Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 2.253
Filter
1.
Ann Med ; 56(1): 2387302, 2024 Dec.
Article in English | MEDLINE | ID: mdl-39101236

ABSTRACT

BACKGROUND: Cushing's syndrome (CS) is associated with increased risk for heart failure, which often initially manifests as left ventricular diastolic dysfunction (LVDD). In this study, we aimed to explore the potential risk factors of LVDD in CS by incorporating body composition parameters. METHODS: A retrospective study was conducted on patients diagnosed with endogenous CS no less than 18 years old. The control group consisted of healthy individuals who were matched to CS patients in terms of gender, age, and BMI. LIFEx software (version 7.3) was applied to measure epicardial adipose tissue volume (EATV) on non-contrast chest CT, as well as abdominal adipose tissue and skeletal muscle mass at the first lumbar vertebral level. Echocardiography was used to evaluate left ventricular (LV) diastolic function. Body compositions and clinical data were examined in relation to early LVDD. RESULTS: A total of 86 CS patients and 86 healthy controls were enrolled. EATV was significantly higher in CS patients compared to control subjects (150.33 cm3 [125.67, 189.41] vs 90.55 cm3 [66.80, 119.84], p < 0.001). CS patients had noticeably increased visceral fat but decreased skeletal muscle in comparison to their healthy counterparts. Higher prevalence of LVDD was found in CS patients based on LV diastolic function evaluated by E/A ratio (p < 0.001). EATV was proved to be an independent risk factor for LVDD in CS patients (OR = 1.015, 95%CI 1.003-1.026, p = 0.011). If the cut-point of EATV was set as 139.252 cm3 in CS patients, the diagnostic sensitivity and specificity of LVDD were 84.00% and 55.60%, respectively. CONCLUSION: CS was associated with marked accumulation of EAT and visceral fat, reduced skeletal muscle mass, and increased prevalence of LVDD. EATV was an independent risk factor for LVDD, suggesting the potential role of EAT in the development of LVDD in CS.


This study explored the potential risk factors of LVDD in endogenous CS by incorporating body composition parameters. EATV was identified as an independent risk factor for LVDD. Targeted therapeutic interventions to reduce excessive cortisol-induced EAT accumulation may be promising to mitigate the risk of LVDD development in patients with CS.


Subject(s)
Adipose Tissue , Cushing Syndrome , Echocardiography , Pericardium , Ventricular Dysfunction, Left , Humans , Male , Cushing Syndrome/physiopathology , Cushing Syndrome/complications , Cushing Syndrome/epidemiology , Female , Retrospective Studies , Adult , Ventricular Dysfunction, Left/physiopathology , Ventricular Dysfunction, Left/diagnostic imaging , Ventricular Dysfunction, Left/epidemiology , Ventricular Dysfunction, Left/etiology , Pericardium/diagnostic imaging , Adipose Tissue/diagnostic imaging , Adipose Tissue/physiopathology , Middle Aged , Diastole , Risk Factors , Case-Control Studies , Tomography, X-Ray Computed , Epicardial Adipose Tissue
2.
Cardiovasc Diabetol ; 23(1): 308, 2024 Aug 22.
Article in English | MEDLINE | ID: mdl-39175063

ABSTRACT

BACKGROUND: Albuminuria is considered an early and sensitive marker of kidney dysfunction, but also an independent cardiovascular risk factor. Considering the possible relationship among metabolic liver disease, cardiovascular disease and chronic kidney disease, we aimed to evaluate the risk of developing albuminuria regarding the presence of epicardial adipose tissue and the steatotic liver disease status. METHODS: A retrospective long-term longitudinal study including 181 patients was carried out. Epicardial adipose tissue and steatotic liver disease were assessed by computed tomography. The presence of albuminuria at follow-up was defined as the outcome. RESULTS: After a median follow up of 11.2 years, steatotic liver disease (HR 3.15; 95% CI, 1.20-8.26; p = 0.02) and excess amount of epicardial adipose tissue (HR 6.12; 95% CI, 1.69-22.19; p = 0.006) were associated with an increased risk of albuminuria after adjustment for visceral adipose tissue, sex, age, weight status, type 2 diabetes, prediabetes, hypertriglyceridemia, hypercholesterolemia, arterial hypertension, and cardiovascular prevention treatment at baseline. The presence of both conditions was associated with a higher risk of developing albuminuria compared to having steatotic liver disease alone (HR 5.91; 95% CI 1.15-30.41, p = 0.033). Compared with the first tertile of visceral adipose tissue, the proportion of subjects with liver steatosis and abnormal epicardial adipose tissue was significantly higher in the second and third tertile. We found a significant correlation between epicardial fat and steatotic liver disease (rho = 0.43 [p < 0.001]). CONCLUSIONS: Identification and management/decrease of excess adiposity must be a target in the primary and secondary prevention of chronic kidney disease development and progression. Visceral adiposity assessment may be an adequate target in the daily clinical setting. Moreover, epicardial adipose tissue and steatotic liver disease assessment may aid in the primary prevention of renal dysfunction.


