Acessibilidade / Reportar erro

Crossroads between Estrogen Loss, Obesity, and Heart Failure with Preserved Ejection Fraction

Abstract

The prevalence of obesity and heart failure with preserved ejection fraction (HFpEF) increases significantly in postmenopausal women. Although obesity is a risk factor for left ventricular diastolic dysfunction (LVDD), the mechanisms that link the cessation of ovarian hormone production, and particularly estrogens, to the development of obesity, LVDD, and HFpEF in aging females are unclear. Clinical, and epidemiologic studies show that postmenopausal women with abdominal obesity (defined by waist circumference) are at greater risk for developing HFpEF than men or women without abdominal obesity. The study presents a review of clinical data that support a mechanistic link between estrogen loss plus obesity and left ventricular remodeling with LVDD. It also seeks to discuss potential cell and molecular mechanisms for estrogen-mediated protection against adverse adipocyte cell types, tissue depots, function, and metabolism that may contribute to LVDD and HFpEF.

Estrogens; Obesity; Heart Failure; Stroke Volume; Menopause; Adiposity; Overweight; Echocardiography/methods; Body Mass Index

Resumo

A prevalência de obesidade e insuficiência cardíaca com fração de ejeção preservada (ICFEP) aumenta significativamente em mulheres na pós-menopausa. Embora a obesidade seja um fator de risco para disfunção diastólica do ventrículo esquerdo (DDFVE), o mecanismo que liga a interrupção da produção de hormônios ovarianos, especialmente o estrogênio, ao desenvolvimento da obesidade, DDFVE, e ICFEP em mulheres em processo de envelhecimento não é claro. Estudos clínicos e epidemiológicos demonstram que mulheres na pós-menopausa com obesidade abdominal (definida pela circunferência de cintura) têm risco maior de desenvolver a ICFEP do que homens ou mulheres sem obesidade abdominal. Este estudo analisa dados clínicos que corroboram a existência de uma ligação de mecanismo entre a perda de estrogênio mais obesidade e o remodelamento ventricular esquerdo com ICFEP. Ele também discute os possíveis mecanismos celulares e moleculares para a proteção mediada por estrogênio contra tipos de células, depósitos de tecidos, função e metabolismo de adipócitos negativos que podem contribuir para a DDFVE e a ICFEP.

Estrogênio; Obesidade; Insuficiência Cardíaca; Volume Sistólico, Menopausa; Adiposidade; Sobrepeso; Ecocardiografia/métodos; Índice de Massa Corporal

Introduction

The prevalence of obesity is steadily increasing worldwide.11. Collaborators GBDO, Afshin A, Forouzanfar MH, Reitsma MB, Sur P, Estep K, et al. Health Effects of Overweight and Obesity in 195 Countries over 25 Years. N Engl J Med. 2017;377(1):13-27. Because obesity is associated with high mortality and the development of comorbid conditions, including diabetes mellitus and cardiovascular disease (CVD), it is one of the most difficult public health issues facing our society. This cluster of obesity-related comorbidities, whether directly or indirectly (e.g., side effect from anthracycline-formulated chemotherapy)22. Guenancia C, Lefebvre A, Cardinale D, Yu AF, Ladoire S, Ghiringhelli F, et al. Obesity As a Risk Factor for Anthracyclines and Trastuzumab Cardiotoxicity in Breast Cancer: A Systematic Review and Meta-Analysis. J Clin Oncol. 2016;34(26):3157-65. often culminates in heart failure (HF).33. Alpert MA, Lavie CJ, Agrawal H, Aggarwal KB, Kumar SA. Obesity and heart failure: epidemiology, pathophysiology, clinical manifestations, and management. Translat Res. 2014;164(4):345-56.

4. Lee HJ, Kim HL, Lim WH, Seo JB, Kim SH, Zo JH, et al. Subclinical alterations in left ventricular structure and function according to obesity and metabolic health status. PloS one. 2019;14(9):e0222118.

5. Russo C, Sera F, Jin Z, Palmieri V, Homma S, Rundek T, et al. Abdominal adiposity, general obesity, and subclinical systolic dysfunction in the elderly: A population-based cohort study. Eur Heart J Fail.2016;18(5):537-44.
- 66. Yang H, Huynh QL, Venn AJ, Dwyer T, Marwick TH. Associations of childhood and adult obesity with left ventricular structure and function. Int J Obes. 2017;41(4):560-8.

While obesity, defined as having a body mass index (BMI) > 30 kg/m22. Guenancia C, Lefebvre A, Cardinale D, Yu AF, Ladoire S, Ghiringhelli F, et al. Obesity As a Risk Factor for Anthracyclines and Trastuzumab Cardiotoxicity in Breast Cancer: A Systematic Review and Meta-Analysis. J Clin Oncol. 2016;34(26):3157-65. , is an independent predictor of incident HF in the general population, evidence shows that even being overweight (BMI 25-29 kg/m22. Guenancia C, Lefebvre A, Cardinale D, Yu AF, Ladoire S, Ghiringhelli F, et al. Obesity As a Risk Factor for Anthracyclines and Trastuzumab Cardiotoxicity in Breast Cancer: A Systematic Review and Meta-Analysis. J Clin Oncol. 2016;34(26):3157-65. ) carries an increased risk of HF.77. Russo C, Jin Z, Homma S, Rundek T, Elkind MS, Sacco RL, et al. Effect of obesity and overweight on left ventricular diastolic function: a community-based study in an elderly cohort. J Am Coll Cardiol. 2011;57(12):1368-74.

8. Rayner JJ, Banerjee R, Holloway CJ, Lewis AJM, Peterzan MA, Francis JM, et al. The relative contribution of metabolic and structural abnormalities to diastolic dysfunction in obesity. Int J Obes (Lond). 2018;42(3):441-7.

9. Lee SL, Daimon M, Di Tullio MR, Homma S, Nakao T, Kawata T, et al. Relationship of Left Ventricular Diastolic Function to Obesity and Overweight in a Japanese Population With Preserved Left Ventricular Ejection Fraction. Circ J. 2016;80(9):1951-6.
- 1010. Pandey A, Patel KV, Vaduganathan M, Sarma S, Haykowsky MJ, Berry JD, et al. Physical Activity, Fitness, and Obesity in Heart Failure With Preserved Ejection Fraction. JACC Heart Fail. 2018;6(12):975-82.

Several studies show that measures of central adiposity, such as waist circumference (WC), are superior to measures of global adiposity, such as weight and BMI, in estimating the risk of CVD.1111. Bigaard J, Frederiksen K, Tjonneland A, Thomsen BL, Overvad K, Heitmann BL, et al. Waist circumference and body composition in relation to all-cause mortality in middle-aged men and women. Int J Obes. 2005;29(7):778-84.

12. Ashwell M, Hsieh SD. Six reasons why the waist-to-height ratio is a rapid and effective global indicator for health risks of obesity and how its use could simplify the international public health message on obesity. Int Nat Food Sci Nutr. 2005;56(5):303-7.

13. Ho SY, Lam TH, Janus ED, Hong Kong Cardiovascular Risk Factor Prevalence Study Steering C. Waist to stature ratio is more strongly associated with cardiovascular risk factors than other simple anthropometric indices. Ann Epidemiol. 2003;13(10):683-91.

14. Janssen I, Katzmarzyk PT, Ross R. Waist circumference and not body mass index explains obesity-related health risk. Am J Clin Nutr. 2004;79(3):379-84.

15. Wei M, Gaskill SP, Haffner SM, Stern MP. Waist circumference as the best predictor of noninsulin dependent diabetes mellitus (NIDDM) compared to body mass index, waist/hip ratio and other anthropometric measurements in Mexican Americans--a 7-year prospective study. Obes Res. 1997;5(1):16-23.
- 1616. Welborn TA, Dhaliwal SS. Preferred clinical measures of central obesity for predicting mortality. Eur J Clin Nutr. 2007;61(12):1373-9. WC is independently associated with left ventricular diastolic dysfunction (LVDD), defined by echocardiographic parameters.1717. Canepa M, Strait JB, Abramov D, Milaneschi Y, AlGhatrif M, Moni M, et al. Contribution of central adiposity to left ventricular diastolic function (from the Baltimore Longitudinal Study of Aging). Am J Cardiol. 2012;109(8):1171-8. Both LVDD and obesity are common factors that contribute to a heart failure with preserved ejection fraction (HFpEF) phenotype, and appear to be causally linked.1717. Canepa M, Strait JB, Abramov D, Milaneschi Y, AlGhatrif M, Moni M, et al. Contribution of central adiposity to left ventricular diastolic function (from the Baltimore Longitudinal Study of Aging). Am J Cardiol. 2012;109(8):1171-8.

18. Haass M, Kitzman DW, Anand IS, Miller A, Zile MR, Massie BM, et al. Body mass index and adverse cardiovascular outcomes in heart failure patients with preserved ejection fraction: results from the Irbesartan in Heart Failure with Preserved Ejection Fraction (I-PRESERVE) trial. Circ Heart Fail. 2011;4(3):324-31.
- 1919. Ho JE, Lyass A, Lee DS, Vasan RS, Kannel WB, Larson MG, et al. Predictors of new-onset heart failure: differences in preserved versus reduced ejection fraction. Circ Heart Fail. 2013;6(2):279-86. Indeed, HF patients can have different phenotypes according to the morpho-functional characteristics of the disease.2020. Silverman DN, Shah SJ. Treatment of Heart Failure With Preserved Ejection Fraction (HFpEF): the Phenotype-Guided Approach. Curr Treat Options Cardiovasc Med. 2019;21(4):20. Briefly, HF patients are classified according to LV function; those with LV ejection fractions less than or equal to 40% fall into the category of heart failure with reduced ejection fraction (HFrEF), and patients with LV ejection fractions equal to or greater than 50% are deemed to have HFpEF. According to the American College of Cardiology and American Heart Association guidelines,2121. Lam CSP, Chandramouli C. Fat, Female, Fatigued: Features of the Obese HFpEF Phenotype. JACC Heart Fail. 2018;6(8):710-3. there is also an intermediate or borderline group of patients who have ejection fractions between 41% and 49%, sometimes referred to as HFmEF. Moreover, a subset of patients with ejection fractions greater than 40% with HFpEF who previously had HFrEF is considered to be clinically distinct from those with persistently preserved or reduced ejection fractions. For purposes of this review, we have focused only on HFpEF, and, specifically, features of the “fat, female, and fatigued” obese HFpEF phenotype.2121. Lam CSP, Chandramouli C. Fat, Female, Fatigued: Features of the Obese HFpEF Phenotype. JACC Heart Fail. 2018;6(8):710-3.

For a narrative of the literature of all the clinical phenotypes of HFpEF, we refer the reader to the review by Silverman.2020. Silverman DN, Shah SJ. Treatment of Heart Failure With Preserved Ejection Fraction (HFpEF): the Phenotype-Guided Approach. Curr Treat Options Cardiovasc Med. 2019;21(4):20. Regardless of the biological phenotype, HFpEF is a heterogeneous clinical syndrome, including cardiomyocyte, extracellular matrix, vascular, and comorbidity-related pathophysiological mechanisms.2222. Lewis GA, Schelbert EB, Williams SG, Cunnington C, Ahmed F, McDonagh TA, et al. Biological Phenotypes of Heart Failure With Preserved Ejection Fraction. J Am Coll Cardiol. 2017;70(17):2186-200. It is characterized by reduced end-diastolic volume, left ventricular hypertrophy, and increased left atrial volume and left ventricular filling pressure. These pathophysiological abnormalities are associated with increased left ventricular stiffness, decreased left ventricular relaxation, cardiomyocyte hypertrophy, myocardial interstitial fibrosis, and reduced intramyocardial capillaries.2323. Mohammed SF, Borlaug BA, Roger VL, Mirzoyev SA, Rodeheffer RJ, Chirinos JA, et al. Comorbidity and ventricular and vascular structure and function in heart failure with preserved ejection fraction: a community-based study. Circ Heart Fail. 2012;5(6):710-9.

24. Lam CS, Roger VL, Rodeheffer RJ, Bursi F, Borlaug BA, Ommen SR, et al. Cardiac structure and ventricular-vascular function in persons with heart failure and preserved ejection fraction from Olmsted County, Minnesota. Circulation. 2007;115(15):1982-90.

25. Zile MR, Gottdiener JS, Hetzel SJ, McMurray JJ, Komajda M, McKelvie R, et al. Prevalence and significance of alterations in cardiac structure and function in patients with heart failure and a preserved ejection fraction. Circulation. 2011;124(23):2491-501.
- 2626. Zile MR, Baicu CF, Gaasch WH. Diastolic heart failure--abnormalities in active relaxation and passive stiffness of the left ventricle. N Engl J Med. 2004;350(19):1953-9.

Another important factor involved in the HFpEF phenotype is sex. HFpEF disproportionally affects more women (sex ratio of about 2:1) then men.2727. Maslov PZ, Kim JK, Argulian E, Ahmadi A, Narula N, Singh M, et al. Is Cardiac Diastolic Dysfunction a Part of Post-Menopausal Syndrome? JACC Heart Fail. 2019;7(3):192-203. , 2828. Pandey A, Omar W, Ayers C, LaMonte M, Klein L, Allen NB, et al. Sex and Race Differences in Lifetime Risk of Heart Failure With Preserved Ejection Fraction and Heart Failure With Reduced Ejection Fraction. Circulation. 2018;137(17):1814-23. The higher prevalence of HFpEF in elderly women2929. Upadhya B, Kitzman DW. Heart Failure with Preserved Ejection Fraction in Older Adults. Heart Fail Clin. 2017;13(3):485-502. appears related to the loss of ovarian hormones, primarily estrogens, that occur after the menopause.

Accordingly, in this review, we will explore pre-clinical and clinical data about the relationships between sex, “fatness”, including mechanisms of fat-induced cardiac impairment, specifically LVDD and HFpEF, and the cardioprotective effects of estrogen on fat metabolism in women.

Link between “fatness”, sex, and HFpEF: clinical evidence

HF is a major problem that is increasing in scope. Despite recent therapeutic advances, morbidity and mortality after the onset of HF still remain substantial.3030. Benjamin EJ, Blaha MJ, Chiuve SE, Cushman M, Das SR, Deo R, et al. Heart Disease and Stroke Statistics-2017 Update: A Report From the American Heart Association. Circulation. 2017;135(10):e146-e603. Consequently, prevention of HF through the identification of the preclinical phases of the disease and the management of risk factors is a priority. Considering that 50 percent of all HF patients have HFpEF,3131. Fonarow GC, Stough WG, Abraham WT, Albert NM, Gheorghiade M, Greenberg BH, et al. Characteristics, treatments, and outcomes of patients with preserved systolic function hospitalized for heart failure: a report from the OPTIMIZE-HF Registry. J Am Coll Cardiol. 2007;50(8):768-77. the complex pathophysiology of this disease is still not fully understood, with no specific therapy available to improve patient outcomes. In this context, several studies have evaluated obesity as a risk factor for LV remodeling and subsequent HFpEF.3232. Alpert MA, Karthikeyan K, Abdullah O, Ghadban R. Obesity and Cardiac Remodeling in Adults: Mechanisms and Clinical Implications. Prog Cardiovasc Dis. 2018;61(2):114-23.

