic-MI rats. Hence, following HF, the benefits of exercise training on cardiac function are blunted in diabetic animals. In conclusion, exercise training only improved the myocardial profile of infarcted lean rats fed the standard diet. NEW & NOTEWORTHY Exercise training is beneficial in patients with heart failure (HF) or diabetes. However, less is known of the possible benefit of exercise training for HF patients with diabetes. Using a rat model where both diabetes and MI had been induced, we showed that 2 mo after MI, 8 wk of exercise training failed to improve cardiac function and metabolism in diabetic animals in contrast to lean animals.
Collapse
Affiliation(s)
- Dalila Boudia
- UMR-S 942 Institut National de la Santé et de la Recherche Médicale (INSERM), Université Paris-Diderot, Sorbonne Paris Cité, France
| | - Valérie Domergue
- UMS IPSIT Animex Platform, Université Paris-Sud, Université Paris-Saclay, Châtenay-Malabry, France
| | - Philippe Mateo
- UMR-S 1180 INSERM, Université Paris-Sud, Université Paris-Saclay, Châtenay-Malabry, France
| | - Loubina Fazal
- UMR-S 942 Institut National de la Santé et de la Recherche Médicale (INSERM), Université Paris-Diderot, Sorbonne Paris Cité, France
| | - Mathilde Prud'homme
- UMR-S 942 Institut National de la Santé et de la Recherche Médicale (INSERM), Université Paris-Diderot, Sorbonne Paris Cité, France
| | - Héloïse Prigent
- UMR-S 942 Institut National de la Santé et de la Recherche Médicale (INSERM), Université Paris-Diderot, Sorbonne Paris Cité, France.,Cardiology, Assistance Publique-Hópitaux de Paris (AP-HP), Ambroise Paré, Paris
| | - Claude Delcayre
- UMR-S 942 Institut National de la Santé et de la Recherche Médicale (INSERM), Université Paris-Diderot, Sorbonne Paris Cité, France
| | - Alain Cohen-Solal
- UMR-S 942 Institut National de la Santé et de la Recherche Médicale (INSERM), Université Paris-Diderot, Sorbonne Paris Cité, France.,Cardiology, Assistance Publique-Hópitaux de Paris (AP-HP), Ambroise Paré, Paris
| | - Anne Garnier
- UMR-S 1180 INSERM, Université Paris-Sud, Université Paris-Saclay, Châtenay-Malabry, France
| | - Renée Ventura-Clapier
- UMR-S 1180 INSERM, Université Paris-Sud, Université Paris-Saclay, Châtenay-Malabry, France
| | - Jane-Lise Samuel
- UMR-S 942 Institut National de la Santé et de la Recherche Médicale (INSERM), Université Paris-Diderot, Sorbonne Paris Cité, France
| |
Collapse
|