Subject(s)
Adiposity , Albuminuria , Fatty Liver , Pericardium , Humans , Retrospective Studies , Male , Female , Pericardium/diagnostic imaging , Albuminuria/epidemiology , Albuminuria/diagnosis , Albuminuria/physiopathology , Middle Aged , Risk Factors , Aged , Fatty Liver/epidemiology , Fatty Liver/diagnosis , Fatty Liver/physiopathology , Longitudinal Studies , Time Factors , Adipose Tissue/diagnostic imaging , Adipose Tissue/physiopathology , Adipose Tissue/metabolism , Risk Assessment , Liver/diagnostic imaging , Liver/pathology , Intra-Abdominal Fat/physiopathology , Intra-Abdominal Fat/diagnostic imaging , Adult
3.
Nat Rev Rheumatol ; 20(9): 565-584, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39112603

ABSTRACT

Obesity has a pivotal and multifaceted role in pain associated with osteoarthritis (OA), extending beyond the mechanistic influence of BMI. It exerts its effects both directly and indirectly through various modifiable risk factors associated with OA-related pain. Adipose tissue dysfunction is highly involved in OA-related pain through local and systemic inflammation, immune dysfunction, and the production of pro-inflammatory cytokines and adipokines. Adipose tissue dysfunction is intricately connected with metabolic syndrome, which independently exerts specific effects on OA-related pain, distinct from its association with BMI. The interplay among obesity, adipose tissue dysfunction and metabolic syndrome influences OA-related pain through diverse pain mechanisms, including nociceptive pain, peripheral sensitization and central sensitization. These complex interactions contribute to the heightened pain experience observed in individuals with OA and obesity. In addition, pain management strategies are less efficient in individuals with obesity. Importantly, therapeutic interventions targeting obesity and metabolic syndrome hold promise in managing OA-related pain. A deeper understanding of the intricate relationship between obesity, metabolic syndrome and OA-related pain is crucial and could have important implications for improving pain management and developing innovative therapeutic options in OA.


Subject(s)
Adipose Tissue , Metabolic Syndrome , Obesity , Osteoarthritis , Humans , Obesity/complications , Obesity/physiopathology , Osteoarthritis/physiopathology , Osteoarthritis/complications , Adipose Tissue/physiopathology , Adipose Tissue/metabolism , Metabolic Syndrome/physiopathology , Metabolic Syndrome/complications , Pain/physiopathology , Pain Management/methods
4.
Lipids Health Dis ; 23(1): 277, 2024 Aug 31.
Article in English | MEDLINE | ID: mdl-39217346

ABSTRACT

BACKGROUND: The global prevalence of obesity has escalated into a formidable health challenge intricately linked with the risk of developing cardiac diastolic disfunction and heart failure with preserved ejection fraction (HFpEF). Abnormal fat distribution is potentially strongly associated with an increased risk of cardiac diastolic dysfunction, and we aimed to scrutinize and elucidate the correlation between them. METHODS: Following the Cochrane Handbook and PRISMA 2020 guidelines, we systematically reviewed the literature from PubMed, Embase, and Web of Science. We focused on studies reporting the mean and standard deviation (SD) of abnormal fat in HFpEF or cardiac diastolic dysfunction patients and the Pearson/Spearman correlation coefficients for the relationship between abnormal fat distribution and the risk of developing cardiac diastolic dysfunction. Data were standardized to the standard mean difference (SMD) and Fisher's z value for meta-analysis. RESULTS: After progressive filtering and selection, 63 studies (43,113 participants) were included in the quantitative analyses. Abnormal fat distribution was significantly greater in participants with cardiac diastolic dysfunction than in controls [SMD 0.88 (0.69, 1.08)], especially in epicardial adipose tissue [SMD 0.99 (0.73, 1.25)]. Abnormal fat distribution was significantly correlated with the risk of developing cardiac diastolic dysfunction [E/E': 0.23 (0.18, 0.27), global longitudinal strain: r=-0.11 (-0.24, 0.02)]. Meta-regression revealed sample size as a potential heterogeneous source, and subgroup analyses revealed a stronger association between abnormal fat distribution and the risk of developing cardiac diastolic dysfunction in the overweight and obese population. CONCLUSION: Abnormal fat distribution was significantly associated with the risk of developing cardiac diastolic dysfunction. TRIAL REGISTRATION: CRD42024543774.


Subject(s)
Heart Failure , Humans , Heart Failure/physiopathology , Stroke Volume , Obesity/physiopathology , Obesity/complications , Diastole , Observational Studies as Topic , Female , Adipose Tissue/physiopathology , Male , Risk Factors
5.
J Endocrinol ; 262(3)2024 Sep 01.
Article in English | MEDLINE | ID: mdl-38967989

ABSTRACT

The root cause of type 2 diabetes (T2D) is insulin resistance (IR), defined by the failure of cells to respond to circulating insulin to maintain lipid and glucose homeostasis. While the causes of whole-body insulin resistance are multifactorial, a major contributing factor is dysregulation of liver and adipose tissue function. Adipose dysfunction, particularly adipose tissue-IR (adipo-IR), plays a crucial role in the development of hepatic insulin resistance and the progression of metabolic dysfunction-associated steatotic liver disease (MASLD) in the context of T2D. In this review, we will focus on molecular mechanisms of hepatic insulin resistance and its association with adipose tissue function. A deeper understanding of the pathophysiological mechanisms of the transition from a healthy state to insulin resistance, impaired glucose tolerance, and T2D may enable us to prevent and intervene in the progression to T2D.