33. Ferron AJT, Francisqueti FV, Minatel IO, Silva C, Bazan SGZ, Kitawara KAH, et al. Association between Cardiac Remodeling and Metabolic Alteration in an Experimental Model of Obesity Induced by Western Diet. Nutrients. 2018;10(11).
- 3434. Antonini-Canterin F, Di Nora C, Poli S, Sparacino L, Cosei I, Ravasel A, et al. Obesity, Cardiac Remodeling, and Metabolic Profile: Validation of a New Simple Index beyond Body Mass Index. J Cardiovasc Echogr. 2018;28(1):18-25. In these studies, obesity has been consistently associated with LV stiffness and diastolic dysfunction, particularly in women.1717. Canepa M, Strait JB, Abramov D, Milaneschi Y, AlGhatrif M, Moni M, et al. Contribution of central adiposity to left ventricular diastolic function (from the Baltimore Longitudinal Study of Aging). Am J Cardiol. 2012;109(8):1171-8. , 3535. Wohlfahrt P, Redfield MM, Lopez-Jimenez F, Melenovsky V, Kane GC, Rodeheffer RJ, et al. Impact of general and central adiposity on ventricular-arterial aging in women and men. JACC Heart failure. 2014;2(5):489-99.

A community-based clinical study of 377 randomly selected participants older than 16 years of age assessed the independent contribution of indices of adiposity to variations in early-to-late (atrial) transmitral velocity (E/A), as determined by echocardiography. The main goal was to clarify why some previous studies failed to establish a contribution of obesity to LV diastolic function, while others demonstrated a relatively minor contribution. For each study participant, the independent relationship between adiposity and diastolic (E/A) or systolic (LV ejection fraction; LVEF) chamber functions were determined using multivariate linear regression analysis with adjustments for age, gender, conventional systolic or diastolic blood pressure measurements, and either LV mass index or relative wall thickness (calculated from echocardiography). Excessive central adiposity (WC), but not elevated BMI, was independently and inversely correlated with E/A; the investigators emphasized that WC might represent a progressive preclinical condition that contributes to obesity-induced diastolic HF.3636. Libhaber CD, Norton GR, Majane OH, Libhaber E, Essop MR, Brooksbank R, et al. Contribution of central and general adiposity to abnormal left ventricular diastolic function in a community sample with a high prevalence of obesity. Am J Cardiol. 2009;104(11):1527-33. WC was second only to age and equivalent to blood pressure in the magnitude of effect on E/A. Findings from this study also suggested that, at a population level, LV mass and geometry play little or no role in the pathogenesis of obesity-induced LV diastolic abnormalities.3636. Libhaber CD, Norton GR, Majane OH, Libhaber E, Essop MR, Brooksbank R, et al. Contribution of central and general adiposity to abnormal left ventricular diastolic function in a community sample with a high prevalence of obesity. Am J Cardiol. 2009;104(11):1527-33. Interestingly, there was no relationship between WC and LVEF (systolic dysfunction), confirming the findings of other researchers that identified WC as a risk factor for HFpEF.77. Russo C, Jin Z, Homma S, Rundek T, Elkind MS, Sacco RL, et al. Effect of obesity and overweight on left ventricular diastolic function: a community-based study in an elderly cohort. J Am Coll Cardiol. 2011;57(12):1368-74. , 99. Lee SL, Daimon M, Di Tullio MR, Homma S, Nakao T, Kawata T, et al. Relationship of Left Ventricular Diastolic Function to Obesity and Overweight in a Japanese Population With Preserved Left Ventricular Ejection Fraction. Circ J. 2016;80(9):1951-6. , 1717. Canepa M, Strait JB, Abramov D, Milaneschi Y, AlGhatrif M, Moni M, et al. Contribution of central adiposity to left ventricular diastolic function (from the Baltimore Longitudinal Study of Aging). Am J Cardiol. 2012;109(8):1171-8. , 3737. Fuster JJ, Ouchi N, Gokce N, Walsh K. Obesity-Induced Changes in Adipose Tissue Microenvironment and Their Impact on Cardiovascular Disease. Circ Res. 2016;118(11):1786-807.

38. Schiattarella GG, Altamirano F, Tong D, French KM, Villalobos E, Kim SY, et al. Nitrosative stress drives heart failure with preserved ejection fraction. Nature. 2019;568(7752):351-6.

39. Chirinos JA, Zamani P. The Nitrate-Nitrite-NO Pathway and Its Implications for Heart Failure and Preserved Ejection Fraction. Curr Heart Fail Rep. 2016;13(1):47-59.

40. Alvarez P, Briasoulis A. Immune Modulation in Heart Failure: the Promise of Novel Biologics. Current treatment. Options Cardiovasc Med. 2018 2018;20(3):26.

41. Hulsmans M, Sager HB, Roh JD, Valero-Munoz M, Houstis NE, Iwamoto Y, et al. Cardiac macrophages promote diastolic dysfunction. J Exper Med.. 2018;215(2):423-40.

42. Van Tassell BW, Trankle CR, Canada JM, Carbone S, Buckley L, Kadariya D, et al. IL-1 Blockade in Patients With Heart Failure With Preserved Ejection Fraction. Circ Heart Fail. 2018;11(8):e005036.
- 4343. Van Tassell BW, Buckley LF, Carbone S, Trankle CR, Canada JM, Dixon DL, et al. Interleukin-1 blockade in heart failure with preserved ejection fraction: rationale and design of the Diastolic Heart Failure Anakinra Response Trial 2 (D-HART2). Clin Cardiol. 2017;40(9):626-32. Finally, it is relevant to comment that the data reported by these authors were restricted to women as they have recruited a limited proportion of male participants.3636. Libhaber CD, Norton GR, Majane OH, Libhaber E, Essop MR, Brooksbank R, et al. Contribution of central and general adiposity to abnormal left ventricular diastolic function in a community sample with a high prevalence of obesity. Am J Cardiol. 2009;104(11):1527-33. Additional data from the clinical study performed by Canepa et al.,1717. Canepa M, Strait JB, Abramov D, Milaneschi Y, AlGhatrif M, Moni M, et al. Contribution of central adiposity to left ventricular diastolic function (from the Baltimore Longitudinal Study of Aging). Am J Cardiol. 2012;109(8):1171-8. in which the sample, from both genders, is part of the Baltimore Longitudinal Study of Aging (BLSA) propose that a possible pathophysiologic explanation for the association of central adiposity with worse cardiovascular outcomes is its relationship with LVDD. They found that central adiposity was strongly associated with LV dysfunction, particularly with impaired LV relaxation. The researchers also found that the effect of central adiposity on LVDD was independent of general adiposity and, surprisingly, this was more pronounced in women than in men. The gender-specific effect of central fat accumulation on LVDD was then graded using various echocardiographic parameters. The study confirmed their previous reports showing a negative correlation between the indices of adiposity and the E/A ratio; however, the authors also found that E/A alone was not enough to discriminate between subjects with normal or abnormal diastolic function and that mitral valve inflow velocities were also significantly influenced by the increase in preload, a condition frequently encountered in obese subjects. When these issues were normalized by combining tissue Doppler measures of mitral annular velocity, or e’ and dynamic (E/A) parameters, it was found that the E/e’ ratio was positively correlated with WC. Thus, this epidemiological study provided further evidence of the link between central obesity (WC) and the prevalence and development of LVDD, and that this link is influenced by sex.1717. Canepa M, Strait JB, Abramov D, Milaneschi Y, AlGhatrif M, Moni M, et al. Contribution of central adiposity to left ventricular diastolic function (from the Baltimore Longitudinal Study of Aging). Am J Cardiol. 2012;109(8):1171-8.

One limitation of cross-sectional studies is that they offer a snapshot of a single moment in time, and thus fail to capture changes that occur over time. The complex relationships between aging, sex, adiposity, and ventricular mechanisms were evaluated in a large longitudinal study conducted over a period of 4 years in 1,402 subjects, 45 years of age and older, who were randomly selected from a community-based population.3535. Wohlfahrt P, Redfield MM, Lopez-Jimenez F, Melenovsky V, Kane GC, Rodeheffer RJ, et al. Impact of general and central adiposity on ventricular-arterial aging in women and men. JACC Heart failure. 2014;2(5):489-99. It was found that weight gain during a 4-year period was associated with significant increases in LV diastolic stiffness in both men and women, but that it was more pronounced in women, indicating a sex difference in the biology of age-related ventricular stiffening. Furthermore, evaluating central obesity in women may help to identify a group at higher risk for incident HFpEF who might benefit from preventive treatment.3535. Wohlfahrt P, Redfield MM, Lopez-Jimenez F, Melenovsky V, Kane GC, Rodeheffer RJ, et al. Impact of general and central adiposity on ventricular-arterial aging in women and men. JACC Heart failure. 2014;2(5):489-99. Finally, the results of this longitudinal study confirmed the findings of cross-sectional investigations regarding the positive relationship between WC and echocardiographic measures of diastolic dysfunction (e.g., E/e’ ratio).

How changes in fat tissue depot might influence the link between obesity and impairments in cardiac function and remodeling, particularly among women, are important to consider. While female sex hormones are believed to cause fat to be stored in the buttocks, thighs, and hips of women, which may be essential for normal reproduction purposes, menopause-related changes in body fat distribution may partially explain the increased risk of cardiovascular and metabolic disease during the postmenopausal years.4444. Garaulet M, Perez-Llamas F, Baraza JC, Garcia-Prieto MD, Fardy PS, Tebar FJ, et al. Body fat distribution in pre-and post-menopausal women: metabolic and anthropometric variables. J Nutr Health Aging. 2002;6(2):123-6.

45. Zamboni M, Armellini F, Milani MP, De Marchi M, Todesco T, Robbi R, et al. Body fat distribution in pre- and post-menopausal women: metabolic and anthropometric variables and their inter-relationships. Int In and related metabolic disorders : journal of the International Association for the Study of Obesity. Int J Obes.1992;16(7):495-504.
- 4646. Toth MJ, Tchernof A, Sites CK, Poehlman ET. Menopause-related changes in body fat distribution. Ann NY Acad Sci. 2000;904:502-6. In 2011, Wehr et al.,4747. Wehr E, Pilz S, Boehm BO, Marz W, Obermayer-Pietsch B. The lipid accumulation product is associated with increased mortality in normal weight postmenopausal women. Obesity. 2011;19(9):1873-80. published results from a longitudinal study of gender-specific differences in the relationship between the lipid accumulation product, which is calculated from WC, and cardiovascular mortality, as well as the presence of type 2 diabetes. The study included 2,279 men and 875 postmenopausal women, with a median follow-up of 77 years. Lipid accumulation product levels were independently associated with congestive HF mortality in all postmenopausal women and with all-cause mortality in diabetic postmenopausal women, but not in men. These data not only support the concept that fat redistribution after estrogen loss may contribute to cardiovascular disease progression, but also endorse an inexpensive and simple risk biomarker, namely, lipid accumulation product, which could identify postmenopausal women at higher cardiovascular risk.4747. Wehr E, Pilz S, Boehm BO, Marz W, Obermayer-Pietsch B. The lipid accumulation product is associated with increased mortality in normal weight postmenopausal women. Obesity. 2011;19(9):1873-80.

Treatment modalities for weight loss: an evidence-based approach to measure outcomes in patients with HFpEF

Given clinical evidence for crosstalk between the heart and “fatness”, with respect to female sex-specific HFpEF, weight reduction or maintenance of ideal body weight is one preventive approach to mitigate age- and estrogen-loss related changes in ventricular structure and function. Essential treatments for weight reduction include changes in eating habits to reduced-calorie and low-fat diets, increased physical activity or exercise, and other behavioral modification strategies, such as self-monitoring (e.g., daily record keeping of food intake and exercise), stimulus control (e.g., avoiding triggers that prompt eating), and problem solving (e.g., identifying barriers and ways to overcome them). Additionally, bariatric surgery is another effective strategy to treat severely obese patients. Thus, it is important to review the literature addressing the effects of different weight loss strategies on the cardiovascular outcomes in patients with LVDD and HFpEF.

Observational studies suggest that overweight or mildly-to-moderately obese patients with HFpEF survive longer than those who are normal-weight.1818. Haass M, Kitzman DW, Anand IS, Miller A, Zile MR, Massie BM, et al. Body mass index and adverse cardiovascular outcomes in heart failure patients with preserved ejection fraction: results from the Irbesartan in Heart Failure with Preserved Ejection Fraction (I-PRESERVE) trial. Circ Heart Fail. 2011;4(3):324-31. , 4848. Joyce E, Lala A, Stevens SR, Cooper LB, AbouEzzeddine OF, Groarke JD, et al. Prevalence, Profile, and Prognosis of Severe Obesity in Contemporary Hospitalized Heart Failure Trial Populations. JACC Heart failure. 2016;4(12):923-31. However, Kitzman et al.4949. Kitzman DW, Brubaker P, Morgan T, Haykowsky M, Hundley G, Kraus WE, et al. Effect of Caloric Restriction or Aerobic Exercise Training on Peak Oxygen Consumption and Quality of Life in Obese Older Patients With Heart Failure With Preserved Ejection Fraction: A Randomized Clinical Trial. JAMA. 2016;315(1):36-46. recently reported that 20 weeks of caloric restriction combined with aerobic exercise training among obese older patients with HFpEF reduced their body weight with additive improvements in exercise capacity, defined by peak V02. Even so, only caloric restriction resulted in decreases in LV mass and relative wall thickness, along with an inkling for improvements in diastolic function, as per the increase in E/A observed in this treatment arm, without effecting resting cardiac function, depicted by ejection fraction or Doppler-derived cardiac output.4949. Kitzman DW, Brubaker P, Morgan T, Haykowsky M, Hundley G, Kraus WE, et al. Effect of Caloric Restriction or Aerobic Exercise Training on Peak Oxygen Consumption and Quality of Life in Obese Older Patients With Heart Failure With Preserved Ejection Fraction: A Randomized Clinical Trial. JAMA. 2016;315(1):36-46. It was further reported that no changes in magnetic resonance imaging (MRI) measures of epicardial or pericardial fat were observed across treatments, but that there were significant reductions in thigh and abdominal subcutaneous and visceral fat depots in the diet only group.4949. Kitzman DW, Brubaker P, Morgan T, Haykowsky M, Hundley G, Kraus WE, et al. Effect of Caloric Restriction or Aerobic Exercise Training on Peak Oxygen Consumption and Quality of Life in Obese Older Patients With Heart Failure With Preserved Ejection Fraction: A Randomized Clinical Trial. JAMA. 2016;315(1):36-46. While these findings favor a role for weight reduction strategies, along with exercise, to improve the detriments in exercise capacity and maximal oxygen consumption associated with HFpEF among obese patients, they also support the concept that extra-cardiac mechanisms are uniquely involved in the pathogenesis of HFpEF.5050. Kitzman DW, Brubaker PH, Herrington DM, Morgan TM, Stewart KP, Hundley WG, et al. Effect of endurance exercise training on endothelial function and arterial stiffness in older patients with heart failure and preserved ejection fraction: a randomized, controlled, single-blind trial. J Am Coll Cardiol. 2013;62(7):584-92. , 5151. Haykowsky MJ, Brubaker PH, Stewart KP, Morgan TM, Eggebeen J, Kitzman DW. Effect of endurance training on the determinants of peak exercise oxygen consumption in elderly patients with stable compensated heart failure and preserved ejection fraction. J Am Coll Cardiol. 2012;60(2):120-8.