Subject(s)
Adipose Tissue , Diabetes Mellitus, Type 2 , Insulin Resistance , Liver , Humans , Insulin Resistance/physiology , Diabetes Mellitus, Type 2/metabolism , Diabetes Mellitus, Type 2/physiopathology , Adipose Tissue/metabolism , Adipose Tissue/physiopathology , Liver/metabolism , Animals , Fatty Liver/metabolism , Fatty Liver/physiopathology
6.
Echocardiography ; 41(7): e15872, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38940234

ABSTRACT

BACKGROUND: Obesity is a significant risk factor for heart failure with preserved ejection fraction (HFpEF). In this study, we explore the relationships between body mass index (BMI) and adipose tissue compartments such as visceral adipose tissue (VAT), subcutaneous adipose tissue (SAT), and epicardial adipose tissue (EAT), with respect to left ventricular (LV) structure and function in subjects with preserved LV systolic function. METHODS: Between January and December 2020, this retrospective study included 749 participants who exhibited preserved LV systolic function and underwent transthoracic echocardiography along with abdominal computed tomography. LV structural and functional variables as well as EAT, VAT, and SAT thickness were evaluated using echocardiography and computed tomography. RESULTS: SAT decreased, while VAT and EAT progressively increased with age. There were significant correlations between BMI and various adipose tissues, with the strongest correlation observed with SAT (r = .491, p < .001) compared to VAT (r = .371, p < .001) or EAT (r = .135, p < .001). However, EAT demonstrated the most substantial association with decreased LV end-diastolic dimension, LV end-systolic dimension, and septal mitral annular velocity and increased relative wall thickness (all p < .05), while VAT and SAT did not show significant associations with LV remodeling and functional parameters after adjusting for clinical variables. CONCLUSION: EAT is the most critical adipose tissue influencing LV geometric and functional changes, compared with VAT or SAT. Thick EAT is associated small LV chamber size, concentric remodeling, and relaxation abnormalities.


Subject(s)
Adiposity , Echocardiography , Ventricular Remodeling , Humans , Male , Female , Retrospective Studies , Ventricular Remodeling/physiology , Adiposity/physiology , Aged , Echocardiography/methods , Middle Aged , Ventricular Dysfunction, Left/physiopathology , Ventricular Dysfunction, Left/diagnostic imaging , Ventricular Dysfunction, Left/etiology , Diastole , Heart Ventricles/diagnostic imaging , Heart Ventricles/physiopathology , Body Mass Index , Adipose Tissue/diagnostic imaging , Adipose Tissue/physiopathology , Ventricular Function, Left/physiology
7.
Int J Cardiol ; 412: 132303, 2024 Oct 01.
Article in English | MEDLINE | ID: mdl-38944349

ABSTRACT

Heart failure is an important cause of mortality and morbidity worldwide. Heart failure with preserved ejection fraction (HFpEF) incidence and prevalence is increasing, and the phenotype associated with obesity is the most frequent. Epicardial adipose tissue (EAT) is directly associated with systemic obesity and several previous studies have shown a clear link between EAT and HFpEF. Moreover, the restriction induced by the pericardium is also linked to HFpEF. In this review we will describe the epidemiological association between the pericardium, EAT and HFpEF, how to quantify EAT, what are the pathophysiological mechanism to explain theses association and how can the pericardium and EAT be a treatment target in patients with HFpEF.


Subject(s)
Adipose Tissue , Heart Failure , Pericardium , Stroke Volume , Humans , Pericardium/physiopathology , Pericardium/diagnostic imaging , Heart Failure/physiopathology , Heart Failure/therapy , Adipose Tissue/physiopathology , Stroke Volume/physiology , Epicardial Adipose Tissue
8.
Ann Endocrinol (Paris) ; 85(3): 214-219, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38871517

ABSTRACT

Osteoarthritis (OA) is the most common musculoskeletal disease, without any curative treatment. Obesity being the main modifiable risk factor for OA, much attention focused on the role of adipose tissues (AT). In addition to the involvement of visceral and subcutaneous AT via systemic ways, many arguments also highlight the involvement of local AT, present in joint tissues. Local AT include intra-articular AT (IAAT), which border the synovium, and bone marrow AT (BMAT) localized within marrow cavities in the bones. This review describes the known features and involvement of IAAT and BMAT in joint homeostasis and OA. Recent findings evidence that alteration in magnetic resonance imaging signal intensity of infrapatellar fat pad can be predictive of the development and progression of knee OA. IAAT and synovium are partners of the same functional unit; IAAT playing an early and pivotal role in synovial inflammation and fibrosis and OA pain. BMAT, whose functions have only recently begun to be studied, is in close functional interaction with its microenvironment. The volume and molecular profile of BMAT change according to the pathophysiological context, enabling fine regulation of haematopoiesis and bone metabolism. Although its role in OA has not yet been studied, the localization of BMAT, its functions and the importance of the bone remodelling processes that occur in OA argue in favour of a role for BMAT in OA.


Subject(s)
Adipose Tissue , Osteoarthritis , Synovial Membrane , Humans , Adipose Tissue/pathology , Adipose Tissue/physiopathology , Osteoarthritis/pathology , Osteoarthritis/physiopathology , Synovial Membrane/pathology , Joints/pathology , Osteoarthritis, Knee/pathology , Osteoarthritis, Knee/etiology , Osteoarthritis, Knee/physiopathology , Obesity/complications , Obesity/physiopathology , Bone Marrow/pathology , Magnetic Resonance Imaging , Animals
9.
High Blood Press Cardiovasc Prev ; 31(4): 389-399, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38874885

ABSTRACT

INTRODUCTION: Central obesity (CO), characterized by an increased waist circumference increases the risk of cardiovascular disease (CVD) and morbidity, yet the underlying mechanisms are not fully understood. CO is often associated with general obesity, hypertension, and abnormal glucose tolerance, confounding the independent contribution of CO to CVD. AIM: We investigated the relationship of CO (without associated disorders) with left ventricular (LV) characteristics and intrathoracic adipose tissue (IAT) by cardiac magnetic resonance. METHODS: LV characteristics, epicardial (EAT), and mediastinal adipose tissue (MAT) were measured from 29 normoglycemic, normotensive males with CO but without general obesity (waist circumference >100 cm, body mass index (BMI) <30 kg/m2) and 18 non-obese male controls. RESULTS: LV maximal wall thickness (LVMWT) and IAT but not LV mass or volumes were increased in CO subjects compared to controls (LVMWT, 12.3±1.2 vs. 10.7±1.5 mm, p < 0.001; EAT, 5.5±3.0 vs. 2.2±2.0 cm2, p = 0.001; MAT, 31.0±12.8 vs. 15.4±10.7 cm2, p < 0.001). The LVMWT was ≥12 mm in 69% of subjects with CO and 22% of controls (p = 0.002). In CO suspects, EAT correlated inversely with LV end-diastolic volume index (r = - 0.403, p = 0.037) and LV stroke volume (SV) (r = - 0.425, p = 0.027). MAT correlated inversely with SV (r = - 0.427, p=0.026) and positively with LVMWT (r = 0.399, p = 0.035). Among CO subjects, the waist-to-hip ratio (WHR) was an independent predictor of LVMWT (B = 22.4, ß = 0.617, p < 0.001). The optimal cut-off with Youden's index for LV hypertrophy was identified at WHR 0.98 (sensitivity 85%, specificity 89%). CONCLUSIONS: CO independent of BMI is associated with LV hypertrophy and intrathoracic adipose tissue contributing to cardiovascular burden.


Subject(s)
Adiposity , Hypertrophy, Left Ventricular , Obesity, Abdominal , Predictive Value of Tests , Ventricular Function, Left , Ventricular Remodeling , Humans , Male , Obesity, Abdominal/physiopathology , Obesity, Abdominal/diagnostic imaging , Obesity, Abdominal/complications , Case-Control Studies , Adult , Middle Aged , Hypertrophy, Left Ventricular/diagnostic imaging , Hypertrophy, Left Ventricular/physiopathology , Hypertrophy, Left Ventricular/etiology , Magnetic Resonance Imaging , Waist Circumference , Magnetic Resonance Imaging, Cine , Adipose Tissue/diagnostic imaging , Adipose Tissue/physiopathology , Body Mass Index , Cross-Sectional Studies
10.
Int J Cardiovasc Imaging ; 40(7): 1455-1463, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38761288

ABSTRACT

To investigate the correlation between quantitative plaque parameters, the perivascular fat attenuation index, and myocardial ischaemia caused by haemodynamic impairment. Patients with stable angina who had invasive flow reserve fraction (FFR) assessment and coronary artery computed tomography (CT) angiography were retrospectively enrolled. A total of 138 patients were included in this study, which were categorized into the FFR < 0.75 group (n = 43), 0.75 ≤ FFR ≤ 0.8 group (n = 37), and FFR > 0.8 group (n = 58), depending on the range of FFR values. The perivascular FAI and CTA-derived parameters, including plaque length (PL), total plaque volume (TPV), minimum lumen area (MLA), and narrowest degree (ND), were recorded for the lesions. An FFR < 0.75 was defined as myocardial-specific ischaemia. The relationships between myocardial ischaemia and parameters such as the PL, TPV, MLA, ND, and FAI were analysed using a logistic regression model and receiver operating characteristic (ROC) curves to compare the diagnostic accuracy of various indicators for myocardial ischaemia. The PL, TPV, ND, and FAI were greater in the FFR < 0.75 group than in the grey area group and the FFR > 0.80 group (all p < 0.05). The MLA in the FFR < 0.75 group was lower than that in the grey area group and the FFR > 0.80 group (both P < 0.05). There were no significant differences in the PL, TPV, or ND between the grey area and the FFR > 0.80 group, but there was a significant difference in the FAI. The coronary artery lesions with FFRs ≤ 0.75 had the greatest FAI values. Multivariate analysis revealed that the perivascular FAI and PL density are significant predictors of myocardial ischaemia. The FAI has some predictive value for myocardial ischaemia (AUC = 0.781). After building a combination model using the FAI and plaque length, the predictive power increased (AUC, 0.781 vs. 0.918), and the change was statistically significant (P < 0.001). The combined model of PL + FAI demonstrated great diagnostic efficacy in identifying myocardial ischaemia caused by haemodynamic impairment; the lower the FFR was, the greater the FAI. Thus, the PL + FAI could be a combined measure to securely rule out myocardial ischaemia.


Subject(s)
Computed Tomography Angiography , Coronary Angiography , Coronary Artery Disease , Coronary Vessels , Fractional Flow Reserve, Myocardial , Hemodynamics , Plaque, Atherosclerotic , Predictive Value of Tests , Humans , Male , Female , Retrospective Studies , Middle Aged , Aged , Coronary Artery Disease/physiopathology , Coronary Artery Disease/diagnostic imaging , Coronary Artery Disease/complications , Coronary Vessels/physiopathology , Coronary Vessels/diagnostic imaging , ROC Curve , Angina, Stable/physiopathology , Angina, Stable/diagnostic imaging , Adipose Tissue/diagnostic imaging , Adipose Tissue/physiopathology , Area Under Curve , Reproducibility of Results , Logistic Models , Severity of Illness Index , Multidetector Computed Tomography , Multivariate Analysis , Cardiac Catheterization , Prognosis , Myocardial Ischemia/physiopathology , Myocardial Ischemia/diagnostic imaging , Myocardial Ischemia/complications , Myocardial Ischemia/etiology
11.
Physiol Rep ; 12(9): e16042, 2024 May.
Article in English | MEDLINE | ID: mdl-38705872