Another weight reduction strategy that is conducive to evaluating the link between “fatness”, LVDD, and HFpEF is bariatric surgery. Indeed, morbidly obese patients who commonly present hemodynamic and cardiac morphometric characteristics, such as elevations in cardiac preload and afterload and increases in LV chamber and wall dimensions, which contribute to myocardial stiffness and impairments in myocardial relaxation.5252. Shin SH, Lee YJ, Heo YS, Park SD, Kwon SW, Woo SI, et al. Beneficial Effects of Bariatric Surgery on Cardiac Structure and Function in Obesity. Obes Surg. 2017;27(3):620-5. Multiple clinical studies5252. Shin SH, Lee YJ, Heo YS, Park SD, Kwon SW, Woo SI, et al. Beneficial Effects of Bariatric Surgery on Cardiac Structure and Function in Obesity. Obes Surg. 2017;27(3):620-5.

53. Aggarwal R, Harling L, Efthimiou E, Darzi A, Athanasiou T, Ashrafian H. The Effects of Bariatric Surgery on Cardiac Structure and Function: a Systematic Review of Cardiac Imaging Outcomes. Obes Surg. 2016;26(5):1030-40.

54. Karason K, Wallentin I, Larsson B, Sjostrom L. Effects of obesity and weight loss on left ventricular mass and relative wall thickness: survey and intervention study. BMJ. 1997;315(7113):912-6.
- 5555. Jhaveri RR, Pond KK, Hauser TH, Kissinger KV, Goepfert L, Schneider B, et al. Cardiac remodeling after substantial weight loss: a prospective cardiac magnetic resonance study after bariatric surgery. Surg Obes Relat Dis. 2009;5(6):648-52. and one systemic review with a meta-analysis5656. Cuspidi C, Rescaldani M, Tadic M, Sala C, Grassi G. Effects of bariatric surgery on cardiac structure and function: a systematic review and meta-analysis. Am J Hypertens. 2014;27(2):146-56. reported the benefits of bariatric surgery with subsequent weight loss on echocardiographic and MRI measures of LV structure, including substantial reductions in LV wall thickness and mass and diastolic function. Along with upgrades in diastolic function, others found that extensive weight loss after bariatric surgery also led to favorable changes in muscle metabolism5757. Leichman JG, Wilson EB, Scarborough T, Aguilar D, Miller CC, 3rd, Yu S, et al. Dramatic reversal of derangements in muscle metabolism and left ventricular function after bariatric surgery. Am J Med. 2008;121(11):966-73. and characteristics of arterial elasticity.5858. Ikonomidis I, Mazarakis A, Papadopoulos C, Patsouras N, Kalfarentzos F, Lekakis J, et al. Weight loss after bariatric surgery improves aortic elastic properties and left ventricular function in individuals with morbid obesity: a 3-year follow-up study. J Hypert.2007;25(2):439-47. , 5959. Iancu ME, Copaescu C, Serban M, Ginghina C. Favorable changes in arterial elasticity, left ventricular mass, and diastolic function after significant weight loss following laparoscopic sleeve gastrectomy in obese individuals. Obes Surg. 2014;24(3):364-70.

Link between cardiac-specific regional adiposity, LVDD, and HFpEF

In addition to peripheral and total body fat linked to LVDD, the potential role of cardiac-specific regional adiposity (e.g. pericardial and epicardial fat depots) in the disease process should not be ignored.6060. Nyman K, Graner M, Pentikainen MO, Lundbom J, Hakkarainen A, Siren R, et al. Cardiac steatosis and left ventricular function in men with metabolic syndrome. J Cardiovasc Magn Reson. 2013;15:103. Pericardial and epicardial fat, commonly found in obese and overweight patients, are considered ectopic adipose depots that can induce a lipotoxic state in close proximity to cardiac muscle and coronary arteries.6161. Konishi M, Sugiyama S, Sugamura K, Nozaki T, Matsubara J, Akiyama E, et al. Accumulation of pericardial fat correlates with left ventricular diastolic dysfunction in patients with normal ejection fraction. J Cardiol. 2012;59(3):344-51. , 6262. Iacobellis G, Ribaudo MC, Assael F, Vecci E, Tiberti C, Zappaterreno A, et al. Echocardiographic epicardial adipose tissue is related to anthropometric and clinical parameters of metabolic syndrome: a new indicator of cardiovascular risk. J Clin Endocrinol Metab. 2003;88(11):5163-8. Moreover, we know that metabolic syndrome, a common condition among obese and overweight patient,6363. Morelli NR, Scavuzzi BM, Miglioranza L, Lozovoy MAB, Simao ANC, Dichi I. Metabolic syndrome components are associated with oxidative stress in overweight and obese patients. Arch Endocrinol Metab. 2018;62(3):309-18. is associated with increased adipose tissue volume around the heart,6060. Nyman K, Graner M, Pentikainen MO, Lundbom J, Hakkarainen A, Siren R, et al. Cardiac steatosis and left ventricular function in men with metabolic syndrome. J Cardiovasc Magn Reson. 2013;15:103. particularly the accumulation of epicardial fat,6464. Iacobellis G. Epicardial and pericardial fat: close, but very different. Obesity. 2009;17(4):625; author reply 6-7. which is significantly linked to adverse cardiovascular events,6565. Ding J, Hsu FC, Harris TB, Liu Y, Kritchevsky SB, Szklo M, et al. The association of pericardial fat with incident coronary heart disease: the Multi-Ethnic Study of Atherosclerosis (MESA). Am J Clin Nutr. 2009;90(3):499-504.

66. Tamarappoo B, Dey D, Shmilovich H, Nakazato R, Gransar H, Cheng VY, et al. Increased pericardial fat volume measured from noncontrast CT predicts myocardial ischemia by SPECT. JACC Cardiovasc Imag. 2010;3(11):1104-12.

67. Aslanabadi N, Salehi R, Javadrashid A, Tarzamni M, Khodadad B, Enamzadeh E, et al. Epicardial and pericardial fat volume correlate with the severity of coronary artery stenosis. J Cardiovasc Thorac Res. 2014;6(4):235-9.

68. Cheng VY, Dey D, Tamarappoo B, Nakazato R, Gransar H, Miranda-Peats R, et al. Pericardial fat burden on ECG-gated noncontrast CT in asymptomatic patients who subsequently experience adverse cardiovascular events. JACC Cardiovasc Imag 2010;3(4):352-60.
- 6969. Mahabadi AA, Berg MH, Lehmann N, Kalsch H, Bauer M, Kara K, et al. Association of epicardial fat with cardiovascular risk factors and incident myocardial infarction in the general population: the Heinz Nixdorf Recall Study. J Am Coll Cardiol. 2013;61(13):1388-95. including HFpEF.6161. Konishi M, Sugiyama S, Sugamura K, Nozaki T, Matsubara J, Akiyama E, et al. Accumulation of pericardial fat correlates with left ventricular diastolic dysfunction in patients with normal ejection fraction. J Cardiol. 2012;59(3):344-51. , 7070. Alonso-Gomez AM, Tojal Sierra L, Fortuny Frau E, Goicolea Guemez L, Aboitiz Uribarri A, Portillo MP, et al. Diastolic dysfunction and exercise capacity in patients with metabolic syndrome and overweight/obesity. Int J Cardiol Heart Vasc. 2019;22:67-72. , 7171. Haykowsky MJ, Nicklas BJ, Brubaker PH, Hundley WG, Brinkley TE, Upadhya B, et al. Regional Adipose Distribution and its Relationship to Exercise Intolerance in Older Obese Patients Who Have Heart Failure With Preserved Ejection Fraction. JACC Heart failure. 2018;6(8):640-9. The direct correlation between these local fat depots and LVDD can be explained, in part, by paracrine processes, whereby proinflammatory cytokines and other damaging mediators (e.g. TNF-α and IL-6),7272. Mazurek T, Zhang L, Zalewski A, Mannion JD, Diehl JT, Arafat H, et al. Human epicardial adipose tissue is a source of inflammatory mediators. Circulation. 2003;108(20):2460-6. collectively called adipokines, are released from the local adipose repositories.6161. Konishi M, Sugiyama S, Sugamura K, Nozaki T, Matsubara J, Akiyama E, et al. Accumulation of pericardial fat correlates with left ventricular diastolic dysfunction in patients with normal ejection fraction. J Cardiol. 2012;59(3):344-51. , 7272. Mazurek T, Zhang L, Zalewski A, Mannion JD, Diehl JT, Arafat H, et al. Human epicardial adipose tissue is a source of inflammatory mediators. Circulation. 2003;108(20):2460-6.

73. Sacks HS, Fain JN. Human epicardial adipose tissue: a review. Am Heart J. 2007;153(6):907-17.
- 7474. Iacobellis G, Corradi D, Sharma AM. Epicardial adipose tissue: anatomic, biomolecular and clinical relationships with the heart. Nature clinical practice Cardiovasc Med. 2005;2(10):536-43.

Distinguishing between the two fat depots and their respective link to LVDD may be important both anatomically and biochemically. For instance, epicardial fat is located between the outer wall of the heart muscle and the visceral layer of pericardium,6464. Iacobellis G. Epicardial and pericardial fat: close, but very different. Obesity. 2009;17(4):625; author reply 6-7. and its proximity to the myocardium is significant in that both tissue layers share the same blood microcirculation, the coronary arteries.6464. Iacobellis G. Epicardial and pericardial fat: close, but very different. Obesity. 2009;17(4):625; author reply 6-7. Potential interactions can be elicited when dysfunctional adipocytes from cardiac fat depots release proinflammatory adipokines into the microcirculation,3737. Fuster JJ, Ouchi N, Gokce N, Walsh K. Obesity-Induced Changes in Adipose Tissue Microenvironment and Their Impact on Cardiovascular Disease. Circ Res. 2016;118(11):1786-807. which in turn can interact with cardiomyocytes and cardiac fibroblasts. These cells independently respond to adipokines, which contribute to the pathologic process of myocardial fibrosis,7575. Schram K, Sweeney G. Implications of myocardial matrix remodeling by adipokines in obesity-related heart failure. Trends Cardiovasc Med. 2008;18(6):199-205. thereby leading to myocardial remodeling, via low grade inflammation and fibrotic processes, which can intensify LV hypertrophy, wall stiffness, and LVDD progression.7676. Packer M. Epicardial Adipose Tissue May Mediate Deleterious Effects of Obesity and Inflammation on the Myocardium. JJ Am Coll Cardiol. 2018;71(20):2360-72.

77. Patel VB, Basu R, Oudit GY. ACE2/Ang 1-7 axis: A critical regulator of epicardial adipose tissue inflammation and cardiac dysfunction in obesity. Adipocyte. 2016;5(3):306-11.

78. Patel VB, Mori J, McLean BA, Basu R, Das SK, Ramprasath T, et al. ACE2 Deficiency Worsens Epicardial Adipose Tissue Inflammation and Cardiac Dysfunction in Response to Diet-Induced Obesity. Diabetes. 2016;65(1):85-95.
- 7979. Wu CK, Tsai HY, Su MM, Wu YF, Hwang JJ, Lin JL, et al. Evolutional change in epicardial fat and its correlation with myocardial diffuse fibrosis in heart failure patients. J Clin Lipidol. 2017;11(6):1421-31. Pericardial fat, which can be more specifically referred to as para -cardial fat or intrathoracic fat,8080. Lee JJ, Yin X, Hoffmann U, Fox CS, Benjamin EJ. Relation of Pericardial Fat, Intrathoracic Fat, and Abdominal Visceral Fat With Incident Atrial Fibrillation (from the Framingham Heart Study). Am J Cardiol. 2016;118(10):1486-92. is that fat which is deposited outside the parietal pericardium. This fat depot originates from primitive thoracic mesenchyme and is supplied by noncoronary sources. While increases in paracardial fat volume in HFpEF have been reported to induce a mechanical compressive-like load on the myocardium, which impairs LV filling,8181. Fox CS, Gona P, Hoffmann U, Porter SA, Salton CJ, Massaro JM, et al. Pericardial fat, intrathoracic fat, and measures of left ventricular structure and function: the Framingham Heart Study. Circulation. 2009;119(12):1586-91. paracrine processes have also been noted. Excessive pericardial adiposity contains high levels of proinflammatory mediators that, when released from adipocytes, promote a collagen turnover, thus leading to myocardial stiffness, impaired lusitropism and subsequent LVDD.8282. Mak GJ, Ledwidge MT, Watson CJ, Phelan DM, Dawkins IR, Murphy NF, et al. Natural history of markers of collagen turnover in patients with early diastolic dysfunction and impact of eplerenone. J Am Coll Cardiol. 2009;54(18):1674-82. Indeed, Konishi et al.,6161. Konishi M, Sugiyama S, Sugamura K, Nozaki T, Matsubara J, Akiyama E, et al. Accumulation of pericardial fat correlates with left ventricular diastolic dysfunction in patients with normal ejection fraction. J Cardiol. 2012;59(3):344-51. reported that a high volume of pericardial fat was significantly correlated with Doppler-derived increases in filling pressure, or E/e’, in HFpEF patients. Moreover, studies have documented a strong potential for epicardial adiposity to be associated with poor prognosis in obese or overweight patients with LVDD and HFpEF.7171. Haykowsky MJ, Nicklas BJ, Brubaker PH, Hundley WG, Brinkley TE, Upadhya B, et al. Regional Adipose Distribution and its Relationship to Exercise Intolerance in Older Obese Patients Who Have Heart Failure With Preserved Ejection Fraction. JACC Heart failure. 2018;6(8):640-9. , 8383. van Woerden G, Gorter TM, Westenbrink BD, Willems TP, van Veldhuisen DJ, Rienstra M. Epicardial fat in heart failure patients with mid-range and preserved ejection fraction. Eur J Heart Fail. 2018;20(11):1559-66.

84. Obokata M, Reddy YNV, Pislaru SV, Melenovsky V, Borlaug BA. Evidence Supporting the Existence of a Distinct Obese Phenotype of Heart Failure With Preserved Ejection Fraction. Circulation. 2017;136(1):6-19.
- 8585. Cavalcante JL, Tamarappoo BK, Hachamovitch R, Kwon DH, Alraies MC, Halliburton S, et al. Association of epicardial fat, hypertension, subclinical coronary artery disease, and metabolic syndrome with left ventricular diastolic dysfunction. Am J Cardiol. 2012;110(12):1793-8.