ABSTRACT

Myosteatosis, or the infiltration of fatty deposits into skeletal muscle, occurs with advancing age and contributes to the health and functional decline of older adults. Myosteatosis and its inflammatory milieu play a larger role in adipose tissue dysfunction, muscle tissue dysfunction, and increased passive muscle stiffness. Combined with the age-related decline of sex hormones and development of anabolic resistance, myosteatosis also contributes to insulin resistance, impaired muscle mechanics, loss of force production from the muscle, and increased risk of chronic disease. Due to its highly inflammatory secretome and the downstream negative effects on muscle metabolism and mechanics, myosteatosis has become an area of interest for aging researchers and clinicians. Thus far, myosteatosis treatments have had limited success, as many lack the potency to completely rescue the metabolic and physical consequences of myosteatosis. Future research is encouraged for the development of reliable assessment methods for myosteatosis, as well as the continued exploration of pharmacological, nutritional, and exercise-related interventions that may lead to the success in attenuating myosteatosis and its clinical consequences within the aging population.


Subject(s)
Aging , Muscle, Skeletal , Humans , Muscle, Skeletal/metabolism , Muscle, Skeletal/physiopathology , Aging/physiology , Aged , Adipose Tissue/metabolism , Adipose Tissue/physiopathology
12.
BMC Cardiovasc Disord ; 24(1): 253, 2024 May 16.
Article in English | MEDLINE | ID: mdl-38750455

ABSTRACT

BACKGROUND: Primary coronary slow flow (CSF) is defined as delayed opacification of the distal epicardial vasculature during coronary angiography in the absence of relevant coronary artery stenoses. Microvascular disease is thought to be the underlying cause of this pathology. Epicardial fat tissue (EFT) is an active endocrine organ directly surrounding the coronary arteries that provides pro-inflammatory factors to the adjacent tissue by paracrine and vasocrine mechanisms. The aim of the present study was to investigate a potential association between EFT and primary CSF and whether EFT can predict the presence of primary CSF. METHODS: Between 2016 and 2017, n = 88 patients with high-grade aortic stenosis who were planned for transcatheter aortic valve implantation (TAVI) were included in this retrospective study. EFT volume was measured by pre-TAVI computed tomography (CT) using dedicated software. The presence of primary CSF was defined based on the TIMI frame count from the pre-TAVI coronary angiograms. RESULTS: Thirty-nine of 88 TAVI patients had CSF (44.3%). EFT volume was markedly higher in patients with CSF (142 ml [IQR 107-180] vs. 113 ml [IQR 89-147]; p = 0.009) and was strongly associated with the presence of CSF (OR 1.012 [95%CI 1.002-1.021]; p = 0.014). After adjustment, EFT volume was still an independent predictor of CSF (OR 1.016 [95%CI 1.004-1.026]; p = 0.009). CONCLUSION: Primary CSF was independently associated with increased EFT volume. Further studies are needed to validate this finding and elucidate whether a causal relationship exists.


Subject(s)
Adipose Tissue , Aortic Valve Stenosis , Coronary Angiography , Coronary Circulation , Pericardium , Predictive Value of Tests , Severity of Illness Index , Transcatheter Aortic Valve Replacement , Humans , Aortic Valve Stenosis/surgery , Aortic Valve Stenosis/physiopathology , Aortic Valve Stenosis/diagnostic imaging , Female , Male , Retrospective Studies , Pericardium/diagnostic imaging , Transcatheter Aortic Valve Replacement/adverse effects , Aged , Adipose Tissue/diagnostic imaging , Adipose Tissue/physiopathology , Aged, 80 and over , Risk Factors , Treatment Outcome , Aortic Valve/surgery , Aortic Valve/diagnostic imaging , Aortic Valve/physiopathology , Aortic Valve/pathology , Computed Tomography Angiography , Coronary Vessels/diagnostic imaging , Coronary Vessels/physiopathology , Epicardial Adipose Tissue
13.
Nutr Metab Cardiovasc Dis ; 34(7): 1741-1750, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38670920

ABSTRACT

BACKGROUND AND AIM: Long-term associations between the alternative healthy eating index (AHEI) score and two predictive indicators for CVD, pericardial adipose tissue (PAT) and coronary artery calcification (CAC) volume, are lacking. Our study aims to investigate the longitudinal associations of the AHEI score with measures of CAC and PAT in adults with and without type 1 diabetes (T1D). METHODS AND RESULTS: The prospective Coronary Artery Calcification in T1D (CACTI) study included 652 people with T1D and 764 people without diabetes (non-DM) (19-56 years old) and was conducted in 2000-2002, 2003-2004, and 2006-2007. At each visit, food frequency questionnaires were collected and PAT and CAC were measured using electron beam computed tomography. Two variables were used for CAC analyses: a continuous variable for the square-root tranformed volume (SRV) for each visit and a second variable identified CAC progression from baseline to visit 3. Mixed effect models and a logistic regression model were used to conduct statistical analyses. A one-point increase in the AHEI score was significantly associated with a -0.12 cm3 (95% CI: -0.17, -0.08; p-value<0.0001) decrease in PAT volume in combined analyses, a -0.16 cm3 (95% CI: -0.22, -0.09; p-value<0.0001) decrease in the non-DM group, a marginally significant -0.07 cm3 (95% CI: -0.14, 0.002; p-value = 0.0571) decrease in the T1D group, and was not associated with either CAC outcome. CONCLUSION: The AHEI score is inversely associated with PAT; the association revealed greater magnitude of PAT reduction in the non-DM group. The AHEI score did not associate with CAC progression.