Given the link between local cardiac fat depots and adverse cardiovascular health, weight reduction strategies should be strongly considered among the armamentarium in the management of the obese LVDD patient. Interestingly, in obese postmenopausal women with HFpEF, Brinkley et al.,8686. Brinkley TE, Ding J, Carr JJ, Nicklas BJ. Pericardial fat loss in postmenopausal women under conditions of equal energy deficit. Med Sci Sports Exerc. 2011;43(5):808-14. showed that caloric restriction, aerobic exercise, or combination therapy significantly reduced body weight and pericardial fat, and the changes in pericardial fat were inversely correlated with cardiorespiratory fitness defined by VO2max. Certainly, future therapies targeting low-grade inflammatory processes arising from epi- and pericardial fat depots could also limit the progression of LVDD.

Link between estrogen and fat-induced cardiovascular risk

The high predilection of HFpEF among older women compared to older men with HF is well accepted.2727. Maslov PZ, Kim JK, Argulian E, Ahmadi A, Narula N, Singh M, et al. Is Cardiac Diastolic Dysfunction a Part of Post-Menopausal Syndrome? JACC Heart Fail. 2019;7(3):192-203. , 2828. Pandey A, Omar W, Ayers C, LaMonte M, Klein L, Allen NB, et al. Sex and Race Differences in Lifetime Risk of Heart Failure With Preserved Ejection Fraction and Heart Failure With Reduced Ejection Fraction. Circulation. 2018;137(17):1814-23. The role that differences in adipocyte distribution among men and woman might have with respect to this sex-specific differential in HF prevalence is new and still coming together. Indeed, women have more body fat than men, but in contrast to the adverse metabolic consequences of central obesity that is typical of men, the pear-shaped, or gluteal-femoral body fat subcutaneous distribution of many women is associated with lower cardiometabolic risk.8787. Karastergiou K, Smith SR, Greenberg AS, Fried SK. Sex differences in human adipose tissues - the biology of pear shape. Biol Sex Differ. 2012;3(1):13. , 8888. Manolopoulos KN, Karpe F, Frayn KN. Gluteofemoral body fat as a determinant of metabolic health. Int J Obes (Lond). 2010;34(6):949-59. However, with advancing age there is a general shift and expansion of fat from the subcutaneous to visceral compartment.8787. Karastergiou K, Smith SR, Greenberg AS, Fried SK. Sex differences in human adipose tissues - the biology of pear shape. Biol Sex Differ. 2012;3(1):13.

88. Manolopoulos KN, Karpe F, Frayn KN. Gluteofemoral body fat as a determinant of metabolic health. Int J Obes (Lond). 2010;34(6):949-59.
- 8989. Frank AP, de Souza Santos R, Palmer BF, Clegg DJ. Determinants of body fat distribution in humans may provide insight about obesity-related health risks. J Lipid Res. 2019;60(10):1710-9. In aging males this means expansion of abdominal visceral adiposity, while in aging females this involves a redistribution of fat from the subcutaneous gluteal-femoral compartment to the visceral-abdominal compartment.8787. Karastergiou K, Smith SR, Greenberg AS, Fried SK. Sex differences in human adipose tissues - the biology of pear shape. Biol Sex Differ. 2012;3(1):13.

88. Manolopoulos KN, Karpe F, Frayn KN. Gluteofemoral body fat as a determinant of metabolic health. Int J Obes (Lond). 2010;34(6):949-59.
- 8989. Frank AP, de Souza Santos R, Palmer BF, Clegg DJ. Determinants of body fat distribution in humans may provide insight about obesity-related health risks. J Lipid Res. 2019;60(10):1710-9. In both cases, cardiovascular disease risk increases with age-related abdominal compartment expansion of visceral fat.8989. Frank AP, de Souza Santos R, Palmer BF, Clegg DJ. Determinants of body fat distribution in humans may provide insight about obesity-related health risks. J Lipid Res. 2019;60(10):1710-9. , 9090. Mancuso P, Bouchard B. The Impact of Aging on Adipose Function and Adipokine Synthesis. Front Endocrinol (Lausanne). 2019;10:137.

As outlined in the introduction, loss of gonadal hormones in older women seems to represent a component associated with increased risk for developing HFpEF. Since women are less likely to develop CVD before menopause,9191. Lobo RA, Davis SR, De Villiers TJ, Gompel A, Henderson VW, Hodis HN, et al. Prevention of diseases after menopause. Climacteric. 2014;17(5):540-56. the production of ovarian estrogen appears to protect against HF.9292. Redfield MM, Jacobsen SJ, Borlaug BA, Rodeheffer RJ, Kass DA. Age- and gender-related ventricular-vascular stiffening: a community-based study. Circulation. 2005;112(15):2254-62. , 9393. Regitz-Zagrosek V, Oertelt-Prigione S, Seeland U, Hetzer R. Sex and gender differences in myocardial hypertrophy and heart failure. Circ J. 2010;74(7):1265-73. Consistently, there are several reports confirming the beneficial effects of estrogen in the cardiovascular system.9494. Gorodeski GI. Update on cardiovascular disease in post-menopausal women. Best Pract Res Clin Obstet Gynaecol. 2002;16(3):329-55.

95. Mendelsohn ME, Karas RH. The protective effects of estrogen on the cardiovascular system. N Engl J Med. 1999;340(23):1801-11.
- 9696. Turgeon JL, McDonnell DP, Martin KA, Wise PM. Hormone therapy: physiological complexity belies therapeutic simplicity. Science. 2004;304(5675):1269-73.

To understand the specific role of gonadal hormones in the expansion of age-related visceral fatness in women and, in turn, its potential influence on diastolic function, a brief review of gonadal hormones, particularly the estrogens and their receptors, is first warranted. The three naturally occurring estrogens in women are estrone (E1), estradiol (E2), and estriol (E3). A fourth form of estrogen, estetrol (E4), is produced only during pregnancy. All these different estrogen forms are synthesized from androgens.9797. Watson CS, Jeng YJ, Kochukov MY. Nongenomic actions of estradiol compared with estrone and estriol in pituitary tumor cell signaling and proliferation. FASEB J. 2008;22(9):3328-36. For simplicity, we will use the term estrogen, to include all forms.

Estrogen binds to multiple receptors, including classical nuclear estrogen receptors (ERs), ERα, and ERβ, and a G protein-coupled receptor, GPER.9898. Haas E, Bhattacharya I, Brailoiu E, Damjanovic M, Brailoiu GC, Gao X, et al. Regulatory role of G protein-coupled estrogen receptor for vascular function and obesity. Circ Res. 2009;104(3):288-91. The ERs signal not only through a “classical” regulation of gene transcription, but also by activating a “non-nuclear” signaling pathway.9494. Gorodeski GI. Update on cardiovascular disease in post-menopausal women. Best Pract Res Clin Obstet Gynaecol. 2002;16(3):329-55. , 9999. Ueda K, Adachi Y, Liu P, Fukuma N, Takimoto E. Regulatory Actions of Estrogen Receptor Signaling in the Cardiovascular System. Front Endocrinol (Lausanne). 2019;10:909. Accumulating findings have been well described and reviewed in the literature, concerning the roles triggered by ERs in maintaining the homeostasis of the cardiovascular system.9999. Ueda K, Adachi Y, Liu P, Fukuma N, Takimoto E. Regulatory Actions of Estrogen Receptor Signaling in the Cardiovascular System. Front Endocrinol (Lausanne). 2019;10:909.

100. Iorga A, Cunningham CM, Moazeni S, Ruffenach G, Umar S, Eghbali M. The protective role of estrogen and estrogen receptors in cardiovascular disease and the controversial use of estrogen therapy. Biol Sex Differ. 2017;8(1):33.
- 101101. Dworatzek E, Mahmoodzadeh S. Targeted basic research to highlight the role of estrogen and estrogen receptors in the cardiovascular system. Pharmacol Res. 2017;119:27-35.

Estrogen directly regulates adiposity distributions through estrogen receptors. In the premenopausal state, subcutaneous fat has relatively more estrogen and progesterone receptors than androgen receptors, whereas visceral fat has higher levels of androgen receptors.102102. Mayes JS, Watson GH. Direct effects of sex steroid hormones on adipose tissues and obesity. Obes Rev. 2004;5(4):197-216. With menopause, the fall of estrogen leads to estrogen receptors on subcutaneous fat to be inactivated, while the androgen receptors on visceral fat become relatively activated, thereby contributing to the inverse relationship between estrogen levels and visceral fat.103103. Bouchard C, Despres JP, Mauriege P. Genetic and nongenetic determinants of regional fat distribution. Endocr Rev. 1993;14(1):72-93. , 104104. Brown LM, Clegg DJ. Central effects of estradiol in the regulation of food intake, body weight, and adiposity. J Steroid Biochem Mol Biol. 2010;122(1-3):65-73. Likewise, in estrogen deficient rodent models induced by ovariectomy, the observed increase in body weight is mainly due to gains in visceral fat.105105. Clegg DJ, Brown LM, Woods SC, Benoit SC. Gonadal hormones determine sensitivity to central leptin and insulin. Diabetes. 2006;55(4):978-87. Estrogenic protection can further be seen upon the systemic administration of estrogen in OVX models whereby the body fat distribution mirrors that of gonad-intact counterparts.106106. Stubbins RE, Holcomb VB, Hong J, Nunez NP. Estrogen modulates abdominal adiposity and protects female mice from obesity and impaired glucose tolerance. Eur J Nutr. 2012;51(7):861-70.

The specific roles may offset the classical steroid receptors ERα and ERβ in the context of fat one to another. In a recent study by Zidon et al.,107107. Zidon TM, Padilla J, Fritsche KL, Welly RJ, McCabe LT, Stricklin OE, et al. Effects of ERbeta and ERalpha on OVX-induced changes in adiposity and insulin resistance. J Endocrinol. 2020;245(1):165-78. gonad intact ERα KO mice were found to be 25% heavier with reduced energy expenditure compared to age-matched gonad intact wild-type and ERβ KO mice.107107. Zidon TM, Padilla J, Fritsche KL, Welly RJ, McCabe LT, Stricklin OE, et al. Effects of ERbeta and ERalpha on OVX-induced changes in adiposity and insulin resistance. J Endocrinol. 2020;245(1):165-78. Furthermore, following OVX, αKO mice did not increase body weight or exhibit more pronounced insulin resistance, whereas WT and βKO mice did, suggesting that the loss of signaling through ERα facilitates OVX-induced metabolic dysfunction. These new data further suggest that following estrogen deficiency, ERβ may mediate protective metabolic benefits.107107. Zidon TM, Padilla J, Fritsche KL, Welly RJ, McCabe LT, Stricklin OE, et al. Effects of ERbeta and ERalpha on OVX-induced changes in adiposity and insulin resistance. J Endocrinol. 2020;245(1):165-78. This contradicts previous preclinical reports showing that the two classical ERs on adipose tissue regulate fat reciprocally.108108. Crandall DL, Busler DE, Novak TJ, Weber RV, Kral JG. Identification of estrogen receptor beta RNA in human breast and abdominal subcutaneous adipose tissue. Biochem Biophys Res Comm. 1998;248(3):523-6.

109. Matelski H, Greene R, Huberman M, Lokich J, Zipoli T. Randomized trial of estrogen vs. tamoxifen therapy for advanced breast cancer. Am J Clin Oncol. 1985;8(2):128-33.
- 110110. Kuiper GG, Enmark E, Pelto-Huikko M, Nilsson S, Gustafsson JA. Cloning of a novel receptor expressed in rat prostate and ovary. Proc Nat Acad Sci USA. 1996;93(12):5925-30. Despite this discrepancy, the linkage between polymorphisms in the structure of both classical ERs and increased CV risk among postmenopausal women corroborate the important role of ER isoforms in the regulation of adiposity, metabolic derangement, and cardiovascular risk.111111. Rexrode KM, Ridker PM, Hegener HH, Buring JE, Manson JE, Zee RY. Polymorphisms and haplotypes of the estrogen receptor-beta gene (ESR2) and cardiovascular disease in men and women. Clin Chem. 2007;53(10):1749-56.

112. Lo JC, Zhao X, Scuteri A, Brockwell S, Sowers MR. The association of genetic polymorphisms in sex hormone biosynthesis and action with insulin sensitivity and diabetes mellitus in women at midlife. Am J Med. 2006;119(9 Suppl 1):S69-78.
- 113113. Schuit SC, Oei HH, Witteman JC, Geurts van Kessel CH, van Meurs JB, Nijhuis RL, et al. Estrogen receptor alpha gene polymorphisms and risk of myocardial infarction. JAMA. 2004;291(24):2969-77.

Fat-derived adipokines and roles in cardiovascular disease risk

The main role of brown fat, mainly located around the neck and large blood vessels of the thorax, is to generate heat by “uncoupling” the respiratory chain of oxidative phosphorylation within mitochondria.114114. Trayhurn P. Hypoxia and adipocyte physiology: implications for adipose tissue dysfunction in obesity. Ann Rev Nutr. 2014;34:207-36. White visceral fat (abdominal fat) is mainly involved in a complex and multidirectional network of autocrine, paracrine, and endocrine signaling that crosstalk between organs and tissues. It is the white fat that mainly participates in the pathogenesis of metabolic diseases, such as type 2 diabetes mellitus, insulin resistance, hypertension, coronary heart disease, stroke, and HF.1717. Canepa M, Strait JB, Abramov D, Milaneschi Y, AlGhatrif M, Moni M, et al. Contribution of central adiposity to left ventricular diastolic function (from the Baltimore Longitudinal Study of Aging). Am J Cardiol. 2012;109(8):1171-8. , 115115. Romacho T, Elsen M, Rohrborn D, Eckel J. Adipose tissue and its role in organ crosstalk. Acta Physiol. 2014;210(4):733-53. , 116116. Xu A, Vanhoutte PM. Adiponectin and adipocyte fatty acid binding protein in the pathogenesis of cardiovascular disease. Am J Physiol Heart Circ Physiol. 2012;302(6):H1231-40. It is currently well accepted that adipose tissue is an active endocrine organ that secretes heterogeneous bioactive factors called adipokines,115115. Romacho T, Elsen M, Rohrborn D, Eckel J. Adipose tissue and its role in organ crosstalk. Acta Physiol. 2014;210(4):733-53. including cytokines and chemokines, vasoactive and coagulation factors, regulators of lipoprotein metabolism, and proteins, such as adiponectin and leptin.115115. Romacho T, Elsen M, Rohrborn D, Eckel J. Adipose tissue and its role in organ crosstalk. Acta Physiol. 2014;210(4):733-53.

In obesity, enlargement of adipose tissue mass has been linked to a dysregulation of adipokine secretion and the related tissue inflammation, which represents a critical pathogenic link between obesity and the development of cardiometabolic diseases.117117. Lehr S, Hartwig S, Lamers D, Famulla S, Muller S, Hanisch FG, et al. Identification and validation of novel adipokines released from primary human adipocytes. Mol Cell Proteom. MCP. 2012;11(1):M111 010504. In obese individuals, adipose tissue is infiltrated with activated macrophages and several other types of inflammatory cells, leading to an augmented production of proinflammatory adipokines, such as TNF-α, IL-6, monocyte chemoattractant protein (MCP)-1, resistin, leptin, lipocalin-2, adipocyte fatty acid binding protein (A-FABP), and plasminogen activator inhibitor-1.116116. Xu A, Vanhoutte PM. Adiponectin and adipocyte fatty acid binding protein in the pathogenesis of cardiovascular disease. Am J Physiol Heart Circ Physiol. 2012;302(6):H1231-40. These inflammatory factors are key components of the “adipo-cardiovascular axis” that mediates crosstalk between adipose tissue and the CV system.

Among the various adipose depots, perivascular adipose tissue is an important contributor to vascular inflammation because of its proximity to the blood vessel wall and its pronounced proinflammatory properties. Proinflammatory cytokines/adipokines released from other major adipose tissue depots, such as subcutaneous and abdominal fat, may further contribute to vascular inflammation by virtue of their endocrine actions.116116. Xu A, Vanhoutte PM. Adiponectin and adipocyte fatty acid binding protein in the pathogenesis of cardiovascular disease. Am J Physiol Heart Circ Physiol. 2012;302(6):H1231-40. These findings explain, in part, why WC may be considered a surrogate biomarker of CVD risk.

Adiponectin is one of the most abundant adipokines secreted by adipocytes, accounting for 0.01% of the total plasma protein content in humans.118118. Arita Y, Kihara S, Ouchi N, Takahashi M, Maeda K, Miyagawa J, et al. Paradoxical decrease of an adipose-specific protein, adiponectin, in obesity. Biochem Biophys Res Comm. 1999;257(1):79-83. The production of adiponectin from white adipocytes, which exerts beneficial effects on insulin sensitivity and cardiovascular function, is markedly reduced in obese individuals.116116. Xu A, Vanhoutte PM. Adiponectin and adipocyte fatty acid binding protein in the pathogenesis of cardiovascular disease. Am J Physiol Heart Circ Physiol. 2012;302(6):H1231-40. , 119119. Zhu W, Cheng KK, Vanhoutte PM, Lam KS, Xu A. Vascular effects of adiponectin: molecular mechanisms and potential therapeutic intervention. Clin Sci. 2008;114(5):361-74. , 120120. Tanaka K, Wilson RM, Essick EE, Duffen JL, Scherer PE, Ouchi N, et al. Effects of adiponectin on calcium-handling proteins in heart failure with preserved ejection fraction. Circ Heart Fail. 2014;7(6):976-85. Epidemiological studies show that low circulating adiponectin levels, particularly the high molecular weight form, is a risk factor for type 2 diabetes, hypertension, atherosclerosis, and myocardial infarction.116116. Xu A, Vanhoutte PM. Adiponectin and adipocyte fatty acid binding protein in the pathogenesis of cardiovascular disease. Am J Physiol Heart Circ Physiol. 2012;302(6):H1231-40.

Adiponectin and adiponectin receptors

The relationship between obesity and LVDD may be linked to adiponectin and adiponectin receptors. The full-length adiponectin consists of 247 amino acid residues, assembled into an N -terminal hypervariable region followed by a conserved collagenous domain of 22 Gly-Xaa-Yaa repeats and a C -terminal C1q-like globular domain.119119. Zhu W, Cheng KK, Vanhoutte PM, Lam KS, Xu A. Vascular effects of adiponectin: molecular mechanisms and potential therapeutic intervention. Clin Sci. 2008;114(5):361-74. In human and mouse plasma, adiponectin is present in three major oligomeric forms.119119. Zhu W, Cheng KK, Vanhoutte PM, Lam KS, Xu A. Vascular effects of adiponectin: molecular mechanisms and potential therapeutic intervention. Clin Sci. 2008;114(5):361-74. , 121121. Tsao TS, Murrey HE, Hug C, Lee DH, Lodish HF. Oligomerization state-dependent activation of NF-kappa B signaling pathway by adipocyte complement-related protein of 30 kDa (Acrp30). J Biol Chem. 2002;277(33):29359-62. , 122122. Xu A, Chan KW, Hoo RL, Wang Y, Tan KC, Zhang J, et al. Testosterone selectively reduces the high molecular weight form of adiponectin by inhibiting its secretion from adipocytes. J Biol Chem. 2005;280(18):18073-80. The monomeric form has never been detected under native conditions. The basic unit of oligomeric adiponectin is a homotrimer called low-molecular-weight (LMW) adiponectin.119119. Zhu W, Cheng KK, Vanhoutte PM, Lam KS, Xu A. Vascular effects of adiponectin: molecular mechanisms and potential therapeutic intervention. Clin Sci. 2008;114(5):361-74. , 123123. Pajvani UB, Du X, Combs TP, Berg AH, Rajala MW, Schulthess T, et al. Structure-function studies of the adipocyte-secreted hormone Acrp30/adiponectin. Implications fpr metabolic regulation and bioactivity. J Biol Chem.. 2003;278(11):9073-85. , 124124. Tsao TS, Tomas E, Murrey HE, Hug C, Lee DH, Ruderman NB, et al. Role of disulfide bonds in Acrp30/adiponectin structure and signaling specificity. Different oligomers activate different signal transduction pathways. J Biol Chem. 2003;278(50):50810-7. Two subunits of the adiponectin trimer are linked by a disulfide bond via cysteine residues in the collagen-like domain, which forms a hexamer termed middle-molecular-weight (MMW) adiponectin. This hexamer provides the building block for the formation of the bouquet-like high-molecular-weight (HMW) adiponectin comprised of 12-18 hexamers.

Post-translational modifications to the adiponectin protein are required for the intracellular assembly of the HMW oligomeric complex in adipocytes.125125. Richards AA, Stephens T, Charlton HK, Jones A, Macdonald GA, Prins JB, et al. Adiponectin multimerization is dependent on conserved lysines in the collagenous domain: evidence for regulation of multimerization by alterations in posttranslational modifications. Mol Endocrinol. 2006;20(7):1673-87. Different oligomeric forms of adiponectin act on different targets and possess distinct biological functions.119119. Zhu W, Cheng KK, Vanhoutte PM, Lam KS, Xu A. Vascular effects of adiponectin: molecular mechanisms and potential therapeutic intervention. Clin Sci. 2008;114(5):361-74.

The two main adiponectin receptors (AdipoRs), AdipoR1 and AdipoR2, are structurally and functionally distinct from classic G-protein-coupled receptors.126126. Yamauchi T, Kamon J, Ito Y, Tsuchida A, Yokomizo T, Kita S, et al. Cloning of adiponectin receptors that mediate antidiabetic metabolic effects. Nature. 2003;423(6941):762-9. AdipoR1 is expressed ubiquitously, whereas AdipoR2 is expressed most abundantly in the liver.126126. Yamauchi T, Kamon J, Ito Y, Tsuchida A, Yokomizo T, Kita S, et al. Cloning of adiponectin receptors that mediate antidiabetic metabolic effects. Nature. 2003;423(6941):762-9. Both AdipoR1 and AdipoR2 are expressed in cardiac cells,127127. Ding G, Qin Q, He N, Francis-David SC, Hou J, Liu J, et al. Adiponectin and its receptors are expressed in adult ventricular cardiomyocytes and upregulated by activation of peroxisome proliferator-activated receptor gamma.J Mol Cell Cardiol. 2007;43(1):73-84. but the exact roles of these two receptors in the antioxidative/nitrative stress and anti-inflammatory actions in cardiomyocytes remain unclear.

Although adipocytes are the major contributors to plasma adiponectin, adiponectin is also expressed in cardiomyocytes,127127. Ding G, Qin Q, He N, Francis-David SC, Hou J, Liu J, et al. Adiponectin and its receptors are expressed in adult ventricular cardiomyocytes and upregulated by activation of peroxisome proliferator-activated receptor gamma.J Mol Cell Cardiol. 2007;43(1):73-84. and cardiomyocyte-derived adiponectin is biologically active in protecting cells against ischemic injury via paracrine/autocrine activation of cardiac AdipoRs in mice.128128. Wang Y, Lau WB, Gao E, Tao L, Yuan Y, Li R, et al. Cardiomyocyte-derived adiponectin is biologically active in protecting against myocardial ischemia-reperfusion injury. Am J Physiol Endocrinol Metab. 2010;298(3):E663-70. In patients with dilated cardiomyopathy, cardiac adiponectin expression is downregulated.129129. Skurk C, Wittchen F, Suckau L, Witt H, Noutsias M, Fechner H, et al. Description of a local cardiac adiponectin system and its deregulation in dilated cardiomyopathy. Eur Heart J. 2008;29(9):1168-80.

Adiponectin and LVDD

In addition to AdipoRs, T-cadherin has also been suggested as a potential receptor for adiponectin,130130. Hug C, Wang J, Ahmad NS, Bogan JS, Tsao TS, Lodish HF. T-cadherin is a receptor for hexameric and high-molecular-weight forms of Acrp30/adiponectin. Proc Nat Acad Sci USA. 2004;101(28):10308-13. and it is highly expressed in the heart, smooth muscle, and endothelium, representing the main target of adiponectin in the cardiovascular system.131131. Ivanov D, Philippova M, Antropova J, Gubaeva F, Iljinskaya O, Tararak E, et al. Expression of cell adhesion molecule T-cadherin in the human vasculature. Histochem Cell Biol. 2001;115(3):231-42. , 132132. Doyle DD, Goings GE, Upshaw-Earley J, Page E, Ranscht B, Palfrey HC. T-cadherin is a major glycophosphoinositol-anchored protein associated with noncaveolar detergent-insoluble domains of the cardiac sarcolemma. J Biol Chem.. 1998;273(12):6937-43. T-cadherin is anchored at the cell surface by glycosyl phosphatidylinositol, and it plays an indispensable role in adiponectin-induced cardioprotection in mice,133133. Denzel MS, Scimia MC, Zumstein PM, Walsh K, Ruiz-Lozano P, Ranscht B. T-cadherin is critical for adiponectin-mediated cardioprotection in mice. J Clin Invest. 2010;120(12):4342-52. acting as a physiological adiponectin-binding receptor that enables the association of this adipokine with cardiac tissue.133133. Denzel MS, Scimia MC, Zumstein PM, Walsh K, Ruiz-Lozano P, Ranscht B. T-cadherin is critical for adiponectin-mediated cardioprotection in mice. J Clin Invest. 2010;120(12):4342-52.

As low levels of adiponectin have been linked to obesity-related cardiometabolic complications, its role in the maintenance of cardiac health should not be ignored.120120. Tanaka K, Wilson RM, Essick EE, Duffen JL, Scherer PE, Ouchi N, et al. Effects of adiponectin on calcium-handling proteins in heart failure with preserved ejection fraction. Circ Heart Fail. 2014;7(6):976-85. , 134134. Pou KM, Massaro JM, Hoffmann U, Vasan RS, Maurovich-Horvat P, Larson MG, et al. Visceral and subcutaneous adipose tissue volumes are cross-sectionally related to markers of inflammation and oxidative stress: the Framingham Heart Study. Circulation. 2007;116(11):1234-41. , 135135. Ouchi N, Shibata R, Walsh K. Cardioprotection by adiponectin. Trends Cardiovasc Med. 2006;16(5):141-6. Preclinical data show that adiponectin can attenuate or prevent the progression of LVDD to HFpEF.120120. Tanaka K, Wilson RM, Essick EE, Duffen JL, Scherer PE, Ouchi N, et al. Effects of adiponectin on calcium-handling proteins in heart failure with preserved ejection fraction. Circ Heart Fail. 2014;7(6):976-85. , 136136. Shibata R, Ouchi N, Ito M, Kihara S, Shiojima I, Pimentel DR, et al. Adiponectin-mediated modulation of hypertrophic signals in the heart. Nature Med. 2004;10(12):1384-9. , 137137. Sam F, Duhaney TA, Sato K, Wilson RM, Ohashi K, Sono-Romanelli S, et al. Adiponectin deficiency, diastolic dysfunction, and diastolic heart failure. Endocrinology. 2010;151(1):322-31. In a mouse model of aldosterone-induced HFpEF, Sam et al.137137. Sam F, Duhaney TA, Sato K, Wilson RM, Ohashi K, Sono-Romanelli S, et al. Adiponectin deficiency, diastolic dysfunction, and diastolic heart failure. Endocrinology. 2010;151(1):322-31. showed that a lack of adiponectin was associated with increased systolic blood pressure, LV remodeling, diastolic dysfunction, and pulmonary congestion, while the chronic hyperadiponectinemia in transgenic mice overexpressing adiponectin, reported by Tanaka et al.,120120. Tanaka K, Wilson RM, Essick EE, Duffen JL, Scherer PE, Ouchi N, et al. Effects of adiponectin on calcium-handling proteins in heart failure with preserved ejection fraction. Circ Heart Fail. 2014;7(6):976-85. ameliorated aldosterone-induced LV hypertrophy, diastolic dysfunction, and lung congestion, regardless of changes in blood pressure. The early filling-to-early mitral annular descent ratio, or E/e’, which was increased in the aldosterone-induced HFpEF mice and indicative of elevated filling pressures,137137. Sam F, Duhaney TA, Sato K, Wilson RM, Ohashi K, Sono-Romanelli S, et al. Adiponectin deficiency, diastolic dysfunction, and diastolic heart failure. Endocrinology. 2010;151(1):322-31. which was significantly attenuated in adiponectin transgenic mice. Tanaka et al.,120120. Tanaka K, Wilson RM, Essick EE, Duffen JL, Scherer PE, Ouchi N, et al. Effects of adiponectin on calcium-handling proteins in heart failure with preserved ejection fraction. Circ Heart Fail. 2014;7(6):976-85. also found that adiponectin overexpression decreased myocardial oxidative stress and calcium handling by preserving protein kinase A (PKA)-dependent phosphorylation of phospholamban. Moreover, adiponectin replacement in adiponectin knock-out mice, attenuated transmitral Doppler indices of pseudonormalization, indicative of impaired LV compliance.120120. Tanaka K, Wilson RM, Essick EE, Duffen JL, Scherer PE, Ouchi N, et al. Effects of adiponectin on calcium-handling proteins in heart failure with preserved ejection fraction. Circ Heart Fail. 2014;7(6):976-85. Taken together, these preclinical findings suggest that adiponectin may have a therapeutic potential in the management of LVDD and HFpEF, via direct actions on the heart.

Our observation that circulating adiponectin was not associated with LV fractional shortening is consistent with existing literature, which suggests that adiponectin acts primarily as an inhibitor of cardiac hypertrophy.