Subject(s)
Adiposity , Coronary Artery Disease , Diabetes Mellitus, Type 1 , Diet, Healthy , Pericardium , Vascular Calcification , Humans , Middle Aged , Male , Female , Vascular Calcification/diagnostic imaging , Pericardium/diagnostic imaging , Adult , Coronary Artery Disease/diagnostic imaging , Prospective Studies , Longitudinal Studies , Diabetes Mellitus, Type 1/complications , Diabetes Mellitus, Type 1/diagnosis , Diabetes Mellitus, Type 1/physiopathology , Young Adult , Time Factors , United States/epidemiology , Risk Assessment , Adipose Tissue/diagnostic imaging , Adipose Tissue/physiopathology , Risk Factors , Protective Factors , Prognosis
14.
Curr Hypertens Rep ; 26(9): 381-388, 2024 Sep.
Article in English | MEDLINE | ID: mdl-38642285

ABSTRACT

PURPOSE OF THE REVIEW: Preserved ejection fraction heart failure and obesity frequently coexist. Whether obesity plays a consistent role in the pathogenesis of preserved ejection fraction heart failure is unclear. Accumulation of visceral adiposity underlies the pathogenic aftermaths of obesity. However, visceral adiposity imaging is assessed by computed tomography or magnetic resonance and thus not routinely available. In contrast, epicardial adiposity thickness is assessed by echocardiography and thus routinely available. We review the rationale for assessing epicardial adiposity thickness in patients with preserved ejection fraction heart failure and elevated body mass index. RECENT FINDINGS: Body mass index correlates poorly with visceral, and epicardial adiposity. Visceral and epicardial adiposity enlarges as preserved ejection fraction heart failure progresses. Epicardial adiposity may hasten the progression of coronary artery disease and impairs left ventricular sub-endocardial perfusion and diastolic function. Epicardial adiposity thickness may help monitor the therapeutic response in patients with preserved ejection failure heart failure and elevated body mass index.


Subject(s)
Adipose Tissue , Body Mass Index , Heart Failure , Obesity , Pericardium , Stroke Volume , Humans , Heart Failure/physiopathology , Pericardium/physiopathology , Pericardium/diagnostic imaging , Pericardium/pathology , Stroke Volume/physiology , Adipose Tissue/physiopathology , Adipose Tissue/pathology , Adipose Tissue/diagnostic imaging , Obesity/physiopathology , Obesity/complications , Adiposity , Echocardiography , Epicardial Adipose Tissue
15.
Intern Emerg Med ; 19(4): 1015-1024, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38578429

ABSTRACT

Epicardial adipose tissue is a novel cardiometabolic risk factor and indicator of subclinical atherosclerosis. We aimed to evaluate the epicardial adipose tissue thickness in rheumatoid arthritis (RA) patients and its association with disease activity scores. A total of 81 rheumatoid arthritis patients and 70 age- and sex-matched healthy individuals were recruited for this cross-sectional study. Epicardial adipose tissue thickness (EATT) was measured by transthoracic two-dimensional echocardiography. Tender and swollen joint counts were recorded at the time of inclusion. The laboratory tests included erythrocyte sedimentation rate (ESR), C-reactive protein (CRP), rheumatoid factor, anti-citrullinated protein antibodies, and serum lipid levels. Disease activity was calculated based on Disease Activity Scores for 28 joints (DAS-28) ESR and CRP, the Simple Disease Activity Index (SDAI), and the Clinical Disease Activity Index (CDAI). Epicardial adipose tissue thickness was significantly higher in the RA patients compared to the healthy controls (p < 0.001). We found statistically significant correlations of EATT with all disease activity indices (p < 0.001) and CRP (p = 0.002). According to a cut-off value of 6.4 mm determined for epicardial adipose tissue thickness, the RA patients with thickness ≥ 6.4 mm had higher disease activity scores and CRP levels. In the multivariable regression analysis, only SDAI score was found as an independent risk factor for increased EATT (OR, (95%CI), 13.70 (3.88-48.43), p < 0.001). Epicardial adipose tissue thickness measurement by echocardiography is a reliable method for assessing subclinical atherosclerosis in rheumatoid arthritis patients, and a higher disease activity score is an independent risk factor for coronary artery disease.


Subject(s)
Adipose Tissue , Arthritis, Rheumatoid , Atherosclerosis , Echocardiography , Pericardium , Humans , Arthritis, Rheumatoid/complications , Arthritis, Rheumatoid/physiopathology , Female , Male , Adipose Tissue/diagnostic imaging , Adipose Tissue/physiopathology , Middle Aged , Cross-Sectional Studies , Pericardium/diagnostic imaging , Atherosclerosis/physiopathology , Echocardiography/methods , Adult , C-Reactive Protein/analysis , Case-Control Studies , Risk Factors , Aged , Epicardial Adipose Tissue
16.
ESC Heart Fail ; 11(4): 2013-2022, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38480481