Clinical studies also support an association between adiponectin and cardiac function and structure.129129. Skurk C, Wittchen F, Suckau L, Witt H, Noutsias M, Fechner H, et al. Description of a local cardiac adiponectin system and its deregulation in dilated cardiomyopathy. Eur Heart J. 2008;29(9):1168-80. , 138138. Hui X, Lam KS, Vanhoutte PM, Xu A. Adiponectin and cardiovascular health: an update. Br J Pharmacol. 2012;165(3):574-90. In a community-based cohort study, McMannus et al.139139. McManus DD, Lyass A, Ingelsson E, Massaro JM, Meigs JB, Aragam J, et al. Relations of circulating resistin and adiponectin and cardiac structure and function: the Framingham Offspring Study. Obesity. 2012;20(9):1882-6. showed that relatively higher plasma levels of adiponectin associated with reduced LV mass, suggesting a cardioprotective effect. There are additional clinical data addressing the cardioprotective roles of adiponectin on the heart.140140. Hong SJ, Park CG, Seo HS, Oh DJ, Ro YM. Associations among plasma adiponectin, hypertension, left ventricular diastolic function and left ventricular mass index. Blood Press. 2004;13(4):236-42.

141. Norvik JV, Schirmer H, Ytrehus K, Jenssen TG, Zykova SN, Eggen AE, et al. Low adiponectin is associated with diastolic dysfunction in women: a cross-sectional study from the Tromso Study. BMC Cardiovasc Disord. 2017;17(1):79.
- 142142. Negi SI, Jeong EM, Shukrullah I, Raicu M, Dudley SC, Jr. Association of low plasma adiponectin with early diastolic dysfunction. Congest Heart Fail. 2012;18(4):187-91. For instance, Francisco et al.143143. Francisco C, Neves JS, Falcao-Pires I, Leite-Moreira A. Can Adiponectin Help us to Target Diastolic Dysfunction? Cardiovasc Dugs Ther. 2016;30(6):635-44. comprehensively reviewed the relevance of adiponectin signaling for the prevention of obesity related diastolic dysfunction with an emphasis on its role in limiting myocardial hypertrophy, cardiac fibrosis, nitrative and oxidative stress, atherosclerosis, and inflammation.

As there are several studies showing adiponectin’s modulatory role in the maintenance of diastolic function, it is worth mentioning that other investigators report no relationship to fat-associated LVDD. Sawada et al.144144. Sawada N, Daimon M, Kawata T, Nakao T, Kimura K, Nakanishi K, et al. The Significance of the Effect of Visceral Adiposity on Left Ventricular Diastolic Function in the General Population. Scientific Rep. 2019;9(1):4435. found that while plasma adiponectin levels significantly decreased with increased visceral adiposity in the general population, the association between adiponectin and diastolic function was not independent of fat. In other words, decreased circulating levels of adiponectin did not appear to have a central role in the association between visceral adiposity and LVDD. As this work was performed in a population with normal or grade 1 LVDD, subjects with more than moderate LVDD were not included. These authors suggested that a larger-scale study, including patients with moderate or severe LVDD are needed to confirm findings.

Sex differences, adiponectin, and cardiac function

Whether there are sex differences in the effects of adiponectin on cardiac function and structure is not clear. Fontes-Carvalho et al.145145. Fontes-Carvalho R, Pimenta J, Bettencourt P, Leite-Moreira A, Azevedo A. Association between plasma leptin and adiponectin levels and diastolic function in the general population. Exper Opin Ther Targets. 2015;19(10):1283-91. reported data from a population-base study involving individuals aged 45 years and older on associations of leptin and adiponectin levels and LV diastolic function. These investigators found significant sex differences for both leptin and adiponectin levels and their relationships with diastolic function. Relatively higher levels of leptin were associated with worse diastolic function, particularly among women, and this was regardless of age and hypertension. Interestingly, adiponectin did not correlate with diastolic function parameters.145145. Fontes-Carvalho R, Pimenta J, Bettencourt P, Leite-Moreira A, Azevedo A. Association between plasma leptin and adiponectin levels and diastolic function in the general population. Exper Opin Ther Targets. 2015;19(10):1283-91. Even so, it is plausible to postulate a sex-specific effect for adiponectin on changes in myocardial structure and function, as women have significantly higher systemic adiponectin levels.146146. Han SH, Quon MJ, Kim JA, Koh KK. Adiponectin and cardiovascular disease: response to therapeutic interventions. J Am Coll Cardiol 2007;49(5):531-8. In two small studies involving patients undergoing coronary angiography147147. Fukuta H, Ohte N, Wakami K, Goto T, Tani T, Kimura G. Relation of plasma levels of adiponectin to left ventricular diastolic dysfunction in patients undergoing cardiac catheterization for coronary artery disease. Am J Cardiol. 2011;108(8):1081-5. or with HF,142142. Negi SI, Jeong EM, Shukrullah I, Raicu M, Dudley SC, Jr. Association of low plasma adiponectin with early diastolic dysfunction. Congest Heart Fail. 2012;18(4):187-91. decreased adiponectin levels were associated with worse diastolic function. Figure 1 summarizes the main mechanisms involved in estrogen loss and obesity in the development of LVDD and HFpEF.

Figure 1
A schematic diagram showing the involvement of estrogen loss and obesity in HFpEF. An expansion and a shift of subcutaneous to visceral fat occurs in women after menopause. Abdominal obesity, defined by increased waist circumference, is a major risk factor for the development of HFpEF, which may involve an uptick in the synthesis and release of adipokines, including TNF-α, IL-6, MCP-1, resistin, leptin, lipocalin-2, and plasminogen activator inhibitor-1. These adipokines play critical roles in cardiac inflammation, oxidative stress, and impaired metabolism. On the other hand, the production of adiponectin from white adipocytes, which exerts beneficial effects on insulin sensitivity and cardiovascular function, is markedly reduced in obese individuals. Abnormalities in adipokines, in addition to estrogen loss, might participate in the development of HFpEF by inducing cardiac inflammation and oxidative stress, and finally leading to concentric cardiac hypertrophy, remodeling, stiffness, and diastolic dysfunction. HFpEF, heart failure with preserved ejection fraction; TNF-α, tumor necrosis factor alpha; IL-6, interleukin 6; MCP- 1, monocyte chemoattractant protein; A-FABP, adipocyte fatty acid binding protein .

Conclusion

In summary, estrogen plays a major role in the regulation of body weight and body fat, and that role may also protect the premenopausal heart from LV dysfunction. Compared with age-matched men, postmenopausal women display increased ventricular and arterial stiffness, and are more likely to develop LVDD and subsequent HFpEF. The lower estrogen levels after menopause are involved in changes in body fat distribution and content, a factor that increases the incidence of CVD. This review collected the recent evidence and clarified the molecular routes by which estrogen triggers these effects and provides new directions for future research in this exciting area of sex-specific cardiac aging and diastolic dysfunction, which is a harbinger for HFpEF.

Referências

  • 1
    Collaborators GBDO, Afshin A, Forouzanfar MH, Reitsma MB, Sur P, Estep K, et al. Health Effects of Overweight and Obesity in 195 Countries over 25 Years. N Engl J Med. 2017;377(1):13-27.
  • 2
    Guenancia C, Lefebvre A, Cardinale D, Yu AF, Ladoire S, Ghiringhelli F, et al. Obesity As a Risk Factor for Anthracyclines and Trastuzumab Cardiotoxicity in Breast Cancer: A Systematic Review and Meta-Analysis. J Clin Oncol. 2016;34(26):3157-65.
  • 3
    Alpert MA, Lavie CJ, Agrawal H, Aggarwal KB, Kumar SA. Obesity and heart failure: epidemiology, pathophysiology, clinical manifestations, and management. Translat Res. 2014;164(4):345-56.
  • 4
    Lee HJ, Kim HL, Lim WH, Seo JB, Kim SH, Zo JH, et al. Subclinical alterations in left ventricular structure and function according to obesity and metabolic health status. PloS one. 2019;14(9):e0222118.
  • 5
    Russo C, Sera F, Jin Z, Palmieri V, Homma S, Rundek T, et al. Abdominal adiposity, general obesity, and subclinical systolic dysfunction in the elderly: A population-based cohort study. Eur Heart J Fail.2016;18(5):537-44.
  • 6
    Yang H, Huynh QL, Venn AJ, Dwyer T, Marwick TH. Associations of childhood and adult obesity with left ventricular structure and function. Int J Obes. 2017;41(4):560-8.
  • 7
    Russo C, Jin Z, Homma S, Rundek T, Elkind MS, Sacco RL, et al. Effect of obesity and overweight on left ventricular diastolic function: a community-based study in an elderly cohort. J Am Coll Cardiol. 2011;57(12):1368-74.
  • 8
    Rayner JJ, Banerjee R, Holloway CJ, Lewis AJM, Peterzan MA, Francis JM, et al. The relative contribution of metabolic and structural abnormalities to diastolic dysfunction in obesity. Int J Obes (Lond). 2018;42(3):441-7.
  • 9
    Lee SL, Daimon M, Di Tullio MR, Homma S, Nakao T, Kawata T, et al. Relationship of Left Ventricular Diastolic Function to Obesity and Overweight in a Japanese Population With Preserved Left Ventricular Ejection Fraction. Circ J. 2016;80(9):1951-6.
  • 10
    Pandey A, Patel KV, Vaduganathan M, Sarma S, Haykowsky MJ, Berry JD, et al. Physical Activity, Fitness, and Obesity in Heart Failure With Preserved Ejection Fraction. JACC Heart Fail. 2018;6(12):975-82.
  • 11
    Bigaard J, Frederiksen K, Tjonneland A, Thomsen BL, Overvad K, Heitmann BL, et al. Waist circumference and body composition in relation to all-cause mortality in middle-aged men and women. Int J Obes. 2005;29(7):778-84.
  • 12
    Ashwell M, Hsieh SD. Six reasons why the waist-to-height ratio is a rapid and effective global indicator for health risks of obesity and how its use could simplify the international public health message on obesity. Int Nat Food Sci Nutr. 2005;56(5):303-7.
  • 13
    Ho SY, Lam TH, Janus ED, Hong Kong Cardiovascular Risk Factor Prevalence Study Steering C. Waist to stature ratio is more strongly associated with cardiovascular risk factors than other simple anthropometric indices. Ann Epidemiol. 2003;13(10):683-91.
  • 14
    Janssen I, Katzmarzyk PT, Ross R. Waist circumference and not body mass index explains obesity-related health risk. Am J Clin Nutr. 2004;79(3):379-84.
  • 15
    Wei M, Gaskill SP, Haffner SM, Stern MP. Waist circumference as the best predictor of noninsulin dependent diabetes mellitus (NIDDM) compared to body mass index, waist/hip ratio and other anthropometric measurements in Mexican Americans--a 7-year prospective study. Obes Res. 1997;5(1):16-23.
  • 16
    Welborn TA, Dhaliwal SS. Preferred clinical measures of central obesity for predicting mortality. Eur J Clin Nutr. 2007;61(12):1373-9.
  • 17
    Canepa M, Strait JB, Abramov D, Milaneschi Y, AlGhatrif M, Moni M, et al. Contribution of central adiposity to left ventricular diastolic function (from the Baltimore Longitudinal Study of Aging). Am J Cardiol. 2012;109(8):1171-8.
  • 18
    Haass M, Kitzman DW, Anand IS, Miller A, Zile MR, Massie BM, et al. Body mass index and adverse cardiovascular outcomes in heart failure patients with preserved ejection fraction: results from the Irbesartan in Heart Failure with Preserved Ejection Fraction (I-PRESERVE) trial. Circ Heart Fail. 2011;4(3):324-31.
  • 19
    Ho JE, Lyass A, Lee DS, Vasan RS, Kannel WB, Larson MG, et al. Predictors of new-onset heart failure: differences in preserved versus reduced ejection fraction. Circ Heart Fail. 2013;6(2):279-86.
  • 20
    Silverman DN, Shah SJ. Treatment of Heart Failure With Preserved Ejection Fraction (HFpEF): the Phenotype-Guided Approach. Curr Treat Options Cardiovasc Med. 2019;21(4):20.
  • 21
    Lam CSP, Chandramouli C. Fat, Female, Fatigued: Features of the Obese HFpEF Phenotype. JACC Heart Fail. 2018;6(8):710-3.
  • 22
    Lewis GA, Schelbert EB, Williams SG, Cunnington C, Ahmed F, McDonagh TA, et al. Biological Phenotypes of Heart Failure With Preserved Ejection Fraction. J Am Coll Cardiol. 2017;70(17):2186-200.
  • 23
    Mohammed SF, Borlaug BA, Roger VL, Mirzoyev SA, Rodeheffer RJ, Chirinos JA, et al. Comorbidity and ventricular and vascular structure and function in heart failure with preserved ejection fraction: a community-based study. Circ Heart Fail. 2012;5(6):710-9.
  • 24
    Lam CS, Roger VL, Rodeheffer RJ, Bursi F, Borlaug BA, Ommen SR, et al. Cardiac structure and ventricular-vascular function in persons with heart failure and preserved ejection fraction from Olmsted County, Minnesota. Circulation. 2007;115(15):1982-90.
  • 25
    Zile MR, Gottdiener JS, Hetzel SJ, McMurray JJ, Komajda M, McKelvie R, et al. Prevalence and significance of alterations in cardiac structure and function in patients with heart failure and a preserved ejection fraction. Circulation. 2011;124(23):2491-501.
  • 26
    Zile MR, Baicu CF, Gaasch WH. Diastolic heart failure--abnormalities in active relaxation and passive stiffness of the left ventricle. N Engl J Med. 2004;350(19):1953-9.
  • 27
    Maslov PZ, Kim JK, Argulian E, Ahmadi A, Narula N, Singh M, et al. Is Cardiac Diastolic Dysfunction a Part of Post-Menopausal Syndrome? JACC Heart Fail. 2019;7(3):192-203.
  • 28
    Pandey A, Omar W, Ayers C, LaMonte M, Klein L, Allen NB, et al. Sex and Race Differences in Lifetime Risk of Heart Failure With Preserved Ejection Fraction and Heart Failure With Reduced Ejection Fraction. Circulation. 2018;137(17):1814-23.
  • 29
    Upadhya B, Kitzman DW. Heart Failure with Preserved Ejection Fraction in Older Adults. Heart Fail Clin. 2017;13(3):485-502.
  • 30
    Benjamin EJ, Blaha MJ, Chiuve SE, Cushman M, Das SR, Deo R, et al. Heart Disease and Stroke Statistics-2017 Update: A Report From the American Heart Association. Circulation. 2017;135(10):e146-e603.
  • 31
    Fonarow GC, Stough WG, Abraham WT, Albert NM, Gheorghiade M, Greenberg BH, et al. Characteristics, treatments, and outcomes of patients with preserved systolic function hospitalized for heart failure: a report from the OPTIMIZE-HF Registry. J Am Coll Cardiol. 2007;50(8):768-77.
  • 32
    Alpert MA, Karthikeyan K, Abdullah O, Ghadban R. Obesity and Cardiac Remodeling in Adults: Mechanisms and Clinical Implications. Prog Cardiovasc Dis. 2018;61(2):114-23.
  • 33
    Ferron AJT, Francisqueti FV, Minatel IO, Silva C, Bazan SGZ, Kitawara KAH, et al. Association between Cardiac Remodeling and Metabolic Alteration in an Experimental Model of Obesity Induced by Western Diet. Nutrients. 2018;10(11).
  • 34
    Antonini-Canterin F, Di Nora C, Poli S, Sparacino L, Cosei I, Ravasel A, et al. Obesity, Cardiac Remodeling, and Metabolic Profile: Validation of a New Simple Index beyond Body Mass Index. J Cardiovasc Echogr. 2018;28(1):18-25.
  • 35
    Wohlfahrt P, Redfield MM, Lopez-Jimenez F, Melenovsky V, Kane GC, Rodeheffer RJ, et al. Impact of general and central adiposity on ventricular-arterial aging in women and men. JACC Heart failure. 2014;2(5):489-99.
  • 36
    Libhaber CD, Norton GR, Majane OH, Libhaber E, Essop MR, Brooksbank R, et al. Contribution of central and general adiposity to abnormal left ventricular diastolic function in a community sample with a high prevalence of obesity. Am J Cardiol. 2009;104(11):1527-33.
  • 37
    Fuster JJ, Ouchi N, Gokce N, Walsh K. Obesity-Induced Changes in Adipose Tissue Microenvironment and Their Impact on Cardiovascular Disease. Circ Res. 2016;118(11):1786-807.
  • 38
    Schiattarella GG, Altamirano F, Tong D, French KM, Villalobos E, Kim SY, et al. Nitrosative stress drives heart failure with preserved ejection fraction. Nature. 2019;568(7752):351-6.
  • 39
    Chirinos JA, Zamani P. The Nitrate-Nitrite-NO Pathway and Its Implications for Heart Failure and Preserved Ejection Fraction. Curr Heart Fail Rep. 2016;13(1):47-59.
  • 40
    Alvarez P, Briasoulis A. Immune Modulation in Heart Failure: the Promise of Novel Biologics. Current treatment. Options Cardiovasc Med. 2018 2018;20(3):26.
  • 41
    Hulsmans M, Sager HB, Roh JD, Valero-Munoz M, Houstis NE, Iwamoto Y, et al. Cardiac macrophages promote diastolic dysfunction. J Exper Med.. 2018;215(2):423-40.
  • 42
    Van Tassell BW, Trankle CR, Canada JM, Carbone S, Buckley L, Kadariya D, et al. IL-1 Blockade in Patients With Heart Failure With Preserved Ejection Fraction. Circ Heart Fail. 2018;11(8):e005036.
  • 43
    Van Tassell BW, Buckley LF, Carbone S, Trankle CR, Canada JM, Dixon DL, et al. Interleukin-1 blockade in heart failure with preserved ejection fraction: rationale and design of the Diastolic Heart Failure Anakinra Response Trial 2 (D-HART2). Clin Cardiol. 2017;40(9):626-32.
  • 44
    Garaulet M, Perez-Llamas F, Baraza JC, Garcia-Prieto MD, Fardy PS, Tebar FJ, et al. Body fat distribution in pre-and post-menopausal women: metabolic and anthropometric variables. J Nutr Health Aging. 2002;6(2):123-6.
  • 45
    Zamboni M, Armellini F, Milani MP, De Marchi M, Todesco T, Robbi R, et al. Body fat distribution in pre- and post-menopausal women: metabolic and anthropometric variables and their inter-relationships. Int In and related metabolic disorders : journal of the International Association for the Study of Obesity. Int J Obes.1992;16(7):495-504.
  • 46
    Toth MJ, Tchernof A, Sites CK, Poehlman ET. Menopause-related changes in body fat distribution. Ann NY Acad Sci. 2000;904:502-6.
  • 47
    Wehr E, Pilz S, Boehm BO, Marz W, Obermayer-Pietsch B. The lipid accumulation product is associated with increased mortality in normal weight postmenopausal women. Obesity. 2011;19(9):1873-80.
  • 48
    Joyce E, Lala A, Stevens SR, Cooper LB, AbouEzzeddine OF, Groarke JD, et al. Prevalence, Profile, and Prognosis of Severe Obesity in Contemporary Hospitalized Heart Failure Trial Populations. JACC Heart failure. 2016;4(12):923-31.
  • 49
    Kitzman DW, Brubaker P, Morgan T, Haykowsky M, Hundley G, Kraus WE, et al. Effect of Caloric Restriction or Aerobic Exercise Training on Peak Oxygen Consumption and Quality of Life in Obese Older Patients With Heart Failure With Preserved Ejection Fraction: A Randomized Clinical Trial. JAMA. 2016;315(1):36-46.
  • 50
    Kitzman DW, Brubaker PH, Herrington DM, Morgan TM, Stewart KP, Hundley WG, et al. Effect of endurance exercise training on endothelial function and arterial stiffness in older patients with heart failure and preserved ejection fraction: a randomized, controlled, single-blind trial. J Am Coll Cardiol. 2013;62(7):584-92.
  • 51
    Haykowsky MJ, Brubaker PH, Stewart KP, Morgan TM, Eggebeen J, Kitzman DW. Effect of endurance training on the determinants of peak exercise oxygen consumption in elderly patients with stable compensated heart failure and preserved ejection fraction. J Am Coll Cardiol. 2012;60(2):120-8.
  • 52
    Shin SH, Lee YJ, Heo YS, Park SD, Kwon SW, Woo SI, et al. Beneficial Effects of Bariatric Surgery on Cardiac Structure and Function in Obesity. Obes Surg. 2017;27(3):620-5.
  • 53
    Aggarwal R, Harling L, Efthimiou E, Darzi A, Athanasiou T, Ashrafian H. The Effects of Bariatric Surgery on Cardiac Structure and Function: a Systematic Review of Cardiac Imaging Outcomes. Obes Surg. 2016;26(5):1030-40.
  • 54
    Karason K, Wallentin I, Larsson B, Sjostrom L. Effects of obesity and weight loss on left ventricular mass and relative wall thickness: survey and intervention study. BMJ. 1997;315(7113):912-6.
  • 55
    Jhaveri RR, Pond KK, Hauser TH, Kissinger KV, Goepfert L, Schneider B, et al. Cardiac remodeling after substantial weight loss: a prospective cardiac magnetic resonance study after bariatric surgery. Surg Obes Relat Dis. 2009;5(6):648-52.
  • 56
    Cuspidi C, Rescaldani M, Tadic M, Sala C, Grassi G. Effects of bariatric surgery on cardiac structure and function: a systematic review and meta-analysis. Am J Hypertens. 2014;27(2):146-56.
  • 57
    Leichman JG, Wilson EB, Scarborough T, Aguilar D, Miller CC, 3rd, Yu S, et al. Dramatic reversal of derangements in muscle metabolism and left ventricular function after bariatric surgery. Am J Med. 2008;121(11):966-73.
  • 58
    Ikonomidis I, Mazarakis A, Papadopoulos C, Patsouras N, Kalfarentzos F, Lekakis J, et al. Weight loss after bariatric surgery improves aortic elastic properties and left ventricular function in individuals with morbid obesity: a 3-year follow-up study. J Hypert.2007;25(2):439-47.
  • 59
    Iancu ME, Copaescu C, Serban M, Ginghina C. Favorable changes in arterial elasticity, left ventricular mass, and diastolic function after significant weight loss following laparoscopic sleeve gastrectomy in obese individuals. Obes Surg. 2014;24(3):364-70.
  • 60
    Nyman K, Graner M, Pentikainen MO, Lundbom J, Hakkarainen A, Siren R, et al. Cardiac steatosis and left ventricular function in men with metabolic syndrome. J Cardiovasc Magn Reson. 2013;15:103.
  • 61
    Konishi M, Sugiyama S, Sugamura K, Nozaki T, Matsubara J, Akiyama E, et al. Accumulation of pericardial fat correlates with left ventricular diastolic dysfunction in patients with normal ejection fraction. J Cardiol. 2012;59(3):344-51.
  • 62
    Iacobellis G, Ribaudo MC, Assael F, Vecci E, Tiberti C, Zappaterreno A, et al. Echocardiographic epicardial adipose tissue is related to anthropometric and clinical parameters of metabolic syndrome: a new indicator of cardiovascular risk. J Clin Endocrinol Metab. 2003;88(11):5163-8.
  • 63
    Morelli NR, Scavuzzi BM, Miglioranza L, Lozovoy MAB, Simao ANC, Dichi I. Metabolic syndrome components are associated with oxidative stress in overweight and obese patients. Arch Endocrinol Metab. 2018;62(3):309-18.
  • 64
    Iacobellis G. Epicardial and pericardial fat: close, but very different. Obesity. 2009;17(4):625; author reply 6-7.
  • 65
    Ding J, Hsu FC, Harris TB, Liu Y, Kritchevsky SB, Szklo M, et al. The association of pericardial fat with incident coronary heart disease: the Multi-Ethnic Study of Atherosclerosis (MESA). Am J Clin Nutr. 2009;90(3):499-504.
  • 66
    Tamarappoo B, Dey D, Shmilovich H, Nakazato R, Gransar H, Cheng VY, et al. Increased pericardial fat volume measured from noncontrast CT predicts myocardial ischemia by SPECT. JACC Cardiovasc Imag. 2010;3(11):1104-12.
  • 67
    Aslanabadi N, Salehi R, Javadrashid A, Tarzamni M, Khodadad B, Enamzadeh E, et al. Epicardial and pericardial fat volume correlate with the severity of coronary artery stenosis. J Cardiovasc Thorac Res. 2014;6(4):235-9.
  • 68
    Cheng VY, Dey D, Tamarappoo B, Nakazato R, Gransar H, Miranda-Peats R, et al. Pericardial fat burden on ECG-gated noncontrast CT in asymptomatic patients who subsequently experience adverse cardiovascular events. JACC Cardiovasc Imag 2010;3(4):352-60.
  • 69
    Mahabadi AA, Berg MH, Lehmann N, Kalsch H, Bauer M, Kara K, et al. Association of epicardial fat with cardiovascular risk factors and incident myocardial infarction in the general population: the Heinz Nixdorf Recall Study. J Am Coll Cardiol. 2013;61(13):1388-95.
  • 70
    Alonso-Gomez AM, Tojal Sierra L, Fortuny Frau E, Goicolea Guemez L, Aboitiz Uribarri A, Portillo MP, et al. Diastolic dysfunction and exercise capacity in patients with metabolic syndrome and overweight/obesity. Int J Cardiol Heart Vasc. 2019;22:67-72.
  • 71
    Haykowsky MJ, Nicklas BJ, Brubaker PH, Hundley WG, Brinkley TE, Upadhya B, et al. Regional Adipose Distribution and its Relationship to Exercise Intolerance in Older Obese Patients Who Have Heart Failure With Preserved Ejection Fraction. JACC Heart failure. 2018;6(8):640-9.