ABSTRACT

AIMS: This study aimed to identify the impact of increased epicardial adipose tissue (EAT) and its regional distribution on cardiac function in patients with diastolic dysfunction. METHODS AND RESULTS: Sixty-eight patients with exertional dyspnoea (New York Heart Association ≥II), preserved ejection fraction (≥50%), and diastolic dysfunction (E/e' ≥ 8) underwent rest and stress right heart catheterization, transthoracic echocardiography, and cardiovascular magnetic resonance (CMR). EAT volumes were depicted from CMR short-axis stacks. First, the impact of increased EAT above the median was investigated. Second, the association of ventricular and atrial EAT with myocardial deformation at rest and during exercise stress was analysed in a multivariable regression analysis. Patients with high EAT had higher HFA-PEFF and H2FPEFF scores as well as N-terminal prohormone of brain natriuretic peptide levels (all P < 0.048). They were diagnosed with manifest heart failure with preserved ejection fraction (HFpEF) more frequently (low EAT: 37% vs. high EAT: 64%; P = 0.029) and had signs of adverse remodelling indicated by higher T1 times (P < 0.001). No differences in biventricular volumetry and left ventricular mass (all P > 0.074) were observed. Patients with high EAT had impaired atrial strain at rest and during exercise stress, and impaired ventricular strain during exercise stress. Regionally increased EAT was independently associated with functional impairment of the adjacent chambers. CONCLUSIONS: Patients with diastolic dysfunction and increased EAT show more pronounced signs of diastolic functional failure and adverse structural remodelling. Despite similar morphological characteristics, patients with high EAT show significant cardiac functional impairment, in particular in the atria. Our results indicate that regionally increased EAT directly induces atrial functional failure, which represents a distinct pathophysiological feature in HFpEF.


Subject(s)
Adipose Tissue , Diastole , Magnetic Resonance Imaging, Cine , Pericardium , Stroke Volume , Humans , Male , Female , Pericardium/diagnostic imaging , Pericardium/physiopathology , Adipose Tissue/physiopathology , Adipose Tissue/diagnostic imaging , Magnetic Resonance Imaging, Cine/methods , Stroke Volume/physiology , Middle Aged , Aged , Ventricular Function, Left/physiology , Echocardiography , Heart Failure, Diastolic/physiopathology , Heart Failure, Diastolic/diagnosis , Heart Ventricles/physiopathology , Heart Ventricles/diagnostic imaging , Follow-Up Studies , Cardiac Catheterization , Epicardial Adipose Tissue
17.
J Cardiovasc Electrophysiol ; 35(6): 1121-1128, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38536760

ABSTRACT

INTRODUCTION: Postoperative atrial fibrillation (POAF) is common following open heart surgery, and is associated with significant morbidity. Medications used for ventricular rate control of POAF may not be effective in controlling rapid ventricular rates during the postoperative period because of increased sympathetic tone. The purpose of this study was to develop nonpharmacologic rate control of POAF by atrioventricular node (AVN) fat pad stimulation using clinically available temporary pacing wires in the canine sterile pericarditis model. METHODS: We studied 10 sterile pericarditis dogs in the closed-chest state on postoperative days 1-3. The AVN fat pad stimulation (amplitude 2-15 mA; frequency 20 Hz; pulse width 0.03-0.2 ms) was performed during sustained POAF (>5 min). We measured ventricular rate and inefficient ventricular contractions during sustained POAF and compared it with and without AVN fat pad stimulation. Also, the parameters of AVN fat pad stimulation to achieve a rate control of POAF were measured over the postoperative days. RESULTS: Eleven episodes of sustained POAF were induced in 5/10 sterile pericarditis dogs in the closed-chest state on postoperative days 1-2. During POAF, the AVN fat pad stimulation decreased the ventricular rate from 178 ± 52 bpm to 100 ± 8 bpm in nine episodes. Nonpharmacologic rate control therapy successfully controlled the ventricular rate and eliminated inefficient ventricular contractions during POAF for the duration of the AVN fat pad stimulation. The AVN fat pad stimulation output remained relatively stable over the postoperative days. CONCLUSION: During sustained POAF, nonpharmacologic rate control by AVN fat pad stimulation effectively and safely controlled rapid ventricular rates throughout the postoperative period.


Subject(s)
Atrial Fibrillation , Disease Models, Animal , Heart Rate , Pericarditis , Animals , Dogs , Atrial Fibrillation/physiopathology , Atrial Fibrillation/diagnosis , Pericarditis/physiopathology , Pericarditis/diagnosis , Cardiac Pacing, Artificial , Atrioventricular Node/physiopathology , Atrioventricular Node/surgery , Male , Time Factors , Cardiac Surgical Procedures/adverse effects , Adipose Tissue/physiopathology , Postoperative Complications/etiology
18.
ESC Heart Fail ; 11(3): 1698-1706, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38438270

ABSTRACT

AIMS: Obesity and epicardial adiposity play a role in the pathophysiology of heart failure with preserved ejection fraction (HFpEF), and both are associated with increased filling pressures and reduced exercise capacity. The haemodynamic basis for these observations remains inaccurately defined. We hypothesize that an abundance of epicardial adipose tissue (EAT) within the pericardial sac is associated with haemodynamic signs of pericardial constraint. METHODS AND RESULTS: HFpEF patients who underwent invasive heart catheterization with simultaneous echocardiography were included. Right atrial pressure (RAP), right ventricular end-diastolic pressure, and pulmonary capillary wedge pressure (PCWP) were invasively measured. The presence of a square root sign on the right ventricular pressure waveform and the RAP/PCWP ratio (surrogate parameters for pericardial constraint) were investigated. EAT thickness alongside the right ventricle was measured on echocardiography. Sixty-four patients were studied, with a mean age of 73 ± 10 years, 64% women, and a mean body mass index (BMI) of 28.6 ± 5.4 kg/m2. In total, 47 patients (73%) had a square root sign. The presence of a square root sign was associated with higher BMI (29.3 vs. 26.7 kg/m2, P = 0.02), higher EAT (4.0 vs. 3.4 mm, P = 0.03), and higher RAP (9 vs. 6 mmHg, P = 0.04). Women had more EAT than men (4.1 vs. 3.5 mm, P = 0.04), despite a comparable BMI. Women with a square root sign had significantly higher EAT (4.3 vs. 3.3 mm, P = 0.02), a higher mean RAP (9 vs. 5 mmHg, P = 0.02), and a higher RAP/PCWP ratio (0.52 vs. 0.26, P = 0.002). In men, such associations were not seen, although there was no significant interaction between men and women (P > 0.05 for all analyses). CONCLUSIONS: Obesity and epicardial adiposity are associated with haemodynamic signs of pericardial constraint in patients with HFpEF. The pathophysiological and therapeutic implications of this finding need further study.