  • 72
    Mazurek T, Zhang L, Zalewski A, Mannion JD, Diehl JT, Arafat H, et al. Human epicardial adipose tissue is a source of inflammatory mediators. Circulation. 2003;108(20):2460-6.
  • 73
    Sacks HS, Fain JN. Human epicardial adipose tissue: a review. Am Heart J. 2007;153(6):907-17.
  • 74
    Iacobellis G, Corradi D, Sharma AM. Epicardial adipose tissue: anatomic, biomolecular and clinical relationships with the heart. Nature clinical practice Cardiovasc Med. 2005;2(10):536-43.
  • 75
    Schram K, Sweeney G. Implications of myocardial matrix remodeling by adipokines in obesity-related heart failure. Trends Cardiovasc Med. 2008;18(6):199-205.
  • 76
    Packer M. Epicardial Adipose Tissue May Mediate Deleterious Effects of Obesity and Inflammation on the Myocardium. JJ Am Coll Cardiol. 2018;71(20):2360-72.
  • 77
    Patel VB, Basu R, Oudit GY. ACE2/Ang 1-7 axis: A critical regulator of epicardial adipose tissue inflammation and cardiac dysfunction in obesity. Adipocyte. 2016;5(3):306-11.
  • 78
    Patel VB, Mori J, McLean BA, Basu R, Das SK, Ramprasath T, et al. ACE2 Deficiency Worsens Epicardial Adipose Tissue Inflammation and Cardiac Dysfunction in Response to Diet-Induced Obesity. Diabetes. 2016;65(1):85-95.
  • 79
    Wu CK, Tsai HY, Su MM, Wu YF, Hwang JJ, Lin JL, et al. Evolutional change in epicardial fat and its correlation with myocardial diffuse fibrosis in heart failure patients. J Clin Lipidol. 2017;11(6):1421-31.
  • 80
    Lee JJ, Yin X, Hoffmann U, Fox CS, Benjamin EJ. Relation of Pericardial Fat, Intrathoracic Fat, and Abdominal Visceral Fat With Incident Atrial Fibrillation (from the Framingham Heart Study). Am J Cardiol. 2016;118(10):1486-92.
  • 81
    Fox CS, Gona P, Hoffmann U, Porter SA, Salton CJ, Massaro JM, et al. Pericardial fat, intrathoracic fat, and measures of left ventricular structure and function: the Framingham Heart Study. Circulation. 2009;119(12):1586-91.
  • 82
    Mak GJ, Ledwidge MT, Watson CJ, Phelan DM, Dawkins IR, Murphy NF, et al. Natural history of markers of collagen turnover in patients with early diastolic dysfunction and impact of eplerenone. J Am Coll Cardiol. 2009;54(18):1674-82.
  • 83
    van Woerden G, Gorter TM, Westenbrink BD, Willems TP, van Veldhuisen DJ, Rienstra M. Epicardial fat in heart failure patients with mid-range and preserved ejection fraction. Eur J Heart Fail. 2018;20(11):1559-66.
  • 84
    Obokata M, Reddy YNV, Pislaru SV, Melenovsky V, Borlaug BA. Evidence Supporting the Existence of a Distinct Obese Phenotype of Heart Failure With Preserved Ejection Fraction. Circulation. 2017;136(1):6-19.
  • 85
    Cavalcante JL, Tamarappoo BK, Hachamovitch R, Kwon DH, Alraies MC, Halliburton S, et al. Association of epicardial fat, hypertension, subclinical coronary artery disease, and metabolic syndrome with left ventricular diastolic dysfunction. Am J Cardiol. 2012;110(12):1793-8.
  • 86
    Brinkley TE, Ding J, Carr JJ, Nicklas BJ. Pericardial fat loss in postmenopausal women under conditions of equal energy deficit. Med Sci Sports Exerc. 2011;43(5):808-14.
  • 87
    Karastergiou K, Smith SR, Greenberg AS, Fried SK. Sex differences in human adipose tissues - the biology of pear shape. Biol Sex Differ. 2012;3(1):13.
  • 88
    Manolopoulos KN, Karpe F, Frayn KN. Gluteofemoral body fat as a determinant of metabolic health. Int J Obes (Lond). 2010;34(6):949-59.
  • 89
    Frank AP, de Souza Santos R, Palmer BF, Clegg DJ. Determinants of body fat distribution in humans may provide insight about obesity-related health risks. J Lipid Res. 2019;60(10):1710-9.
  • 90
    Mancuso P, Bouchard B. The Impact of Aging on Adipose Function and Adipokine Synthesis. Front Endocrinol (Lausanne). 2019;10:137.
  • 91
    Lobo RA, Davis SR, De Villiers TJ, Gompel A, Henderson VW, Hodis HN, et al. Prevention of diseases after menopause. Climacteric. 2014;17(5):540-56.
  • 92
    Redfield MM, Jacobsen SJ, Borlaug BA, Rodeheffer RJ, Kass DA. Age- and gender-related ventricular-vascular stiffening: a community-based study. Circulation. 2005;112(15):2254-62.
  • 93
    Regitz-Zagrosek V, Oertelt-Prigione S, Seeland U, Hetzer R. Sex and gender differences in myocardial hypertrophy and heart failure. Circ J. 2010;74(7):1265-73.
  • 94
    Gorodeski GI. Update on cardiovascular disease in post-menopausal women. Best Pract Res Clin Obstet Gynaecol. 2002;16(3):329-55.
  • 95
    Mendelsohn ME, Karas RH. The protective effects of estrogen on the cardiovascular system. N Engl J Med. 1999;340(23):1801-11.
  • 96
    Turgeon JL, McDonnell DP, Martin KA, Wise PM. Hormone therapy: physiological complexity belies therapeutic simplicity. Science. 2004;304(5675):1269-73.
  • 97
    Watson CS, Jeng YJ, Kochukov MY. Nongenomic actions of estradiol compared with estrone and estriol in pituitary tumor cell signaling and proliferation. FASEB J. 2008;22(9):3328-36.
  • 98
    Haas E, Bhattacharya I, Brailoiu E, Damjanovic M, Brailoiu GC, Gao X, et al. Regulatory role of G protein-coupled estrogen receptor for vascular function and obesity. Circ Res. 2009;104(3):288-91.
  • 99
    Ueda K, Adachi Y, Liu P, Fukuma N, Takimoto E. Regulatory Actions of Estrogen Receptor Signaling in the Cardiovascular System. Front Endocrinol (Lausanne). 2019;10:909.
  • 100
    Iorga A, Cunningham CM, Moazeni S, Ruffenach G, Umar S, Eghbali M. The protective role of estrogen and estrogen receptors in cardiovascular disease and the controversial use of estrogen therapy. Biol Sex Differ. 2017;8(1):33.
  • 101
    Dworatzek E, Mahmoodzadeh S. Targeted basic research to highlight the role of estrogen and estrogen receptors in the cardiovascular system. Pharmacol Res. 2017;119:27-35.
  • 102
    Mayes JS, Watson GH. Direct effects of sex steroid hormones on adipose tissues and obesity. Obes Rev. 2004;5(4):197-216.
  • 103
    Bouchard C, Despres JP, Mauriege P. Genetic and nongenetic determinants of regional fat distribution. Endocr Rev. 1993;14(1):72-93.
  • 104
    Brown LM, Clegg DJ. Central effects of estradiol in the regulation of food intake, body weight, and adiposity. J Steroid Biochem Mol Biol. 2010;122(1-3):65-73.
  • 105
    Clegg DJ, Brown LM, Woods SC, Benoit SC. Gonadal hormones determine sensitivity to central leptin and insulin. Diabetes. 2006;55(4):978-87.
  • 106
    Stubbins RE, Holcomb VB, Hong J, Nunez NP. Estrogen modulates abdominal adiposity and protects female mice from obesity and impaired glucose tolerance. Eur J Nutr. 2012;51(7):861-70.
  • 107
    Zidon TM, Padilla J, Fritsche KL, Welly RJ, McCabe LT, Stricklin OE, et al. Effects of ERbeta and ERalpha on OVX-induced changes in adiposity and insulin resistance. J Endocrinol. 2020;245(1):165-78.
  • 108
    Crandall DL, Busler DE, Novak TJ, Weber RV, Kral JG. Identification of estrogen receptor beta RNA in human breast and abdominal subcutaneous adipose tissue. Biochem Biophys Res Comm. 1998;248(3):523-6.
  • 109
    Matelski H, Greene R, Huberman M, Lokich J, Zipoli T. Randomized trial of estrogen vs. tamoxifen therapy for advanced breast cancer. Am J Clin Oncol. 1985;8(2):128-33.
  • 110
    Kuiper GG, Enmark E, Pelto-Huikko M, Nilsson S, Gustafsson JA. Cloning of a novel receptor expressed in rat prostate and ovary. Proc Nat Acad Sci USA. 1996;93(12):5925-30.
  • 111
    Rexrode KM, Ridker PM, Hegener HH, Buring JE, Manson JE, Zee RY. Polymorphisms and haplotypes of the estrogen receptor-beta gene (ESR2) and cardiovascular disease in men and women. Clin Chem. 2007;53(10):1749-56.
  • 112
    Lo JC, Zhao X, Scuteri A, Brockwell S, Sowers MR. The association of genetic polymorphisms in sex hormone biosynthesis and action with insulin sensitivity and diabetes mellitus in women at midlife. Am J Med. 2006;119(9 Suppl 1):S69-78.
  • 113
    Schuit SC, Oei HH, Witteman JC, Geurts van Kessel CH, van Meurs JB, Nijhuis RL, et al. Estrogen receptor alpha gene polymorphisms and risk of myocardial infarction. JAMA. 2004;291(24):2969-77.
  • 114
    Trayhurn P. Hypoxia and adipocyte physiology: implications for adipose tissue dysfunction in obesity. Ann Rev Nutr. 2014;34:207-36.
  • 115
    Romacho T, Elsen M, Rohrborn D, Eckel J. Adipose tissue and its role in organ crosstalk. Acta Physiol. 2014;210(4):733-53.
  • 116
    Xu A, Vanhoutte PM. Adiponectin and adipocyte fatty acid binding protein in the pathogenesis of cardiovascular disease. Am J Physiol Heart Circ Physiol. 2012;302(6):H1231-40.
  • 117
    Lehr S, Hartwig S, Lamers D, Famulla S, Muller S, Hanisch FG, et al. Identification and validation of novel adipokines released from primary human adipocytes. Mol Cell Proteom. MCP. 2012;11(1):M111 010504.
  • 118
    Arita Y, Kihara S, Ouchi N, Takahashi M, Maeda K, Miyagawa J, et al. Paradoxical decrease of an adipose-specific protein, adiponectin, in obesity. Biochem Biophys Res Comm. 1999;257(1):79-83.
  • 119
    Zhu W, Cheng KK, Vanhoutte PM, Lam KS, Xu A. Vascular effects of adiponectin: molecular mechanisms and potential therapeutic intervention. Clin Sci. 2008;114(5):361-74.
  • 120
    Tanaka K, Wilson RM, Essick EE, Duffen JL, Scherer PE, Ouchi N, et al. Effects of adiponectin on calcium-handling proteins in heart failure with preserved ejection fraction. Circ Heart Fail. 2014;7(6):976-85.
  • 121
    Tsao TS, Murrey HE, Hug C, Lee DH, Lodish HF. Oligomerization state-dependent activation of NF-kappa B signaling pathway by adipocyte complement-related protein of 30 kDa (Acrp30). J Biol Chem. 2002;277(33):29359-62.
  • 122
    Xu A, Chan KW, Hoo RL, Wang Y, Tan KC, Zhang J, et al. Testosterone selectively reduces the high molecular weight form of adiponectin by inhibiting its secretion from adipocytes. J Biol Chem. 2005;280(18):18073-80.
  • 123
    Pajvani UB, Du X, Combs TP, Berg AH, Rajala MW, Schulthess T, et al. Structure-function studies of the adipocyte-secreted hormone Acrp30/adiponectin. Implications fpr metabolic regulation and bioactivity. J Biol Chem.. 2003;278(11):9073-85.
  • 124
    Tsao TS, Tomas E, Murrey HE, Hug C, Lee DH, Ruderman NB, et al. Role of disulfide bonds in Acrp30/adiponectin structure and signaling specificity. Different oligomers activate different signal transduction pathways. J Biol Chem. 2003;278(50):50810-7.
  • 125
    Richards AA, Stephens T, Charlton HK, Jones A, Macdonald GA, Prins JB, et al. Adiponectin multimerization is dependent on conserved lysines in the collagenous domain: evidence for regulation of multimerization by alterations in posttranslational modifications. Mol Endocrinol. 2006;20(7):1673-87.
  • 126
    Yamauchi T, Kamon J, Ito Y, Tsuchida A, Yokomizo T, Kita S, et al. Cloning of adiponectin receptors that mediate antidiabetic metabolic effects. Nature. 2003;423(6941):762-9.
  • 127
    Ding G, Qin Q, He N, Francis-David SC, Hou J, Liu J, et al. Adiponectin and its receptors are expressed in adult ventricular cardiomyocytes and upregulated by activation of peroxisome proliferator-activated receptor gamma.J Mol Cell Cardiol. 2007;43(1):73-84.
  • 128
    Wang Y, Lau WB, Gao E, Tao L, Yuan Y, Li R, et al. Cardiomyocyte-derived adiponectin is biologically active in protecting against myocardial ischemia-reperfusion injury. Am J Physiol Endocrinol Metab. 2010;298(3):E663-70.
  • 129
    Skurk C, Wittchen F, Suckau L, Witt H, Noutsias M, Fechner H, et al. Description of a local cardiac adiponectin system and its deregulation in dilated cardiomyopathy. Eur Heart J. 2008;29(9):1168-80.
  • 130
    Hug C, Wang J, Ahmad NS, Bogan JS, Tsao TS, Lodish HF. T-cadherin is a receptor for hexameric and high-molecular-weight forms of Acrp30/adiponectin. Proc Nat Acad Sci USA. 2004;101(28):10308-13.
  • 131
    Ivanov D, Philippova M, Antropova J, Gubaeva F, Iljinskaya O, Tararak E, et al. Expression of cell adhesion molecule T-cadherin in the human vasculature. Histochem Cell Biol. 2001;115(3):231-42.
  • 132
    Doyle DD, Goings GE, Upshaw-Earley J, Page E, Ranscht B, Palfrey HC. T-cadherin is a major glycophosphoinositol-anchored protein associated with noncaveolar detergent-insoluble domains of the cardiac sarcolemma. J Biol Chem.. 1998;273(12):6937-43.
  • 133
    Denzel MS, Scimia MC, Zumstein PM, Walsh K, Ruiz-Lozano P, Ranscht B. T-cadherin is critical for adiponectin-mediated cardioprotection in mice. J Clin Invest. 2010;120(12):4342-52.
  • 134
    Pou KM, Massaro JM, Hoffmann U, Vasan RS, Maurovich-Horvat P, Larson MG, et al. Visceral and subcutaneous adipose tissue volumes are cross-sectionally related to markers of inflammation and oxidative stress: the Framingham Heart Study. Circulation. 2007;116(11):1234-41.
  • 135
    Ouchi N, Shibata R, Walsh K. Cardioprotection by adiponectin. Trends Cardiovasc Med. 2006;16(5):141-6.
  • 136
    Shibata R, Ouchi N, Ito M, Kihara S, Shiojima I, Pimentel DR, et al. Adiponectin-mediated modulation of hypertrophic signals in the heart. Nature Med. 2004;10(12):1384-9.
  • 137
    Sam F, Duhaney TA, Sato K, Wilson RM, Ohashi K, Sono-Romanelli S, et al. Adiponectin deficiency, diastolic dysfunction, and diastolic heart failure. Endocrinology. 2010;151(1):322-31.
  • 138
    Hui X, Lam KS, Vanhoutte PM, Xu A. Adiponectin and cardiovascular health: an update. Br J Pharmacol. 2012;165(3):574-90.
  • 139
    McManus DD, Lyass A, Ingelsson E, Massaro JM, Meigs JB, Aragam J, et al. Relations of circulating resistin and adiponectin and cardiac structure and function: the Framingham Offspring Study. Obesity. 2012;20(9):1882-6.
  • 140
    Hong SJ, Park CG, Seo HS, Oh DJ, Ro YM. Associations among plasma adiponectin, hypertension, left ventricular diastolic function and left ventricular mass index. Blood Press. 2004;13(4):236-42.
  • 141
    Norvik JV, Schirmer H, Ytrehus K, Jenssen TG, Zykova SN, Eggen AE, et al. Low adiponectin is associated with diastolic dysfunction in women: a cross-sectional study from the Tromso Study. BMC Cardiovasc Disord. 2017;17(1):79.
  • 142
    Negi SI, Jeong EM, Shukrullah I, Raicu M, Dudley SC, Jr. Association of low plasma adiponectin with early diastolic dysfunction. Congest Heart Fail. 2012;18(4):187-91.
  • 143
    Francisco C, Neves JS, Falcao-Pires I, Leite-Moreira A. Can Adiponectin Help us to Target Diastolic Dysfunction? Cardiovasc Dugs Ther. 2016;30(6):635-44.
  • 144
    Sawada N, Daimon M, Kawata T, Nakao T, Kimura K, Nakanishi K, et al. The Significance of the Effect of Visceral Adiposity on Left Ventricular Diastolic Function in the General Population. Scientific Rep. 2019;9(1):4435.
  • 145
    Fontes-Carvalho R, Pimenta J, Bettencourt P, Leite-Moreira A, Azevedo A. Association between plasma leptin and adiponectin levels and diastolic function in the general population. Exper Opin Ther Targets. 2015;19(10):1283-91.
  • 146
    Han SH, Quon MJ, Kim JA, Koh KK. Adiponectin and cardiovascular disease: response to therapeutic interventions. J Am Coll Cardiol 2007;49(5):531-8.
  • 147
    Fukuta H, Ohte N, Wakami K, Goto T, Tani T, Kimura G. Relation of plasma levels of adiponectin to left ventricular diastolic dysfunction in patients undergoing cardiac catheterization for coronary artery disease. Am J Cardiol. 2011;108(8):1081-5.
  • Study Association
    This study is not associated with any thesis or dissertation work.
  • Ethics approval and consent to participate
    This article does not contain any studies with human participants or animals performed by any of the authors.
  • Sources of Funding: This study was funded by National Institute on Aging of the National Institute of Health to Leanne Groban (AG033727; AG061588) and partially funded by Conselho Nacional de Desenvolvimento Científico e Tecnológico (CNPq), Coordenação de Aperfeiçoamento de Pessoal de Nível Superior (CAPES), and Fundação Carlos Chagas Filho de Amparo à Pesquisa do Estado do Rio de Janeiro (FAPERJ) to Gisele Zapata-Sudo and Allan Kardec Nogueira de Alencar.

Publication Dates

  • Publication in this collection
    08 Oct 2021
  • Date of issue
    Dec 2021

History

  • Received
    31 July 2020
  • Reviewed
    16 Dec 2020
  • Accepted
    27 Jan 2021
Sociedade Brasileira de Cardiologia - SBC Avenida Marechal Câmara, 160, sala: 330, Centro, CEP: 20020-907, (21) 3478-2700 - Rio de Janeiro - RJ - Brazil, Fax: +55 21 3478-2770 - São Paulo - SP - Brazil
E-mail: revista@cardiol.br