Subject(s)
Adipose Tissue , Echocardiography , Heart Failure , Pericardium , Stroke Volume , Humans , Female , Heart Failure/physiopathology , Male , Stroke Volume/physiology , Pericardium/physiopathology , Pericardium/diagnostic imaging , Aged , Adipose Tissue/physiopathology , Adipose Tissue/diagnostic imaging , Cardiac Catheterization/methods , Heart Ventricles/physiopathology , Heart Ventricles/diagnostic imaging , Epicardial Adipose Tissue
19.
J Cardiovasc Transl Res ; 17(4): 851-858, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38409474

ABSTRACT

Following the placement of endovascular implants, perivascular adipose tissue (PVAT) becomes an early sensor of vascular injury to which it responds by undergoing phenotypic changes characterized by reduction in the secretion of adipocyte-derived relaxing factors and a shift to a proinflammatory and pro-contractile state. Thus, activated PVAT loses its anti-inflammatory function, secretes proinflammatory cytokines and chemokines, and generates reactive oxygen species, which are accompanied by differentiation of fibroblasts into myofibroblasts and proliferation of smooth muscle cells. These subsequently migrate into the intima, leading to intimal growth. In addition, periadventitial vasa vasorum undergoes neovascularization and functions as a portal for extravasation of inflammatory infiltrates and mobilization of PVAT resident stem/progenitor cells into the intima. This review focuses on the response of PVAT to endovascular intervention-induced injury and discusses potential therapeutic targets to suppress the PVAT-initiated pathways that mediate the formation of neointima.


Subject(s)
Adipose Tissue , Endovascular Procedures , Neointima , Signal Transduction , Humans , Animals , Adipose Tissue/metabolism , Adipose Tissue/pathology , Adipose Tissue/physiopathology , Endovascular Procedures/adverse effects , Endovascular Procedures/instrumentation , Vascular System Injuries/pathology , Vascular System Injuries/metabolism , Vascular System Injuries/physiopathology , Vascular System Injuries/etiology , Inflammation Mediators/metabolism , Cell Proliferation , Phenotype
20.
Sci Rep ; 13(1): 2831, 2023 02 17.
Article in English | MEDLINE | ID: mdl-36805000

ABSTRACT

In this study, we explored the relationship between inflammatory adipokine levels and coronary artery disease (CAD). We collected subcutaneous adipose tissues(SAT), pericardial adipose tissues(PAT), and epicardial adipose tissues (EAT) and serum samples from 26 inpatients with CAD undergone coronary artery bypass grafting and 20 control inpatients without CAD. Serum inflammatory adipokines were measured by ELISA. Quantitative real-time PCR and western blot were used to measure gene and protein expression. Adipocyte morphology was assessed by H&E staining. Immunohistochemistry and immunofluorescence were used to measure endothelial and inflammatory markers. Serum pro- and anti-inflammatory adipokine levels were higher and lower, respectively, in the CAD group than those in the control group (P < 0.05). In CAD, the pro-inflammatory adipokine levels via ELISA in EAT and PAT were elevated. Pro-inflammatory adipokine mRNA expression was increased, while anti-inflammatory adipokine mRNA expression decreased, in CAD relative to NCAD in EAT and PAT rather than SAT. In EAT, adipocyte area and macrophage-specific staining were lower, while lymphatic vessel marker expression was higher in CAD. Additionally, the endothelial marker expression in EAT was higher than PAT in CAD. The three tissue types had different blood vessel amounts in CAD. The regulation and imbalance expression of the novel biomarkers, including inflammatory adipokine, macrophage infiltration, angiogenesis, and lymphangiogenesis in EAT and PAT, may be related to the pathogenesis of CAD. The serum levels of inflammatory adipokines may correlate to CAD, which requires large sample size studies to get further validation before clinic practice.


Subject(s)
Adipose Tissue , Coronary Artery Disease , Pericardium , Humans , Adipokines/blood , Adipokines/genetics , Adipokines/metabolism , Adipose Tissue/blood supply , Adipose Tissue/metabolism , Adipose Tissue/physiopathology , Biomarkers/blood , Biomarkers/metabolism , Coronary Artery Disease/blood , Coronary Artery Disease/genetics , Coronary Artery Disease/metabolism , Coronary Artery Disease/physiopathology , Lymphangiogenesis/physiology , Neovascularization, Pathologic/blood , Neovascularization, Pathologic/metabolism , Neovascularization, Pathologic/physiopathology , Pericardium/metabolism , Pericardium/physiopathology , RNA, Messenger/genetics , RNA, Messenger/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL