1
|
Sawali MA, Zahid MA, Abdelsalam SS, Al-Zoubi RM, Shkoor M, Agouni A. The role of PTP1B in cardiometabolic disorders and endothelial dysfunction. J Drug Target 2025:1-16. [PMID: 39996501 DOI: 10.1080/1061186x.2025.2473024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2024] [Revised: 01/15/2025] [Accepted: 02/23/2025] [Indexed: 02/26/2025]
Abstract
Cardiovascular diseases (CVD) are a global health concern that accounts for a large share of annual mortality. Endothelial dysfunction is the main underlying factor that eventually leads to cardiovascular events. Recent studies have underscored the critical function of Protein Tyrosine Phosphatase 1B (PTP1B) in the onset of endothelial dysfunction, chiefly through its involvement in metabolic diseases such as diabetes, obesity, and leptin resistance. PTP1B attenuates insulin and leptin signalling by dephosphorylating their respective receptors at key tyrosine residues, resulting in resistance-both of which are significant mechanisms underpinning the development of endothelial dysfunction. PTP1B also contributes to the disruption of the endoplasmic reticulum, causing endoplasmic reticulum stress, another molecular driver of endothelial dysfunction. Efforts to inhibit PTP1B activity hold the promise of advancing the prevention and management of CVD and metabolic disorders, as these conditions share common risk factors and underlying cellular mechanisms. Numerous small molecules have been reported as PTP1B inhibitors; however, their progression to advanced clinical trials has been hindered by major challenges such as low selectivity and undesirable side effects. This review provides an in-depth analysis of PTP1B's involvement in metabolic diseases and its interaction with CVD and examines the strategies and challenges related to inhibiting this enzyme.
Collapse
Affiliation(s)
- Mona A Sawali
- Department of Pharmaceutical Sciences, College of Pharmacy, QU Health, Qatar University, Doha, Qatar
| | - Muhammad Ammar Zahid
- Department of Pharmaceutical Sciences, College of Pharmacy, QU Health, Qatar University, Doha, Qatar
| | - Shahenda Salah Abdelsalam
- Department of Pharmaceutical Sciences, College of Pharmacy, QU Health, Qatar University, Doha, Qatar
| | - Raed M Al-Zoubi
- Surgical Research Section, Department of Surgery, Hamad Medical Corporation, Doha, Qatar
- Department of Biomedical Sciences, College of Health Sciences, QU Health, Qatar University, Doha, Qatar
- Department of Chemistry, Jordan University of Science and Technology, Irbid, Jordan
| | - Mohanad Shkoor
- Department of Chemistry, College of Arts and Science, Qatar University, Doha, Qatar
| | - Abdelali Agouni
- Department of Pharmaceutical Sciences, College of Pharmacy, QU Health, Qatar University, Doha, Qatar
| |
Collapse
|
2
|
Sun J, Chen J, Cicchella A. Correlation of Adiponectin and Leptin with Anthropometrics and Behavioral and Physical Performance in Overweight and Obese Chinese College Students. BIOLOGY 2024; 13:567. [PMID: 39194505 DOI: 10.3390/biology13080567] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/30/2024] [Revised: 07/17/2024] [Accepted: 07/20/2024] [Indexed: 08/29/2024]
Abstract
The aim of this study is to assess the relationship of leptin (LEP) and adiponectin (ADPN) with other circulating fat markers, physical capacity, behaviors, and anthropometric indices in a population of overweight and obese Chinese university students. LEP and ADPN levels, as well as behavioral, anthropometric, biochemical, and performance characteristics, were measured. METHOD A total of 17 anthropometric parameters, 8 questionnaires (investigating quality of life, sleep, eating, perceived functioning, stress, and depression), 9 biochemical parameters, and 12 functional parameters were investigated. RESULTS In contrast to previous studies, our work found an unusually strong relationship between LEP and ADPN (r = 0.961, p = 0.000) that can be related to ethnicity. We also found that LEP and ADPN were associated with stress and bodily pain. A total of 12 anthropometric measures were also associated with LEP/ADNP levels. Moreover, LEP and ADPN were found to be related to lower limb, hand, and abdominal strength; blood pressure; and basic metabolism. However, we did not find associations with sleep; eating habits; or cardiovascular fitness, which was measured in the form of resting heart rate and VO2max. CONCLUSION This study reveals new relationships of LEP and ADPN with selected anthropometric and behavioral parameters in obese Chinese college students.
Collapse
Affiliation(s)
- Jingyu Sun
- Sports and Health Research Center, Department of Physical Education, Tongji University, Shanghai 200092, China
| | - Jiajia Chen
- Sports and Health Research Center, Department of Physical Education, Tongji University, Shanghai 200092, China
| | - Antonio Cicchella
- International College of Football, Tongji University, Shanghai 200092, China
- Department for Quality of Life Studies, Bologna University, 40127 Bologna, Italy
| |
Collapse
|
3
|
Naik N, Patel M, Sen R. Developmental Impacts of Epigenetics and Metabolism in COVID-19. J Dev Biol 2024; 12:9. [PMID: 38390960 PMCID: PMC10885083 DOI: 10.3390/jdb12010009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2023] [Revised: 02/04/2024] [Accepted: 02/06/2024] [Indexed: 02/24/2024] Open
Abstract
Developmental biology is intricately regulated by epigenetics and metabolism but the mechanisms are not completely understood. The situation becomes even more complicated during diseases where all three phenomena are dysregulated. A salient example is COVID-19, where the death toll exceeded 6.96 million in 4 years, while the virus continues to mutate into different variants and infect people. Early evidence during the pandemic showed that the host's immune and inflammatory responses to COVID-19 (like the cytokine storm) impacted the host's metabolism, causing damage to the host's organs and overall physiology. The involvement of angiotensin-converting enzyme 2 (ACE2), the pivotal host receptor for the SARS-CoV-2 virus, was identified and linked to epigenetic abnormalities along with other contributing factors. Recently, studies have revealed stronger connections between epigenetics and metabolism in COVID-19 that impact development and accelerate aging. Patients manifest systemic toxicity, immune dysfunction and multi-organ failure. Single-cell multiomics and other state-of-the-art high-throughput studies are only just beginning to demonstrate the extent of dysregulation and damage. As epigenetics and metabolism directly impact development, there is a crucial need for research implementing cutting-edge technology, next-generation sequencing, bioinformatics analysis, the identification of biomarkers and clinical trials to help with prevention and therapeutic interventions against similar threats in the future.
Collapse
Affiliation(s)
- Noopur Naik
- Department of Molecular, Cellular & Developmental Biology, University of Colorado Boulder, Boulder, CO 80309, USA
| | - Mansi Patel
- Institute of Genomics and Integrative Biology, Delhi 110007, India
| | - Rwik Sen
- Active Motif, Inc., Carlsbad, CA 92008, USA
| |
Collapse
|
4
|
Wang W, Zhang W, Li L, Hu D, Liu S, Cui L, Liu J, Xu J, Guo X, Deng F. Obesity-related cardiometabolic indicators modify the associations of personal noise exposure with heart rate variability: A further investigation on the Study among Obese and Normal-weight Adults (SONA). ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2023; 336:122446. [PMID: 37625771 DOI: 10.1016/j.envpol.2023.122446] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/03/2023] [Revised: 08/17/2023] [Accepted: 08/22/2023] [Indexed: 08/27/2023]
Abstract
Elucidating the associations between environmental noise and heart rate variability (HRV) would be beneficial for the prevention and control of detrimental cardiovascular changes. Obese people have been found to manifest heightened susceptibility to the adverse effects of noise on HRV. However, the underlying mechanisms remain unclear. Based on 53 normal-weight and 44 obese young adults aged 18-26 years in Beijing, China, this study aimed to investigate the role of obesity-related cardiometabolic indicators for associations between short-term environmental noise exposure and HRV in the real-world context. The participants underwent personal noise exposure and ambulatory electrocardiogram monitoring using portable devices at 5-min intervals for 24 continuous hours. Obesity-related blood pressure, glucose and lipid metabolism, and inflammatory indicators were subsequently examined. Generalized mixed-effect models were used to estimate the associations between noise exposure and HRV parameters. The C-peptide, homeostasis model assessment of insulin resistance (HOMA-IR), and leptin levels were higher in obese participants compared to normal-weight participants. We observed amplified associations between short-term noise exposure and decreases in HRV among participants with higher C-peptide, HOMA-IR, and leptin levels. For instance, a 1 dB(A) increment in 3 h-average noise exposure level preceding each measurement was associated with changes of -0.20% (95%CI: -0.45%, 0.04%) and -1.35% (95%CI: -1.85%, -0.86%) in standard deviation of all normal to normal intervals (SDNN) among participants with lower and higher C-peptide levels, respectively (P for interaction <0.05). Meanwhile, co-existing fine particulate matter (PM2.5) could amplify the associations between noise and HRV among obese participants and participants with higher C-peptide, HOMA-IR, and leptin levels. The more apparent associations of short-term exposure to environmental noise with HRV and the effect modification by PM2.5 may be partially explained by the higher C-peptide, HOMA-IR, and leptin levels of obese people.
Collapse
Affiliation(s)
- Wanzhou Wang
- Department of Occupational and Environmental Health Sciences, School of Public Health, Peking University, Beijing, 100191, China
| | - Wenlou Zhang
- Department of Occupational and Environmental Health Sciences, School of Public Health, Peking University, Beijing, 100191, China
| | - Luyi Li
- Department of Occupational and Environmental Health Sciences, School of Public Health, Peking University, Beijing, 100191, China
| | - Dayu Hu
- Department of Occupational and Environmental Health Sciences, School of Public Health, Peking University, Beijing, 100191, China
| | - Shan Liu
- Department of Occupational and Environmental Health Sciences, School of Public Health, Peking University, Beijing, 100191, China
| | - Liyan Cui
- Department of Laboratory Medicine, Peking University Third Hospital, Beijing, 100191, China
| | - Junxiu Liu
- Department of Otolaryngology Head and Neck Surgery, Peking University Third Hospital, Beijing, 100191, China
| | - Junhui Xu
- Department of Occupational and Environmental Health Sciences, School of Public Health, Peking University, Beijing, 100191, China
| | - Xinbiao Guo
- Department of Occupational and Environmental Health Sciences, School of Public Health, Peking University, Beijing, 100191, China
| | - Furong Deng
- Department of Occupational and Environmental Health Sciences, School of Public Health, Peking University, Beijing, 100191, China; Center for Environment and Health, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, 100871, China.
| |
Collapse
|
5
|
Talati M, Brittain E, Agrawal V, Fortune N, Simon K, Shay S, Zeng X, Freeman ML, West J, Hemnes A. A potential adverse role for leptin and cardiac leptin receptor in the right ventricle in pulmonary arterial hypertension: effect of metformin is BMPR2 mutation-specific. Front Med (Lausanne) 2023; 10:1276422. [PMID: 37869164 PMCID: PMC10586504 DOI: 10.3389/fmed.2023.1276422] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2023] [Accepted: 09/15/2023] [Indexed: 10/24/2023] Open
Abstract
Introduction Pulmonary arterial hypertension is a fatal cardiopulmonary disease. Leptin, a neuroendocrine hormone released by adipose tissue, has a complex relationship with cardiovascular diseases, including PAH. Leptin is thought to be an important factor linking metabolic syndrome and cardiovascular disorders. Given the published association between metabolic syndrome and RV dysfunction in PAH, we sought to determine the association between leptin and RV dysfunction. We hypothesized that in PAH-RV, leptin influences metabolic changes via leptin receptors, which can be manipulated by metformin. Methods Plasma leptin was measured in PAH patients and healthy controls from a published trial of metformin in PAH. Leptin receptor localization was detected in RV from PAH patients, healthy controls, animal models of PH with RV dysfunction before and after metformin treatment, and cultured cardiomyocytes with two different BMPR2 mutants by performing immunohistochemical and cell fractionation studies. Functional studies were conducted in cultured cardiomyocytes to examine the role of leptin and metformin in lipid-driven mitochondrial respiration. Results In human studies, we found that plasma leptin levels were higher in PAH patients and moderately correlated with higher BMI, but not in healthy controls. Circulating leptin levels were reduced by metformin treatment, and these findings were confirmed in an animal model of RV dysfunction. Leptin receptor expression was increased in PAH-RV cardiomyocytes. In animal models of RV dysfunction and cultured cardiomyocytes with BMPR2 mutation, we found increased expression and membrane localization of the leptin receptor. In cultured cardiomyocytes with BMPR2 mutation, leptin moderately influences palmitate uptake, possibly via CD36, in a mutation-specific manner. Furthermore, in cultured cardiomyocytes, the Seahorse XFe96 Extracellular Flux Analyzer and gene expression data indicate that leptin may not directly influence lipid-driven mitochondrial respiration in BMPR2 mutant cardiomyocytes. However, metformin alone or when supplemented with leptin can improve lipid-driven mitochondrial respiration in BMPR2 mutant cardiomyocytes. The effect of metformin on lipid-driven mitochondrial respiration in cardiomyocytes is BMPR2 mutation-specific. Conclusion In PAH, increased circulating leptin can influence metabolic signaling in RV cardiomyocytes via the leptin receptor; in particular, it may alter lipid-dependent RV metabolism in combination with metformin in a mutation-specific manner and warrants further investigation.
Collapse
Affiliation(s)
- Megha Talati
- Division of Allergy, Pulmonary and Critical Care Medicine, Vanderbilt University Medical Center, Nashville, TN, United States
| | - Evan Brittain
- Division of Cardiovascular Medicine, Vanderbilt University Medical Center, Nashville, TN, United States
| | - Vineet Agrawal
- Division of Cardiovascular Medicine, Vanderbilt University Medical Center, Nashville, TN, United States
| | - Niki Fortune
- Division of Allergy, Pulmonary and Critical Care Medicine, Vanderbilt University Medical Center, Nashville, TN, United States
| | - Katie Simon
- Division of Allergy, Pulmonary and Critical Care Medicine, Vanderbilt University Medical Center, Nashville, TN, United States
| | - Sheila Shay
- Division of Allergy, Pulmonary and Critical Care Medicine, Vanderbilt University Medical Center, Nashville, TN, United States
| | - Xiaofang Zeng
- Department of Cardiology, Xiangya Hospital, Central South University, Changsha, China
| | - Michael L. Freeman
- Department of Radiation Oncology, Vanderbilt University Medical Center, Nashville, TN, United States
| | - James West
- Division of Allergy, Pulmonary and Critical Care Medicine, Vanderbilt University Medical Center, Nashville, TN, United States
| | - Anna Hemnes
- Division of Allergy, Pulmonary and Critical Care Medicine, Vanderbilt University Medical Center, Nashville, TN, United States
| |
Collapse
|
6
|
Vasamsetti SB, Natarajan N, Sadaf S, Florentin J, Dutta P. Regulation of cardiovascular health and disease by visceral adipose tissue-derived metabolic hormones. J Physiol 2023; 601:2099-2120. [PMID: 35661362 PMCID: PMC9722993 DOI: 10.1113/jp282728] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Accepted: 05/04/2022] [Indexed: 11/08/2022] Open
Abstract
Visceral adipose tissue (VAT) is a metabolic organ known to regulate fat mass, and glucose and nutrient homeostasis. VAT is an active endocrine gland that synthesizes and secretes numerous bioactive mediators called 'adipocytokines/adipokines' into systemic circulation. These adipocytokines act on organs of metabolic importance like the liver and skeletal muscle. Multiple preclinical and in vitro studies showed strong evidence of the roles of adipocytokines in the regulation of metabolic disorders like diabetes, obesity and insulin resistance. Adipocytokines, such as adiponectin and omentin, are anti-inflammatory and have been shown to prevent atherogenesis by increasing nitric oxide (NO) production by the endothelium, suppressing endothelium-derived inflammation and decreasing foam cell formation. By inhibiting differentiation of vascular smooth muscle cells (VSMC) into osteoblasts, adiponectin and omentin prevent vascular calcification. On the other hand, adipocytokines like leptin and resistin induce inflammation and endothelial dysfunction that leads to vasoconstriction. By promoting VSMC migration and proliferation, extracellular matrix degradation and inflammatory polarization of macrophages, leptin and resistin increase the risk of atherosclerotic plaque vulnerability and rupture. Additionally, the plasma concentrations of these adipocytokines alter in ageing, rendering older humans vulnerable to cardiovascular disease. The disturbances in the normal physiological concentrations of these adipocytokines secreted by VAT under pathological conditions impede the normal functions of various organs and affect cardiovascular health. These adipokines could be used for both diagnostic and therapeutic purposes in cardiovascular disease.
Collapse
Affiliation(s)
- Sathish Babu Vasamsetti
- Pittsburgh Heart, Lung, Blood, and Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, PA, USA 15213
- Pittsburgh VA Medical Center-University Drive, University Drive C, Pittsburgh, PA, USA
| | - Niranjana Natarajan
- Pittsburgh Heart, Lung, Blood, and Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, PA, USA 15213
| | - Samreen Sadaf
- Pittsburgh Heart, Lung, Blood, and Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, PA, USA 15213
- Pittsburgh VA Medical Center-University Drive, University Drive C, Pittsburgh, PA, USA
| | - Jonathan Florentin
- Pittsburgh Heart, Lung, Blood, and Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, PA, USA 15213
| | - Partha Dutta
- Pittsburgh Heart, Lung, Blood, and Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, PA, USA 15213
- Pittsburgh VA Medical Center-University Drive, University Drive C, Pittsburgh, PA, USA
- Department of Bioengineering, Swanson School of Engineering, University of Pittsburgh, Pittsburgh, PA, USA, 15213
- Division of Cardiology, Department of Medicine, University of Pittsburgh, Pittsburgh, PA, USA, 15213
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| |
Collapse
|
7
|
Smooth Muscle Cells from a Rat Model of Obesity and Hyperleptinemia Are Partially Resistant to Leptin-Induced Reactive Oxygen Species Generation. Antioxidants (Basel) 2023; 12:antiox12030728. [PMID: 36978976 PMCID: PMC10045401 DOI: 10.3390/antiox12030728] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Revised: 03/05/2023] [Accepted: 03/11/2023] [Indexed: 03/18/2023] Open
Abstract
The aim of this study was to evaluate the effect of leptin on reactive oxygen species’ (ROS) generation of smooth muscle cells (SMCs) from a rat model of obesity and hyperleptinemia. Obesity and hyperleptinemia were induced in rats by a sucrose-based diet for 24 weeks. ROS generation was detected by using dichloro-dihydrofluorescein (DCF), a fluorescent ROS probe in primary SMCs culture. An increase in plasma leptin and oxidative stress markers was observed in sucrose-fed (SF) rats. At baseline SMCs from SF rats showed a more than twofold increase in fluorescence intensity (FI) compared to that obtained in control (C) cells. When the C cells were treated with 20 ng leptin, the FI increased by about 200%, whereas the leptin-induced FI in the SF cells increased only by 60%. In addition, sucrose feeding increased the levels of p22phox and gp91phox, subunits of Nox as an O2•− source in SMCs. Treatment of cells with leptin significantly increased p22phox and gp91phox levels in C cells and did not affect SF cells. Regarding STAT3 phosphorylation and the content of PTP1B and SOCS3 as protein markers of leptin resistance, they were found to be significantly increased in SF cells. These results suggest that SF aortic SMCs are partially resistant to leptin-induced ROS generation.
Collapse
|
8
|
López-Acosta O, Ruiz-Ramírez A, Barrios-Maya MÁ, Alarcon-Aguilar J, Alarcon-Enos J, Céspedes Acuña CL, El-Hafidi M. Lipotoxicity, glucotoxicity and some strategies to protect vascular smooth muscle cell against proliferative phenotype in metabolic syndrome. Food Chem Toxicol 2023; 172:113546. [PMID: 36513245 DOI: 10.1016/j.fct.2022.113546] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2022] [Revised: 11/16/2022] [Accepted: 11/29/2022] [Indexed: 12/14/2022]
Abstract
Metabolic syndrome (MetS) is a risk factor for the development of cardiovascular disease (CVD) and atherosclerosis through a mechanism that involves vascular smooth muscle cell (VSMC) proliferation, lipotoxicity and glucotoxicity. Several molecules found to be increased in MetS, including free fatty acids, fatty acid binding protein 4, leptin, resistin, oxidized lipoprotein particles, and advanced glycation end products, influence VSMC proliferation. Most of these molecules act through their receptors on VSMCs by activating several signaling pathways associated with ROS generation in various cellular compartments. ROS from NADPH-oxidase and mitochondria have been found to promote VSMC proliferation and cell cycle progression. In addition, most of the natural or synthetic substances described in this review, including pharmaceuticals with hypoglycemic and hypolipidemic properties, attenuate VSMC proliferation by their simultaneous modulation of cell signaling and their scavenging property due to the presence of a phenolic ring in their structure. This review discusses recent data in the literature on the role that several MetS-related molecules and ROS play in the change from contractile to proliferative phenotype of VSMCs. Hence the importance of proposing an appropriate strategy to prevent uncontrolled VSMC proliferation using antioxidants, hypoglycemic and hypolipidemic agents.
Collapse
Affiliation(s)
- Ocarol López-Acosta
- Depto de Biomedicina Cardiovascular, Instituto Nacional de Cardiología Ignacio Chávez, Juan Badiano No 1, Colonia Sección XVI, Tlalpan, 14080, México D.F., Mexico
| | - Angélica Ruiz-Ramírez
- Depto de Biomedicina Cardiovascular, Instituto Nacional de Cardiología Ignacio Chávez, Juan Badiano No 1, Colonia Sección XVI, Tlalpan, 14080, México D.F., Mexico
| | - Miguel-Ángel Barrios-Maya
- Depto de Biomedicina Cardiovascular, Instituto Nacional de Cardiología Ignacio Chávez, Juan Badiano No 1, Colonia Sección XVI, Tlalpan, 14080, México D.F., Mexico
| | - Javier Alarcon-Aguilar
- Laboratorio de Farmacología, Depto. de Ciencias Biológicas y de la Salud, Universidad Autónoma Metropolitana Unidad Iztapalapa, Iztapalapa, Mexico
| | - Julio Alarcon-Enos
- Departamento de Ciencias Básicas, Facultad de Ciencias, Universidad del Bio Bio, Av. Andres Bello 720, Chillan, Chile
| | - Carlos L Céspedes Acuña
- Departamento de Ciencias Básicas, Facultad de Ciencias, Universidad del Bio Bio, Av. Andres Bello 720, Chillan, Chile.
| | - Mohammed El-Hafidi
- Depto de Biomedicina Cardiovascular, Instituto Nacional de Cardiología Ignacio Chávez, Juan Badiano No 1, Colonia Sección XVI, Tlalpan, 14080, México D.F., Mexico.
| |
Collapse
|
9
|
Shih YL, Shih YH, Huang TC, Shih CC, Chen JY. Association between sedentary time and plasma leptin levels in middle-aged and older adult population in Taiwan: A community-based, cross-sectional study. Front Cardiovasc Med 2023; 9:1057497. [PMID: 36698957 PMCID: PMC9868819 DOI: 10.3389/fcvm.2022.1057497] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Accepted: 12/13/2022] [Indexed: 01/12/2023] Open
Abstract
Background Association of sedentary behavior and plasma leptin levels is a popular topic in recent research. Aged populations often suffer from cardiometabolic diseases, and leptin is considered a novel marker for many cardiometabolic diseases. To further explore this topic, our research investigates the relationship between sedentary time and serum leptin levels in middle-aged and older populations. Methods A total of 396 middle-aged and older adult Taiwanese participants were included in this study. We recorded their self-reported sitting time as sedentary time. Participants were categorized into low leptin, medium leptin group, and high leptin groups according to the tertile of serum leptin level in the study. We also analyzed the anthropometric and cardiometabolic parameters between the three groups. Spearman's correlation coefficient was used to analyze the correlation between leptin level, sedentary time, and other cardiometabolic risk factors. The relationsip between leptin and sedentary time was also shown in a scatter plot. Multivariate linear regression was performed to determine the association between serum leptin levels and sedentary time after adjusting for age, sex, alcohol consumption, smoking, triglycerides, body mass index (BMI), fasting plasma glucose, systolic blood pressure, uric acid, creatinine, and alanine transaminase (ALT). Results In our study, data from a total of 396 participants were analyzed. The average age of participants was 64.75 (±8.75) years, and ~41.4% were male. A longer period of sedentary time was observed in the high leptin group. A positive correlation was found between serum leptin level and sedentary time in Spearman's correlation, in all BMI groups. Serum leptin levels were positively associated with sedentary time (B = 0.603, p = 0.016) in the multivariate linear regression after adjusting for age, sex, alcohol consumption, smoking, triglycerides, BMI, fasting plasma glucose, systolic blood pressure, uric acid, creatinine, and ALT. Conclusion Prolonged sedentary time can be an independent risk factor for high serum leptin levels, and high leptin levels can be a novel marker in future healthcare to screen the individual with prolonged sedentary time. Furthermore, based on our study, future research can further explore the relationship between leptin levels and health promotion, especially decreasing sedentary time in the middle-aged and elder population, which is vulnerable to cardiometabolic diseases.
Collapse
Affiliation(s)
- Yu-Lin Shih
- Department of Family Medicine, Chang Gung Memorial Hospital, Taoyuan, Taiwan
| | - Yun-Hsiang Shih
- School of Medicine, Medical University of Lublin, Lublin, Poland
| | - Tzu-Cheng Huang
- Department of Family Medicine, Chang Gung Memorial Hospital, Taoyuan, Taiwan
| | - Chin-Chuan Shih
- General Administrative Department, United Safety Medical Group, New Taipei City, Taiwan
| | - Jau-Yuan Chen
- Department of Family Medicine, Chang Gung Memorial Hospital, Taoyuan, Taiwan,College of Medicine, Chang Gung University, Taoyuan, Taiwan,*Correspondence: Jau-Yuan Chen ✉
| |
Collapse
|
10
|
Khokhlova A, Myachina T, Butova X, Kochurova A, Polyakova E, Galagudza M, Solovyova O, Kopylova G, Shchepkin D. The Acute Effects of Leptin on the Contractility of Isolated Rat Atrial and Ventricular Cardiomyocytes. Int J Mol Sci 2022; 23:ijms23158356. [PMID: 35955485 PMCID: PMC9369024 DOI: 10.3390/ijms23158356] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Revised: 07/22/2022] [Accepted: 07/27/2022] [Indexed: 02/01/2023] Open
Abstract
Leptin is a pleiotropic peptide playing an important role in the regulation of cardiac functions. It is not clear whether leptin directly modulates the mechanical function of atrial cardiomyocytes. We compared the acute effects of leptin on the characteristics of mechanically non-loaded sarcomere shortening and cytosolic Ca2+ concentration ([Ca2+]i) transients in single rat atrial and ventricular cardiomyocytes. We also studied the functional properties of myosin obtained from cardiomyocytes using an in vitro motility assay and assessed the sarcomeric protein phosphorylation. Single cardiomyocytes were exposed to 5, 20, and 60 nM leptin for 60 min. In ventricular cardiomyocytes, 60 nM leptin depressed sarcomere shortening amplitude and decreased the rates of shortening and relaxation. These effects were accompanied by a decrease in the phosphorylation of cMyBP-C, an increase in Tpm phosphorylation, and a slowdown of the sliding velocity of thin filaments over myosin in the in vitro motility assay. In contrast, in atrial cardiomyocytes, the phosphorylation of cMyBP-C and TnI increased, and the characteristics of sarcomere shortening did not change. Leptin had no effect on the characteristics of [Ca2+]i transients in ventricular cardiomyocytes, while 5 nM leptin prolonged [Ca2+]i transients in atrial cardiomyocytes. Thus, leptin-induced changes in contractility of ventricular cardiomyocytes may be attributed to the direct effects of leptin on cross-bridge kinetics and sarcomeric protein properties rather than changes in [Ca2+]i. We also suggest that the observed differences between atrial and ventricular cardiomyocytes may be associated with the peculiarities of the expression of leptin receptors, as well as signaling pathways in the atrial and ventricular myocardium.
Collapse
Affiliation(s)
- Anastasia Khokhlova
- Institute of Immunology and Physiology, Russian Academy of Sciences, Pervomajskaya Str. 106, 620049 Yekaterinburg, Russia; (T.M.); (X.B.); (A.K.); (O.S.); (G.K.); (D.S.)
- Correspondence:
| | - Tatiana Myachina
- Institute of Immunology and Physiology, Russian Academy of Sciences, Pervomajskaya Str. 106, 620049 Yekaterinburg, Russia; (T.M.); (X.B.); (A.K.); (O.S.); (G.K.); (D.S.)
| | - Xenia Butova
- Institute of Immunology and Physiology, Russian Academy of Sciences, Pervomajskaya Str. 106, 620049 Yekaterinburg, Russia; (T.M.); (X.B.); (A.K.); (O.S.); (G.K.); (D.S.)
| | - Anastasia Kochurova
- Institute of Immunology and Physiology, Russian Academy of Sciences, Pervomajskaya Str. 106, 620049 Yekaterinburg, Russia; (T.M.); (X.B.); (A.K.); (O.S.); (G.K.); (D.S.)
| | - Ekaterina Polyakova
- Almazov National Medical Research Centre, Institute of Experimental Medicine, Akkuratova Str. 2, 197341 Saint-Petersburg, Russia; (E.P.); (M.G.)
| | - Michael Galagudza
- Almazov National Medical Research Centre, Institute of Experimental Medicine, Akkuratova Str. 2, 197341 Saint-Petersburg, Russia; (E.P.); (M.G.)
| | - Olga Solovyova
- Institute of Immunology and Physiology, Russian Academy of Sciences, Pervomajskaya Str. 106, 620049 Yekaterinburg, Russia; (T.M.); (X.B.); (A.K.); (O.S.); (G.K.); (D.S.)
| | - Galina Kopylova
- Institute of Immunology and Physiology, Russian Academy of Sciences, Pervomajskaya Str. 106, 620049 Yekaterinburg, Russia; (T.M.); (X.B.); (A.K.); (O.S.); (G.K.); (D.S.)
| | - Daniil Shchepkin
- Institute of Immunology and Physiology, Russian Academy of Sciences, Pervomajskaya Str. 106, 620049 Yekaterinburg, Russia; (T.M.); (X.B.); (A.K.); (O.S.); (G.K.); (D.S.)
| |
Collapse
|
11
|
Misch M, Puthanveetil P. The Head-to-Toe Hormone: Leptin as an Extensive Modulator of Physiologic Systems. Int J Mol Sci 2022; 23:ijms23105439. [PMID: 35628271 PMCID: PMC9141226 DOI: 10.3390/ijms23105439] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Revised: 04/30/2022] [Accepted: 05/10/2022] [Indexed: 12/12/2022] Open
Abstract
Leptin is a well-known hunger-sensing peptide hormone. The role of leptin in weight gain and metabolic homeostasis has been explored for the past two decades. In this review, we have tried to shed light upon the impact of leptin signaling on health and diseases. At low or moderate levels, this peptide hormone supports physiological roles, but at chronically higher doses exhibits detrimental effects on various systems. The untoward effects we observe with chronically higher levels of leptin are due to their receptor-mediated effect or due to leptin resistance and are not well studied. This review will help us in understanding the non-anorexic roles of leptin, including their contribution to the metabolism of various systems and inflammation. We will be able to get an alternative perspective regarding the physiological and pathological roles of this mysterious peptide hormone.
Collapse
Affiliation(s)
- Monica Misch
- Chicago College of Osteopathic Medicine, Midwestern University, Downers Grove, IL 60515, USA;
| | - Prasanth Puthanveetil
- Department of Pharmacology, College of Graduate Studies, Midwestern University, Downers Grove, IL 60515, USA
- Correspondence: ; Tel.: +1-630-960-3935
| |
Collapse
|
12
|
Delaney KZ, Santosa S. Sex differences in regional adipose tissue depots pose different threats for the development of Type 2 diabetes in males and females. Obes Rev 2022; 23:e13393. [PMID: 34985183 DOI: 10.1111/obr.13393] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Revised: 10/26/2021] [Accepted: 10/28/2021] [Indexed: 01/08/2023]
Abstract
Type 2 diabetes mellitus (T2DM) affects males and females disproportionately. In midlife, more males have T2DM than females. The sex difference in T2DM prevalence is, in part, explained by differences in regional adipose tissue characteristics. With obesity, changes to regional adipokine and cytokine release increases the risk of T2DM in both males and females with males having greater levels of TNFα and females having greater levels of leptin, CRP, and adiponectin. Regional immune cell infiltration appears to be pathogenic in both sexes via different routes as males with obesity have greater VAT ATM and a decrease in the protective Treg cells, whereas females have greater SAT ATM and T cells. Lastly, the ability of female adipose tissue to expand all regions through hyperplasia, rather than hypertrophy, protects them against the development of large insulin-resistant adipocytes that dominate male adipose tissue. The objective of this review is to discuss how sex may affect regional differences in adipose tissue characteristics and how these differences may distinguish the development of T2DM in males and females. In doing so, we will show that the origins of T2DM development differ between males and females.
Collapse
Affiliation(s)
- Kerri Z Delaney
- Department of Health, Kinesiology and Applied Physiology, Concordia University, Montréal, Québec, Canada.,Metabolism, Obesity and Nutrition Lab, PERFORM Centre, Concordia University, Montréal, Québec, Canada.,Centre de recherche - Axe maladies chroniques, Centre intégré universitaire de santé et de services sociaux du Nord-de-l'Ile-de-Montréal, Hôpital du Sacré-Coeur de Montréal, Montréal, Québec, Canada
| | - Sylvia Santosa
- Department of Health, Kinesiology and Applied Physiology, Concordia University, Montréal, Québec, Canada.,Metabolism, Obesity and Nutrition Lab, PERFORM Centre, Concordia University, Montréal, Québec, Canada.,Centre de recherche - Axe maladies chroniques, Centre intégré universitaire de santé et de services sociaux du Nord-de-l'Ile-de-Montréal, Hôpital du Sacré-Coeur de Montréal, Montréal, Québec, Canada
| |
Collapse
|
13
|
Malenica M, Meseldžić N. Oxidative stress and obesity. ARHIV ZA FARMACIJU 2022. [DOI: 10.5937/arhfarm72-36123] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022] Open
Abstract
Obesity is a disease of excessive accumulation of adipose tissue due to an increased energy intake which is disproportionate to the energy expenditure in the body. The visceral adipose tissue in the obese accumulated in that way increases the risk of developing a number of metabolic and cardiovascular diseases. Disorders such as diabetes, dyslipidemia, inflammation, endothelial dysfunction and mitochondria can contribute to the development of oxidative stress, which is especially pronounced in the abdominal type of obesity. Obesity can induce systemic oxidative stress through a variety of biochemical mechanisms. Although ROS is generated in a large number of cells, mitochondria play a significant role in their intracellular production through the process of oxidative phosphorylation of the respiratory chain, and in fatty acid oxidation reactions. Oxidative stress is a unique link between the various molecular disorders present in the development of insulin resistance that plays a key role in the pathogenesis and progression of chronic metabolic, proinflammatory diseases. The progression of insulin resistance is also affected by inflammation. Both of these can be the cause and the consequence of obesity. The synthesis of the inflammatory mediators is induced by oxidative stress, thus bringing the inflammation and the oxidative stress into a very significant relation. This review aims to highlight recent findings on the role of oxidative stress in the pathogenesis of obesity, with special reference to the mechanisms that explain its occurrence.
Collapse
|
14
|
Larsson A, Lipcsey M, Hultström M, Frithiof R, Eriksson M. Plasma Leptin Is Increased in Intensive Care Patients with COVID-19-An Investigation Performed in the PronMed-Cohort. Biomedicines 2021; 10:biomedicines10010004. [PMID: 35052684 PMCID: PMC8773415 DOI: 10.3390/biomedicines10010004] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Revised: 12/17/2021] [Accepted: 12/19/2021] [Indexed: 12/27/2022] Open
Abstract
COVID-19 has shaken the world and intensive care units (ICU) have been challenged by numerous patients suffering from a previously unknown disease. Leptin is a polypeptide pleiotropic hormone, mainly expressed by adipocytes. It acts as a proinflammatory cytokine and is associated with several conditions, known to increase the risk of severe COVID-19. Very little is known about leptin in severe viral disorders. Plasma leptin was analyzed in 222 out of 229 patients with severe COVID-19 on admission to an ICU at Uppsala University Hospital, a tertiary care hospital in Sweden, and compared to plasma leptin in 25 healthy blood donors. COVID-19 was confirmed by positive PCR. Leptin levels were significantly higher in patients with COVID-19 (18.3 ng × mL−1; IQR = 30.4), than in healthy controls (7.8 ng × mL−1; IQR = 6.4). Women had significantly higher leptin values (22.9 ng × mL−1; IQR = 29.8) than men (17.5 ng × mL−1; IQR = 29.9). Mortality at 30 days was 23% but was not associated with increased leptin levels. Neither median duration of COVID-19 before admission to ICU (10 days; IQR = 4) or median length of ICU stay (8 days; IQR = 11) correlated with the plasma leptin levels. Leptin levels in COVID-19 were higher in females than in males. Both treatment (e.g., use of corticosteroids) and prophylaxis (vaccines) have been improved since the start of the COVID-19 pandemic, which may contribute to some difficulties in deciphering relations between COVID-19 and leptin.
Collapse
Affiliation(s)
- Anders Larsson
- Department of Medical Sciences, Clinical Chemistry, Uppsala University, 751 85 Uppsala, Sweden
- Correspondence: ; Tel.: +46-186114271
| | - Miklós Lipcsey
- Department of Surgical Sciences, Anaesthesiology and Intensive Care Medicine, Uppsala University, 751 85 Uppsala, Sweden; (M.L.); (M.H.); (R.F.); (M.E.)
- Hedenstierna Laboratory, Department of Surgical Sciences, Uppsala University, 751 85 Uppsala, Sweden
| | - Michael Hultström
- Department of Surgical Sciences, Anaesthesiology and Intensive Care Medicine, Uppsala University, 751 85 Uppsala, Sweden; (M.L.); (M.H.); (R.F.); (M.E.)
- Department of Medical Cell Biology, Integrative Physiology, Uppsala University, 751 23 Uppsala, Sweden
| | - Robert Frithiof
- Department of Surgical Sciences, Anaesthesiology and Intensive Care Medicine, Uppsala University, 751 85 Uppsala, Sweden; (M.L.); (M.H.); (R.F.); (M.E.)
| | - Mats Eriksson
- Department of Surgical Sciences, Anaesthesiology and Intensive Care Medicine, Uppsala University, 751 85 Uppsala, Sweden; (M.L.); (M.H.); (R.F.); (M.E.)
| |
Collapse
|
15
|
The Role of Obesity-Induced Perivascular Adipose Tissue (PVAT) Dysfunction in Vascular Homeostasis. Nutrients 2021; 13:nu13113843. [PMID: 34836100 PMCID: PMC8621306 DOI: 10.3390/nu13113843] [Citation(s) in RCA: 54] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2021] [Revised: 10/23/2021] [Accepted: 10/26/2021] [Indexed: 12/17/2022] Open
Abstract
Perivascular adipose tissue (PVAT) is an additional special type of adipose tissue surrounding blood vessels. Under physiological conditions, PVAT plays a significant role in regulation of vascular tone, intravascular thermoregulation, and vascular smooth muscle cell (VSMC) proliferation. PVAT is responsible for releasing adipocytes-derived relaxing factors (ADRF) and perivascular-derived relaxing factors (PDRF), which have anticontractile properties. Obesity induces increased oxidative stress, an inflammatory state, and hypoxia, which contribute to PVAT dysfunction. The exact mechanism of vascular dysfunction in obesity is still not well clarified; however, there are some pathways such as renin-angiotensin-aldosterone system (RAAS) disorders and PVAT-derived factor dysregulation, which are involved in hypertension and endothelial dysfunction development. Physical activity has a beneficial effect on PVAT function among obese patients by reducing the oxidative stress and inflammatory state. Diet, which is the second most beneficial non-invasive strategy in obesity treatment, may have a positive impact on PVAT-derived factors and may restore the balance in their concentration.
Collapse
|
16
|
Dadousis C, Somavilla A, Ilska JJ, Johnsson M, Batista L, Mellanby RJ, Headon D, Gottardo P, Whalen A, Wilson D, Dunn IC, Gorjanc G, Kranis A, Hickey JM. A genome-wide association analysis for body weight at 35 days measured on 137,343 broiler chickens. Genet Sel Evol 2021; 53:70. [PMID: 34496773 PMCID: PMC8424881 DOI: 10.1186/s12711-021-00663-w] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Accepted: 08/23/2021] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND Body weight (BW) is an economically important trait in the broiler (meat-type chickens) industry. Under the assumption of polygenicity, a "large" number of genes with "small" effects is expected to control BW. To detect such effects, a large sample size is required in genome-wide association studies (GWAS). Our objective was to conduct a GWAS for BW measured at 35 days of age with a large sample size. METHODS The GWAS included 137,343 broilers spanning 15 pedigree generations and 392,295 imputed single nucleotide polymorphisms (SNPs). A false discovery rate of 1% was adopted to account for multiple testing when declaring significant SNPs. A Bayesian ridge regression model was implemented, using AlphaBayes, to estimate the contribution to the total genetic variance of each region harbouring significant SNPs (1 Mb up/downstream) and the combined regions harbouring non-significant SNPs. RESULTS GWAS revealed 25 genomic regions harbouring 96 significant SNPs on 13 Gallus gallus autosomes (GGA1 to 4, 8, 10 to 15, 19 and 27), with the strongest associations on GGA4 at 65.67-66.31 Mb (Galgal4 assembly). The association of these regions points to several strong candidate genes including: (i) growth factors (GGA1, 4, 8, 13 and 14); (ii) leptin receptor overlapping transcript (LEPROT)/leptin receptor (LEPR) locus (GGA8), and the STAT3/STAT5B locus (GGA27), in connection with the JAK/STAT signalling pathway; (iii) T-box gene (TBX3/TBX5) on GGA15 and CHST11 (GGA1), which are both related to heart/skeleton development); and (iv) PLAG1 (GGA2). Combined together, these 25 genomic regions explained ~ 30% of the total genetic variance. The region harbouring significant SNPs that explained the largest portion of the total genetic variance (4.37%) was on GGA4 (~ 65.67-66.31 Mb). CONCLUSIONS To the best of our knowledge, this is the largest GWAS that has been conducted for BW in chicken to date. In spite of the identified regions, which showed a strong association with BW, the high proportion of genetic variance attributed to regions harbouring non-significant SNPs supports the hypothesis that the genetic architecture of BW35 is polygenic and complex. Our results also suggest that a large sample size will be required for future GWAS of BW35.
Collapse
Affiliation(s)
| | | | - Joanna J. Ilska
- The Roslin Institute, University of Edinburgh, Midlothian, UK
| | - Martin Johnsson
- The Roslin Institute, University of Edinburgh, Midlothian, UK
- Department of Animal Breeding and Genetics, Swedish University of Agricultural Sciences, Uppsala, Sweden
| | - Lorena Batista
- The Roslin Institute, University of Edinburgh, Midlothian, UK
| | | | - Denis Headon
- The Roslin Institute, University of Edinburgh, Midlothian, UK
| | - Paolo Gottardo
- Italian Brown Breeders Association, Loc. Ferlina 204, 37012 Bussolengo, Italy
| | - Andrew Whalen
- The Roslin Institute, University of Edinburgh, Midlothian, UK
| | - David Wilson
- The Roslin Institute, University of Edinburgh, Midlothian, UK
| | - Ian C. Dunn
- The Roslin Institute, University of Edinburgh, Midlothian, UK
| | - Gregor Gorjanc
- The Roslin Institute, University of Edinburgh, Midlothian, UK
| | - Andreas Kranis
- The Roslin Institute, University of Edinburgh, Midlothian, UK
- Aviagen Ltd, Midlothian, UK
| | - John M. Hickey
- The Roslin Institute, University of Edinburgh, Midlothian, UK
| |
Collapse
|
17
|
Wang X, Karvonen-Gutierrez CA, Mukherjee B, Herman WH, Park SK. Urinary metals and adipokines in midlife women: The Study of Women's Health Across the nation (SWAN). ENVIRONMENTAL RESEARCH 2021; 196:110426. [PMID: 33157106 PMCID: PMC8093324 DOI: 10.1016/j.envres.2020.110426] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/09/2020] [Revised: 10/23/2020] [Accepted: 10/31/2020] [Indexed: 05/05/2023]
Abstract
BACKGROUND Information on the associations between metal exposures and adipokines in human populations is limited and results are inconsistent. We evaluated the associations between metals and adipokines. METHODS Urinary concentrations of 15 metals (arsenic, barium, cadmium, cobalt, cesium, copper, mercury, manganese, molybdenum, nickel, lead, antimony, tin, thallium, and zinc) were measured in 1999-2000 among 1228 women of the Study of Women's Health Across the Nation Multi-Pollutant Study. Serum adipokines including high molecular weight (HMW)-adiponectin, leptin, and soluble leptin receptor (sOB-R) were measured at the follow-up visit (2002-2003). Linear regression models with adaptive elastic-net (AENET) were fit to identify metals associated with adipokines and to compute estimated percent changes in adipokines for one standard deviation increase in log-transformed urinary metal concentrations. RESULTS After adjustment for confounders, urinary molybdenum was associated with a 5.54% higher level (95% CI: 1.36%, 9.90%), whereas cadmium was associated with a 4.53% lower level (95% CI: -8.17%, -0.76%) of HMW-adiponectin. Urinary molybdenum was also associated with a 5.95% lower leptin level (95% CI: -10.15%, -1.56%) and a 2.98% (95% CI: 0.69%, 5.32%) higher sOB-R level. Urinary cesium and lead were associated with a 3.58% (95% CI: -6.06%, -1.03%) and a 2.53% (95% CI: -4.80%, -0.21%) lower level of sOB-R, respectively. CONCLUSIONS Our findings suggest that molybdenum was associated with favorable profiles of HMW-adiponectin, leptin, and sOB-R. Exposures to cadmium, cesium, and lead were associated with adverse adipokine profiles.
Collapse
Affiliation(s)
- Xin Wang
- Department of Epidemiology, School of Public Health, University of Michigan, Ann Arbor, MI, USA
| | | | - Bhramar Mukherjee
- Department of Biostatistics, School of Public Health, University of Michigan, Ann Arbor, MI, USA
| | - William H Herman
- Department of Epidemiology, School of Public Health, University of Michigan, Ann Arbor, MI, USA; Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Sung Kyun Park
- Department of Epidemiology, School of Public Health, University of Michigan, Ann Arbor, MI, USA; Department of Environmental Health Sciences, School of Public Health, University of Michigan, Ann Arbor, MI, USA.
| |
Collapse
|
18
|
Dessie G, Ayelign B, Akalu Y, Shibabaw T, Molla MD. Effect of Leptin on Chronic Inflammatory Disorders: Insights to Therapeutic Target to Prevent Further Cardiovascular Complication. Diabetes Metab Syndr Obes 2021; 14:3307-3322. [PMID: 34305402 PMCID: PMC8296717 DOI: 10.2147/dmso.s321311] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/22/2021] [Accepted: 06/30/2021] [Indexed: 01/08/2023] Open
Abstract
In response to obesity-associated chronic inflammatory disorders, adipose tissue releases a biologically active peptide known as leptin. Leptin activates the secretion of chemical mediators, which contribute to the pathogenesis of chronic inflammatory disorders, such as rheumatoid arthritis (RA), systemic lupus erythematosus (SLE) and psoriasis. Conversely, adiposity and obesity are the major aggravating risk factors in the pathogenesis of metabolic syndrome (MetS), including type II diabetes mellitus and obesity-associated hypertension. Elevated level of leptin in obesity-associated hypertension causes an increase in the production of aldosterone, which also results in elevation of arterial blood pressure. Hyperleptinemia is associated with the progress of the atherosclerosis through secretion of pro-inflammatory cytokines, like interleukin 6 (IL-6), tumor necrosis factor α (TNF-α), IL-17, and other cytokines to promote inflammation. The release of those cytokines leads to chronic inflammatory disorders and obesity-associated MetS. Thus, the aberrant leptin level in both MetS and chronic inflammatory disorders also leads to the complication of cardiovascular diseases (CVD). Therapeutic target of leptin regarding its pro-inflammatory effect and dysregulated sympathetic nervous system activity may prevent further cardiovascular complication. This review mainly assesses the mechanism of leptin on the pathogenesis and further cardiovascular risk complication of chronic inflammatory disorders.
Collapse
Affiliation(s)
- Gashaw Dessie
- Department of Biochemistry, School of Medicine, College of Medicine and Health Sciences, University of Gondar, Gondar, Ethiopia
- Correspondence: Gashaw Dessie Tel +251 975152796 Email
| | - Birhanu Ayelign
- Department of Immunology and Molecular Biology, School of Biomedical and Laboratory Science, College of Medicine and Health Sciences, University of Gondar, Gondar, Ethiopia
| | - Yonas Akalu
- Department of Physiology, School of Medicine, College of Medicine and Health Sciences, University of Gondar, Gondar, Ethiopia
| | - Tewodros Shibabaw
- Department of Biochemistry, School of Medicine, College of Medicine and Health Sciences, University of Gondar, Gondar, Ethiopia
| | - Meseret Derbew Molla
- Department of Biochemistry, School of Medicine, College of Medicine and Health Sciences, University of Gondar, Gondar, Ethiopia
| |
Collapse
|
19
|
Muhammad HFL, Pratama SA, Sulistyoningrum DC, Hartono MN, Huriyati E, Lee YY, Muda WAMW. The impact of sleep quality and duration on leptin, appetite, and obesity indices in adults. MEDICAL JOURNAL OF INDONESIA 2020. [DOI: 10.13181/mji.oa.203707] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022] Open
Abstract
BACKGROUND Poor sleep duration and quality were previously reported to increase the risk of obesity. This study was aimed to evaluate the impact of sleep quality and duration on leptin, appetite, and adiposity in Indonesian adults.
METHODS This cross-sectional study recruited adults in Yogyakarta, Indonesia, in 2016. Sleep quality was assessed using the Pittsburgh sleep quality index, appetite was evaluated using the community nutrition appetite questionnaire, and dietary intake was assessed by interviews using the semi-quantitative food frequency questionnaire. Sleep duration was also asked. Obesity indices were measured using the body mass index (BMI), waist-hip circumference, and percentage of body fat. Leptin was analyzed using the enzyme-linked immunosorbent assay. A Spearman analysis was done to evaluate the correlation between sleep quality, sleep duration, anthropometric measures, dietary intake, appetite, and leptin.
RESULTS 244 adults participated in this study. This study showed that lower sleep quality and duration was significantly correlated with higher body weight (r = 0.129, p = 0.043 and r = −0.228, p<0.001), BMI (r = 0.176, p = 0.006 and r = −0.202, p = 0.001), and waist circumference (r = 0.179, p = 0.005 and r = −0.254, p<0.001). There was a correlation between poor sleep quality and higher leptin concentration (r = 0.186, p = 0.004). Sleep quality and duration were not associated with appetite (r = 0.109, p = 0.109 and r = −0.043, p = 0.500).
CONCLUSIONS This study found that lower sleep quality was correlated with higher BMI, higher leptin concentration, but not appetite.
Collapse
|
20
|
Zvolinskaya EY, Mamedov MN, Potievskaya VI, Ivanov SA, Kaprin AD. [Role of modified cardiovascular risk factors in development of oncologic diseases]. KARDIOLOGIIA 2020; 60:110-121. [PMID: 33131482 DOI: 10.18087/cardio.2020.9.n910] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/28/2019] [Revised: 01/20/2020] [Accepted: 01/20/2020] [Indexed: 12/24/2022]
Abstract
Cardiovascular (CVD) and oncological diseases (OD) are the main causes of death worldwide and account for a heavy burden on economy, disability and mortality in many countries. Clear understanding of the mechanisms shared by CVD and cancer is important for increasing the life span and quality of life in cancer survivors as well as for preventing comorbidities and correct instructing the patients about risk factors and lifestyle modifications. Both groups of diseases share risk factors, including smoking, obesity, diabetes mellitus, alcohol consumption, unhealthy diet, etc. Along with these factors, inflammation may play a key role as it promotes both types of diseases and accompanies obesity, diabetes mellitus, arterial hypertension, and dyslipidemia. Better understanding of the interaction between CVD and cancer will allow creating common effective diagnostic and preventive strategies and safe approaches to the treatment.
Collapse
Affiliation(s)
- E Yu Zvolinskaya
- National Medical Research Center for Therapy and Preventive Medicine of the Ministry of Healthcare, Moscow, Russia
| | - M N Mamedov
- National Medical Research Center for Therapy and Preventive Medicine of the Ministry of Healthcare, Moscow, Russia
| | - V I Potievskaya
- National Medical Radiology Research Center of the Ministry of Healthcare, Moscow, Russia
| | - S A Ivanov
- Medical Radiological Research Center the branch of National Medical Radiology Research Center of the Ministry of Healthcare, Obninsk, Russia
| | - A D Kaprin
- National Medical Radiology Research Center of the Ministry of Healthcare, Moscow, Russia
| |
Collapse
|
21
|
Nigro E, Perrotta F, Polito R, D'Agnano V, Scialò F, Bianco A, Daniele A. Metabolic Perturbations and Severe COVID-19 Disease: Implication of Molecular Pathways. Int J Endocrinol 2020; 2020:8896536. [PMID: 33312199 PMCID: PMC7703458 DOI: 10.1155/2020/8896536] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/16/2020] [Revised: 11/04/2020] [Accepted: 11/17/2020] [Indexed: 01/08/2023] Open
Abstract
Coronavirus disease (COVID-19) is caused by SARS-CoV-2 virus, which can result in serious respiratory illnesses such as pneumonia leading to respiratory failure. It was first reported in Wuhan, Hubei, China, in December 2019 and rapidly spread globally, becoming a pandemic in March 2020. Among comorbidities observed in SARS-CoV-2 positive patients, hypertension (68.3%) and type 2-diabetes (30.1%) are the most frequent conditions. Although symptoms are highly heterogeneous (ranging from absence of symptoms to severe acute respiratory failure), patients with metabolic-associated diseases often experience worse COVID-19 outcomes. This review investigates the association between metabolic disorders and COVID-19 severity, exploring the molecular mechanisms potentially underlying this relationship and those that are responsible for more severe COVID-19 outcomes. In addition, the role of the main biological processes that may connect metabolic alterations to SARS-CoV-2 infection such as hyperglycemia, immune system deregulation, ACE-2 receptor modulation, and inflammatory response is described. The impact of metabolic disorders on the prognosis of COVID-19 has major implications in public health especially for countries affected by a high incidence of metabolic diseases.
Collapse
Affiliation(s)
- Ersilia Nigro
- Dipartimento di Scienze e Tecnologie Ambientali Biologiche Farmaceutiche, Università Degli Studi Della Campania “Luigi Vanvitelli”, Via G. Vivaldi 42, Caserta 81100, Italy
- CEINGE-Biotecnologie Avanzate Scarl, Via G. Salvatore 486, Napoli 80145, Italy
| | - Fabio Perrotta
- Dipartimento di Medicina e Scienze Della Salute “V. Tiberio”, Università Del Molise, Campobasso 86100, Italy
| | - Rita Polito
- CEINGE-Biotecnologie Avanzate Scarl, Via G. Salvatore 486, Napoli 80145, Italy
| | - Vito D'Agnano
- Dipartimento di Scienze Mediche Traslazionali e Chirurgiche, Università Della Campania “L. Vanvitelli”, Napoli 80131, Italy
| | - Filippo Scialò
- Dipartimento di Scienze Mediche Traslazionali e Chirurgiche, Università Della Campania “L. Vanvitelli”, Napoli 80131, Italy
| | - Andrea Bianco
- Dipartimento di Scienze Mediche Traslazionali e Chirurgiche, Università Della Campania “L. Vanvitelli”, Napoli 80131, Italy
| | - Aurora Daniele
- Dipartimento di Scienze e Tecnologie Ambientali Biologiche Farmaceutiche, Università Degli Studi Della Campania “Luigi Vanvitelli”, Via G. Vivaldi 42, Caserta 81100, Italy
- CEINGE-Biotecnologie Avanzate Scarl, Via G. Salvatore 486, Napoli 80145, Italy
| |
Collapse
|
22
|
Imerbtham T, Thitiwuthikiat P, Jongjitwimol J, Nuamchit T, Yingchoncharoen T, Siriwittayawan D. Leptin Levels are Associated with Subclinical Cardiac Dysfunction in Obese Adolescents. Diabetes Metab Syndr Obes 2020; 13:925-933. [PMID: 32273744 PMCID: PMC7108875 DOI: 10.2147/dmso.s245048] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/13/2020] [Accepted: 03/11/2020] [Indexed: 12/24/2022] Open
Abstract
PURPOSE The purposes of this study were to use speckle tracking echocardiography to confirm the influence of obesity on cardiac functions and to assess their relationships with leptin and uric acid levels in obese adolescents. METHODS Eighty-one participants aged 16-19 years were recruited and classified as either non-obese (n = 30) or obese (n = 51). Global longitudinal strain (GLS), leptin and uric acid levels for each group were assessed and compared. The data from obese participants were then compared based on their leptin levels and analyzed for correlation using regression analysis. RESULTS The obese group had significantly lower absolute GLS compared to the non-obese group (19.10 ± 0.30 versus 21.10 ± 0.30%, p < 0.001). In obese group, subclinical cardiac dysfunction was worse in the hyperleptinemic group than that of the normoleptinemic group (p = 0.03). Multivariate regression analysis showed that leptin and triglyceride levels were negatively associated with absolute GLS. Leptin could predict the absolute GLS with β = -0.35 (p = 0.02). CONCLUSION Subclinical left ventricular systolic dysfunction was found in obese adolescents, while GLS was worse in the hyperleptinemic subjects. Leptin, but not uric acid, levels were associated with a worsening of GLS.
Collapse
Affiliation(s)
- Thamonwan Imerbtham
- Department of Cardio-Thoracic Technology, Faculty of Allied Health Sciences, Naresuan University, Phitsanulok, Thailand
| | - Piyanuch Thitiwuthikiat
- Department of Cardio-Thoracic Technology, Faculty of Allied Health Sciences, Naresuan University, Phitsanulok, Thailand
| | - Jirapas Jongjitwimol
- Department of Medical Technology, Faculty of Allied Health Sciences, Naresuan University, Phitsanulok, Thailand
| | - Teonchit Nuamchit
- Department of Cardio-Thoracic Technology, Faculty of Allied Health Sciences, Naresuan University, Phitsanulok, Thailand
| | | | - Duangduan Siriwittayawan
- Department of Cardio-Thoracic Technology, Faculty of Allied Health Sciences, Naresuan University, Phitsanulok, Thailand
- Correspondence: Duangduan Siriwittayawan Department of Cardio-Thoracic Technology, Faculty of Allied Health Sciences, Naresuan University, Phitsanulok65000, ThailandTel +66 55 966 417Fax +66 55 966 234 Email
| |
Collapse
|
23
|
Muhammad HFL, van Baak MA, Mariman EC, Sulistyoningrum DC, Huriyati E, Lee YY, Wan Muda WAM. Dietary Inflammatory Index Score and Its Association with Body Weight, Blood Pressure, Lipid Profile, and Leptin in Indonesian Adults. Nutrients 2019; 11:nu11010148. [PMID: 30641979 PMCID: PMC6356884 DOI: 10.3390/nu11010148] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2018] [Revised: 12/24/2018] [Accepted: 12/25/2018] [Indexed: 02/06/2023] Open
Abstract
It was previously reported that dietary intake is an important trigger for systemic inflammation and one of the lifestyle factors for the development of cardiovascular diseases. The aim of this study was to evaluate the association between Dietary Inflammatory Index (DII) score and body weight, blood pressure, lipid profile and leptin in an Indonesian population. This was a cross-sectional study conducted in 503 Indonesian adults. The DII score was calculated based on data of 30 nutrients and food components. Anthropometric profile, blood pressure, lipid profile, and leptin were measured. The association of these variables with the DII score was analyzed. The DII score was not associated with body weight, body mass index (BMI), body fat, waist circumference, hip circumference, systolic and diastolic blood pressure, triglycerides, and high-density lipoprotein (HDL) (both unadjusted and after adjustment for covariates). However, plasma leptin concentration was significantly associated with the DII score (B = 0.096, p = 0.020). Plasma leptin also increased significantly across tertiles of the DII score (ANCOVA, p = 0.031). This positive association between the DII score and plasma leptin concentration suggests a role for the inflammatory properties of the diet in regulating adipose tissue inflammation.
Collapse
Affiliation(s)
- Harry Freitag Luglio Muhammad
- Department of Nutrition and Health, Universitas Gadjah Mada, Yogyakarta 55281, Indonesia.
- NUTRIM School of Translational Nutrition and Metabolism, Department of Human Biology, Maastricht University, 6200 MD Maastricht, The Netherlands.
| | - Marleen A van Baak
- NUTRIM School of Translational Nutrition and Metabolism, Department of Human Biology, Maastricht University, 6200 MD Maastricht, The Netherlands.
| | - Edwin C Mariman
- NUTRIM School of Translational Nutrition and Metabolism, Department of Human Biology, Maastricht University, 6200 MD Maastricht, The Netherlands.
| | | | - Emy Huriyati
- Department of Nutrition and Health, Universitas Gadjah Mada, Yogyakarta 55281, Indonesia.
| | - Yi Yi Lee
- International Institute for Global Health, United Nations University, Kuala Lumpur 16150, Malaysia.
| | | |
Collapse
|
24
|
Kain D, Simon AJ, Greenberg A, Ben Zvi D, Gilburd B, Schneiderman J. Cardiac leptin overexpression in the context of acute MI and reperfusion potentiates myocardial remodeling and left ventricular dysfunction. PLoS One 2018; 13:e0203902. [PMID: 30312306 PMCID: PMC6193573 DOI: 10.1371/journal.pone.0203902] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2018] [Accepted: 08/29/2018] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND Acute MI induces leptin expression in the heart, however the role of myocardial leptin in cardiac ischemia and reperfusion (IR) remains unknown. To shed light on the effects of elevated levels of leptin in the myocardium, we overexpressed cardiac leptin and assessed local remodeling and myocardial function in this context. METHODS AND RESULTS Cardiac leptin overexpression was stimulated in mice undergoing IR by a single intraperitoneal injection of leptin antagonist (LepA). All mice exhibited a normal pattern of body weight gain. A rapid, long-term upregulation of leptin mRNA was demonstrated in the heart, adipose, and liver tissues in IR/LepA-treated mice. Overexpressed cardiac leptin mRNA extended beyond postoperative day (POD) 30. Plasma leptin peaked 7.5 hours postoperatively, especially in IR/LepA-treated mice, subsiding to normal levels by 24 hours. On POD-30 IR/LepA-treated mice demonstrated cardiomyocyte hypertrophy and perivascular fibrosis compared to IR/saline controls. Echocardiography on POD-30 demonstrated eccentric hypertrophy and systolic dysfunction in IR/LepA. We recorded reductions in Ejection Fraction (p<0.001), Fraction Shortening (p<0.01), and Endocardial Fraction Area Change (p<0.01), and an increase in Endocardial Area Change (p<0.01). Myocardial remodeling in the context of IR and cardiac leptin overexpression was associated with increased cardiac TGFβ ligand expression, activated Smad2, and downregulation of STAT3 activity. CONCLUSIONS Cardiac IR coinciding with increased myocardial leptin synthesis promotes cardiomyocyte hypertrophy and fibrosis and potentiates myocardial dysfunction. Plasma leptin levels do not reflect cardiac leptin synthesis, and may not predict leptin-related cardiovascular morbidity. Targeting cardiac leptin is a potential treatment for cardiac IR damage.
Collapse
Affiliation(s)
- David Kain
- Department of Neurobiology, Tel Aviv University, Tel Aviv, Israel
| | - Amos J. Simon
- Cancer Research and Institute of Hematology, Sheba Medical Center, Sackler Faculty of Medicine, Tel Aviv University, Israel
| | - Avraham Greenberg
- Department of Developmental Biology and Cancer Research, The institute for Medical Research Israel-Canada, The Hebrew University-Hadassah medical School, Jerusalem, Israel
| | - Danny Ben Zvi
- Department of Developmental Biology and Cancer Research, The institute for Medical Research Israel-Canada, The Hebrew University-Hadassah medical School, Jerusalem, Israel
| | - Boris Gilburd
- Center for Autoimmune Diseases, Sheba Medical Center, Sackler Faculty of Medicine, Tel Aviv University, Israel
| | - Jacob Schneiderman
- Department of Vascular Surgery, Sheba Medical Center, Sackler Faculty of Medicine, Tel Aviv University, Isreal
- * E-mail: ,
| |
Collapse
|
25
|
Katsiki N, Mikhailidis DP, Banach M. Leptin, cardiovascular diseases and type 2 diabetes mellitus. Acta Pharmacol Sin 2018; 39:1176-1188. [PMID: 29877321 PMCID: PMC6289384 DOI: 10.1038/aps.2018.40] [Citation(s) in RCA: 171] [Impact Index Per Article: 24.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/18/2018] [Accepted: 05/02/2018] [Indexed: 02/07/2023]
Abstract
Leptin, an adipokine that is implicated in the control of food intake via appetite suppression, may also stimulate oxidative stress, inflammation, thrombosis, arterial stiffness, angiogenesis and atherogenesis. These leptin-induced effects may predispose to the development of cardiovascular diseases. In the present review we discuss the evidence linking leptin levels with the presence, severity and/or prognosis of both coronary artery disease and non-cardiac vascular diseases such as stroke, carotid artery disease, peripheral artery disease (PAD) and abdominal aortic aneurysms (AAA) as well as with chronic kidney disease (CKD) and type 2 diabetes mellitus (T2DM). Leptin levels have been positively associated with the presence, severity, extent and lesion complexity of coronary atherosclerosis as well as with the presence, severity and poor clinical outcomes of both ischemic and hemorrhagic strokes. But conflicting results also exist. Furthermore, leptin was reported to independently predict common carotid intima-media thickness and carotid plaque instability. A link between hyperleptinemia and PAD has been reported, whereas limited data were available on the potential association between leptin and AAA. Elevated leptin concentrations have also been related to CKD incidence and progression as well as with insulin resistance, T2DM, micro- and macrovascular diabetic complications. Statins and antidiabetic drugs (including sitagliptin, metformin, pioglitazone, liraglutide and empagliflozin) may affect leptin levels. Further research is needed to establish the potential use (if any) of leptin as a therapeutic target in these diseases.
Collapse
Affiliation(s)
- Niki Katsiki
- Second Propedeutic Department of Internal Medicine, Medical School, Aristotle University of Thessaloniki, Hippocration Hospital, Thessaloniki, Greece
| | - Dimitri P Mikhailidis
- Department of Clinical Biochemistry, Royal Free Hospital Campus, University College London Medical School, University College London (UCL), London, UK.
| | - Maciej Banach
- Department of Hypertension, Medical University of Lodz, Lodz, Poland
| |
Collapse
|
26
|
Han YC, Ma B, Guo S, Yang M, Li LJ, Wang SJ, Tan J. Leptin regulates disc cartilage endplate degeneration and ossification through activation of the MAPK-ERK signalling pathway in vivo and in vitro. J Cell Mol Med 2018; 22:2098-2109. [PMID: 29372627 PMCID: PMC5867127 DOI: 10.1111/jcmm.13398] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2016] [Accepted: 08/22/2017] [Indexed: 12/23/2022] Open
Abstract
Recent findings demonstrate that leptin plays a significant role in chondrocyte and osteoblast differentiation. However, the mechanisms by which leptin acts on cartilage endplate (CEP) cells to give rise to calcification are still unclear. The aim of this study was to evaluate the effects of leptin that induced mineralization of CEP cells in vitro and in vivo. We constructed a rat model of lumbar disc degeneration and determined that leptin was highly expressed in the presence of CEP calcification. Rat CEP cells treated with or without leptin were used for in vitro analysis using RT‐PCR and Western blotting to examine the expression of osteocalcin (OCN) and runt‐related transcription factor 2 (Runx2). Both OCN and Runx2 expression levels were significantly increased in a dose‐ and time‐dependent manner. Leptin activated ERK1/2 and STAT3 phosphorylation in a time‐dependent manner. Inhibition of phosphorylated ERK1/2 using targeted siRNA suppressed leptin‐induced OCN and Runx2 expression and blocked the formation of mineralized nodules in CEP cells. We further demonstrated that exogenous leptin induced matrix mineralization of CEP cells in vivo. We suggest that leptin promotes the osteoblastic differentiation of CEP cells via the MAPK/ERK signal transduction pathway and may be used to investigate the mechanisms of disc degeneration.
Collapse
Affiliation(s)
- Ying-Chao Han
- Department of Orthopedics, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Bin Ma
- Department of Orthopedics, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Song Guo
- Department of Orthopedics, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Mingjie Yang
- Department of Orthopedics, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Li-Jun Li
- Department of Orthopedics, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Shan-Jin Wang
- Department of Spinal Surgery, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Jun Tan
- Department of Orthopedics, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| |
Collapse
|
27
|
Xie Y, Potter CMF, Le Bras A, Nowak WN, Gu W, Bhaloo SI, Zhang Z, Hu Y, Zhang L, Xu Q. Leptin Induces Sca-1 + Progenitor Cell Migration Enhancing Neointimal Lesions in Vessel-Injury Mouse Models. Arterioscler Thromb Vasc Biol 2017; 37:2114-2127. [PMID: 28935755 PMCID: PMC5671780 DOI: 10.1161/atvbaha.117.309852] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2017] [Accepted: 09/08/2017] [Indexed: 12/20/2022]
Abstract
Supplemental Digital Content is available in the text. Objective— Leptin is an adipokine initially thought to be a metabolic factor. Recent publications have shown its roles in inflammation and vascular disease, to which Sca-1+ vascular progenitor cells within the vessel wall may contribute. We sought to elucidate the effects of leptin on Sca-1+ progenitor cells migration and neointimal formation and to understand the underlying mechanisms. Approach and Results— Sca-1+ progenitor cells from the vessel wall of Lepr+/+ and Lepr−/− mice were cultured and purified. The migration of Lepr+/+ Sca-1+ progenitor cells in vitro was markedly induced by leptin. Western blotting and kinase assays revealed that leptin induced the activation of phosphorylated signal transducer and activator of transcription 3, phosphorylated extracellular signal–regulated kinases 1/2, pFAK (phosphorylated focal adhesion kinase), and Rac1 (ras-related C3 botulinum toxin substrate 1)/Cdc42 (cell division control protein 42 homolog). In a mouse femoral artery guidewire injury model, an increased expression of leptin in both injured vessels and serum was observed 24 hours post-surgery. RFP (red fluorescent protein)-Sca-1+ progenitor cells in Matrigel were applied to the adventitia of the injured femoral artery. RFP+ cells were observed in the intima 24 hours post-surgery, subsequently increasing neointimal lesions at 2 weeks when compared with the arteries without seeded cells. This increase was reduced by pre-treatment of Sca-1+ cells with a leptin antagonist. Guidewire injury could only induce minor neointima in Lepr−/− mice 2 weeks post-surgery. However, transplantation of Lepr+/+ Sca-1+ progenitor cells into the adventitial side of injured artery in Lepr−/− mice significantly enhanced neointimal formation. Conclusions— Upregulation of leptin levels in both the vessel wall and the circulation after vessel injury promoted the migration of Sca-1+ progenitor cells via leptin receptor–dependent signal transducer and activator of transcription 3- Rac1/Cdc42-ERK (extracellular signal–regulated kinase)-FAK pathways, which enhanced neointimal formation.
Collapse
Affiliation(s)
- Yao Xie
- From the Cardiovascular Division, King's College London BHF Centre, United Kingdom (Y.X., C.M.F.P., W.N.N., A.L.B., W.G., S.I.B., Z.Z., Y.H., Q.X.); and Department of Cardiology, The First Affiliated Hospital, School of Medicine, Zhejiang University, China (L.Z.)
| | - Claire M F Potter
- From the Cardiovascular Division, King's College London BHF Centre, United Kingdom (Y.X., C.M.F.P., W.N.N., A.L.B., W.G., S.I.B., Z.Z., Y.H., Q.X.); and Department of Cardiology, The First Affiliated Hospital, School of Medicine, Zhejiang University, China (L.Z.)
| | - Alexandra Le Bras
- From the Cardiovascular Division, King's College London BHF Centre, United Kingdom (Y.X., C.M.F.P., W.N.N., A.L.B., W.G., S.I.B., Z.Z., Y.H., Q.X.); and Department of Cardiology, The First Affiliated Hospital, School of Medicine, Zhejiang University, China (L.Z.)
| | - Witold N Nowak
- From the Cardiovascular Division, King's College London BHF Centre, United Kingdom (Y.X., C.M.F.P., W.N.N., A.L.B., W.G., S.I.B., Z.Z., Y.H., Q.X.); and Department of Cardiology, The First Affiliated Hospital, School of Medicine, Zhejiang University, China (L.Z.)
| | - Wenduo Gu
- From the Cardiovascular Division, King's College London BHF Centre, United Kingdom (Y.X., C.M.F.P., W.N.N., A.L.B., W.G., S.I.B., Z.Z., Y.H., Q.X.); and Department of Cardiology, The First Affiliated Hospital, School of Medicine, Zhejiang University, China (L.Z.)
| | - Shirin Issa Bhaloo
- From the Cardiovascular Division, King's College London BHF Centre, United Kingdom (Y.X., C.M.F.P., W.N.N., A.L.B., W.G., S.I.B., Z.Z., Y.H., Q.X.); and Department of Cardiology, The First Affiliated Hospital, School of Medicine, Zhejiang University, China (L.Z.)
| | - Zhongyi Zhang
- From the Cardiovascular Division, King's College London BHF Centre, United Kingdom (Y.X., C.M.F.P., W.N.N., A.L.B., W.G., S.I.B., Z.Z., Y.H., Q.X.); and Department of Cardiology, The First Affiliated Hospital, School of Medicine, Zhejiang University, China (L.Z.)
| | - Yanhua Hu
- From the Cardiovascular Division, King's College London BHF Centre, United Kingdom (Y.X., C.M.F.P., W.N.N., A.L.B., W.G., S.I.B., Z.Z., Y.H., Q.X.); and Department of Cardiology, The First Affiliated Hospital, School of Medicine, Zhejiang University, China (L.Z.)
| | - Li Zhang
- From the Cardiovascular Division, King's College London BHF Centre, United Kingdom (Y.X., C.M.F.P., W.N.N., A.L.B., W.G., S.I.B., Z.Z., Y.H., Q.X.); and Department of Cardiology, The First Affiliated Hospital, School of Medicine, Zhejiang University, China (L.Z.)
| | - Qingbo Xu
- From the Cardiovascular Division, King's College London BHF Centre, United Kingdom (Y.X., C.M.F.P., W.N.N., A.L.B., W.G., S.I.B., Z.Z., Y.H., Q.X.); and Department of Cardiology, The First Affiliated Hospital, School of Medicine, Zhejiang University, China (L.Z.).
| |
Collapse
|
28
|
Magnussen LV, Andersen PE, Diaz A, Ostojic J, Højlund K, Hougaard DM, Christensen AN, Nielsen TL, Andersen M. MR spectroscopy of hepatic fat and adiponectin and leptin levels during testosterone therapy in type 2 diabetes: a randomized, double-blinded, placebo-controlled trial. Eur J Endocrinol 2017; 177:157-168. [PMID: 28522646 DOI: 10.1530/eje-17-0071] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/26/2017] [Revised: 04/30/2017] [Accepted: 05/18/2017] [Indexed: 01/06/2023]
Abstract
BACKGROUND Men with type 2 diabetes mellitus (T2D) often have lowered testosterone levels and an increased risk of cardiovascular disease (CVD). Ectopic fat increases the risk of CVD, whereas subcutaneous gluteofemoral fat protects against CVD and has a beneficial adipokine-secreting profile. HYPOTHESIS Testosterone replacement therapy (TRT) may reduce the content of ectopic fat and improve the adipokine profile in men with T2D. DESIGN AND METHODS A randomized, double-blinded, placebo-controlled study in 39 men aged 50-70 years with T2D and bioavailable testosterone levels <7.3 nmol/L. Patients were randomized to TRT (n = 20) or placebo gel (n = 19) for 24 weeks. Thigh subcutaneous fat area (TFA, %fat of total thigh volume), subcutaneous abdominal adipose tissue (SAT, % fat of total abdominal volume) and visceral adipose tissue (VAT, % fat of total abdominal volume) were measured by magnetic resonance (MR) imaging. Hepatic fat content was estimated by single-voxel MR spectroscopy. Adiponectin and leptin levels were measured by in-house immunofluorometric assay. Coefficients (b) represent the placebo-controlled mean effect of intervention. RESULTS TFA (b = -3.3 percentage points (pp), P = 0.009), SAT (b = -3.0 pp, P = 0.006), levels of adiponectin (b = -0.4 mg/L, P = 0.045), leptin (b = -4.3 µg/mL, P < 0.001), leptin:adiponectin ratio (b = -0.53, P = 0.001) and HDL cholesterol (b = -0.11 mmol/L, P = 0.009) decreased during TRT compared with placebo. Hepatic fat content and VAT were unchanged. CONCLUSIONS The effects of TRT on cardiovascular risk markers were ambiguous. We observed potentially harmful changes in cardiovascular risk parameters, markedly reduced subcutaneous fat and unchanged ectopic fat during TRT and a reduction in adiponectin levels. On the other hand, the decrease in leptin and leptin:adiponectin ratio assessments could reflect an amelioration of the cardiovascular risk profile linked to hyperleptinaemia in ageing men with T2D.
Collapse
Affiliation(s)
| | - P E Andersen
- Departments of Radiology, Odense University Hospital, Odense, Denmark
- Clinical Institute, University of Southern Denmark, Odense, Denmark
| | - A Diaz
- Departments of Radiology, Odense University Hospital, Odense, Denmark
| | - J Ostojic
- Centre of Radiology, Clinical Centre of Vojvodina, Faculty of Medicine-University of Novi Sad, Novi Sad, Serbia
| | - K Højlund
- Departments of Endocrinology and Metabolism
- Section of Molecular Diabetes & Metabolism, Institute of Molecular Medicine, University of Southern Denmark, Odense, Denmark
| | - D M Hougaard
- Department for Congenital Disorders, Statens Serum Institut, Copenhagen, Denmark
| | - A N Christensen
- Department of Applied Mathematics and Computer Science, Technical University of Denmark, Lyngby, Denmark
| | | | - M Andersen
- Departments of Endocrinology and Metabolism
| |
Collapse
|
29
|
Hasin T, Iakobishvili Z, Weisz G. Associated Risk of Malignancy in Patients with Cardiovascular Disease: Evidence and Possible Mechanism. Am J Med 2017; 130:780-785. [PMID: 28344133 DOI: 10.1016/j.amjmed.2017.02.024] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/04/2017] [Revised: 02/10/2017] [Accepted: 02/13/2017] [Indexed: 12/31/2022]
Abstract
Cardiovascular disease and malignancy are leading causes of morbidity and mortality. Increased risk of malignancy was identified in patients with cardiovascular disease, including patients with heart failure, heart failure after myocardial infarction, patients undergoing cardiac intervention, and patients after a thrombotic event. Common risk factors and biological pathways can explain this association and are explored in this review. Further research is needed to establish the causes of malignancy in this population and direct possible intervention.
Collapse
Affiliation(s)
- Tal Hasin
- Department of Cardiology, Shaare Zedek Medical Center, Jerusalem, Israel.
| | - Zaza Iakobishvili
- Department of Cardiology, Rabin Medical Center, Petach Tiqwa, Israel
| | - Giora Weisz
- Department of Cardiology, Shaare Zedek Medical Center, Jerusalem, Israel
| |
Collapse
|
30
|
Lekva T, Roland MCP, Michelsen AE, Friis CM, Aukrust P, Bollerslev J, Henriksen T, Ueland T. Large Reduction in Adiponectin During Pregnancy Is Associated With Large-for-Gestational-Age Newborns. J Clin Endocrinol Metab 2017; 102:2552-2559. [PMID: 28460045 DOI: 10.1210/jc.2017-00289] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/30/2017] [Accepted: 04/25/2017] [Indexed: 01/05/2023]
Abstract
CONTEXT Fetuses exposed to an obese intrauterine environment are more likely to be born large-for-gestational age (LGA) and are at increased risk of obesity in childhood and cardiovascular disease and/or type 2 diabetes mellitus as adults, but which factors that influence the intrauterine environment is less clear. OBJECTIVE To investigate the association between circulating levels of leptin and adiponectin, measured multiple times during pregnancy, and birth weight and prevalence of LGA or small-for-gestational-age infants. The association between birth weight and messenger RNA (mRNA) expression of adiponectin receptors and genes involved in nutrient transport in the placenta was also investigated. DESIGN Population-based prospective cohort [substudy of the STORK study (STORe barn og Komplikasjoner, translated as Large Babies and Complications)] from 2001 to 2008. SETTING University hospital. Patients or other participants: 300 women. MAIN OUTCOME MEASURES Oral glucose tolerance test was performed twice along with adiponectin and leptin levels measured four times during pregnancy. RESULTS Circulating adiponectin was lower in mothers who gave birth to LGA offspring or had fetuses with high intrauterine abdominal circumference late in pregnancy. Adiponectin decreased most from early to late pregnancy in mothers who gave birth to LGA offspring, and the decrease was an independent predictor of birth weight. Adiponectin receptor 2 and system A amino acid transporter mRNA expression in placentas was negatively correlated with birth weight and was lower in placentas from LGA infants. CONCLUSIONS Our findings suggest that maternal adiponectin may be an important predictor of fetal growth and birth weight, independent of body mass index and insulin resistance.
Collapse
Affiliation(s)
- Tove Lekva
- Research Institute of Internal Medicine, Oslo University Hospital, Rikshospitalet, 0027 Oslo, Norway
| | - Marie Cecilie Paasche Roland
- Department of Obstetrics, Oslo University Hospital, Rikshospitalet, 0027 Oslo, Norway
- National Advisory Unit for Womens Health, Oslo University Hospital, Rikshospitalet, 0027 Oslo, Norway
| | - Annika E Michelsen
- Research Institute of Internal Medicine, Oslo University Hospital, Rikshospitalet, 0027 Oslo, Norway
- Faculty of Medicine, University of Oslo, 0027 Oslo, Norway
| | | | - Pål Aukrust
- Research Institute of Internal Medicine, Oslo University Hospital, Rikshospitalet, 0027 Oslo, Norway
- Faculty of Medicine, University of Oslo, 0027 Oslo, Norway
- Section of Clinical Immunology and Infectious Diseases, Oslo University Hospital, Rikshospitalet, 0027 Oslo, Norway
- K.G. Jebsen Inflammatory Research Center, University of Oslo, 0027 Oslo, Norway
- K. G. Jebsen Thrombosis Research and Expertise Center, University of Tromsø, 9037 Tromsø, Norway
| | - Jens Bollerslev
- Faculty of Medicine, University of Oslo, 0027 Oslo, Norway
- Section of Specialized Endocrinology, Department of Endocrinology, Oslo University Hospital, Rikshospitalet, 0027 Oslo, Norway
| | - Tore Henriksen
- Department of Obstetrics, Oslo University Hospital, Rikshospitalet, 0027 Oslo, Norway
- Faculty of Medicine, University of Oslo, 0027 Oslo, Norway
| | - Thor Ueland
- Research Institute of Internal Medicine, Oslo University Hospital, Rikshospitalet, 0027 Oslo, Norway
- Faculty of Medicine, University of Oslo, 0027 Oslo, Norway
- K. G. Jebsen Thrombosis Research and Expertise Center, University of Tromsø, 9037 Tromsø, Norway
| |
Collapse
|
31
|
Lekva T, Michelsen AE, Aukrust P, Henriksen T, Bollerslev J, Ueland T. Leptin and adiponectin as predictors of cardiovascular risk after gestational diabetes mellitus. Cardiovasc Diabetol 2017; 16:5. [PMID: 28068986 PMCID: PMC5223461 DOI: 10.1186/s12933-016-0492-4] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/06/2016] [Accepted: 12/26/2016] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Gestational diabetes mellitus (GDM) is a significant risk factor for cardiovascular disease (CVD) in later life, but the mechanism remains unclear. Adipokine imbalance in the presence of metabolic dysfunction may be a key event in promoting CVD. The aim of the study was to examine the relationships between GDM, cardiovascular risk, and plasma adiponectin, leptin and the leptin/adiponectin (L/A) ratio in pregnancy and at 5 years after the index pregnancy. METHODS This population-based prospective cohort included 300 women who had an oral glucose tolerance test (OGTT) during pregnancy. Five years later, the OGTT was repeated along with dual-energy X-ray absorptiometry, lipid analysis, and pulse wave velocity analysis. Fasting adiponectin and leptin levels were measured four times during pregnancy and at follow-up. RESULTS We found the L/A ratio higher in GDM women both during pregnancy and follow-up compared to non-GDM women. A high L/A ratio during pregnancy was associated with CV risk based on lipid ratios at follow-up, especially the TG/HDL-C ratio. Further, interaction analysis indicated that an increase in the L/A ratio of 1 unit was associated with a higher CV risk in GDM compared to normal pregnancy. Finally, low adiponectin levels independently predicted increased lipid ratios at follow-up. CONCLUSIONS Taken together, our findings suggest that high L/A ratio in pregnancy and in particularly in those with GDM are associated with an unfavorable CVD risk profile during follow-up. Future studies should investigate if a dysregulated leptin and adiponectin profile during pregnancy is associated with atherosclerotic disease during long-term follow-up.
Collapse
Affiliation(s)
- Tove Lekva
- Research Institute of Internal Medicine, Oslo University Hospital, Rikshospitalet, Oslo, Norway.
| | - Annika Elisabet Michelsen
- Research Institute of Internal Medicine, Oslo University Hospital, Rikshospitalet, Oslo, Norway.,Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Pål Aukrust
- Research Institute of Internal Medicine, Oslo University Hospital, Rikshospitalet, Oslo, Norway.,Section of Clinical Immunology and Infectious Diseases, Oslo University Hospital, Rikshospitalet, Oslo, Norway.,Faculty of Medicine, University of Oslo, Oslo, Norway.,K. G. Jebsen Thrombosis Research and Expertise Center, University of Tromsø, Tromsø, Norway
| | - Tore Henriksen
- Department of Obstetrics, Oslo University Hospital, Rikshospitalet, Oslo, Norway.,Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Jens Bollerslev
- Section of Specialized Endocrinology, Department of Endocrinology, Oslo University Hospital, Rikshospitalet, Oslo, Norway.,Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Thor Ueland
- Research Institute of Internal Medicine, Oslo University Hospital, Rikshospitalet, Oslo, Norway.,Faculty of Medicine, University of Oslo, Oslo, Norway.,K. G. Jebsen Thrombosis Research and Expertise Center, University of Tromsø, Tromsø, Norway
| |
Collapse
|
32
|
Bi R, Ding F, He Y, Jiang L, Jiang Z, Mei J, Liu H. miR-503 inhibits platelet-derived growth factor-induced human aortic vascular smooth muscle cell proliferation and migration through targeting the insulin receptor. Biomed Pharmacother 2016; 84:1711-1716. [PMID: 27829550 DOI: 10.1016/j.biopha.2016.10.081] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2016] [Revised: 10/19/2016] [Accepted: 10/27/2016] [Indexed: 12/14/2022] Open
Abstract
Abnormal proliferation and migration of vascular smooth muscle cells (VSMC) is a common feature of disease progression in atherosclerosis. Here, we investigated the potential role of miR-503 in platelet-derived growth factor (PDGF)-induced proliferation and migration of human aortic smooth muscle cells and the underlying mechanisms of action. miR-503 expression was significantly downregulated in a dose- and time-dependent manner following PDGF treatment. Introduction of miR-503 mimics into cultured SMCs significantly attenuated cell proliferation and migration induced by PDGF. Bioinformatics analyses revealed that the insulin receptor (INSR) is a target candidate of miR-503. miR-503 suppressed luciferase activity driven by a vector containing the 3'-untranslated region of INSR in a sequence-specific manner. Downregulation of INSR appeared critical for miR-503-mediated inhibitory effects on PDGF-induced cell proliferation and migration in human aortic SMCs. Based on the collective data, we suggest a novel role of miR-503 as a regulator of VSMC proliferation and migration through modulating INSR.
Collapse
Affiliation(s)
- Rui Bi
- Department of Cardiothoracic Surgery, Xinhua Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, 200092, China
| | - Fangbao Ding
- Department of Cardiothoracic Surgery, Xinhua Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, 200092, China
| | - Yi He
- Department of Cardiothoracic Surgery, Xinhua Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, 200092, China
| | - Lianyong Jiang
- Department of Cardiothoracic Surgery, Xinhua Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, 200092, China
| | - Zhaolei Jiang
- Department of Cardiothoracic Surgery, Xinhua Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, 200092, China
| | - Ju Mei
- Department of Cardiothoracic Surgery, Xinhua Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, 200092, China.
| | - Hao Liu
- Department of Cardiothoracic Surgery, Xinhua Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, 200092, China.
| |
Collapse
|
33
|
Ding N, Liu B, Song J, Bao S, Zhen J, Lv Z, Wang R. Leptin promotes endothelial dysfunction in chronic kidney disease through AKT/GSK3β and β-catenin signals. Biochem Biophys Res Commun 2016; 480:544-551. [PMID: 27789284 DOI: 10.1016/j.bbrc.2016.10.079] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2016] [Accepted: 10/21/2016] [Indexed: 12/01/2022]
Abstract
Endothelial dysfunction (ED) is a well-recognized instigator of cardiovascular diseases and develops in chronic kidney disease (CKD) with high rate. Recent studies have implicated that leptin is associated with endothelial dysfunction. We investigated the relationship between leptin and markers of ED in CKD patients and how leptin contributed to endothelial damage. 140 CKD patients and 140 healthy subjects were studied. Serum leptin levels were significantly higher in CKD than in controls and displayed significantly positive association with the increase levels of sICAM-1 and sVCAM-1 but negative correlation with flow-mediated dilatation (FMD) reduction in patients. Our in vitro study demonstrated that leptin induced overexpression of ICAM-1 and VCAM-1, led to f-actin reorganization and vinculin assembly, increased endothelial monolayer permeability for FITC-dextran, and accelerated endothelial cell migration; these changes were markedly reversed when the cells were transfected with AKT or β-catenin shRNA vectors. Notably, high leptin resulted in hyper-phosphorylation of AKT and GSK3β, along with nuclear accumulation of β-catenin. In conclusion, serum leptin was elevated in CKD patients and it might contribute to endothelial dysfunction by disarrangement of f-actin cytoskeleton via a mechanism involving the AKT/GSK3β and β-catenin pathway.
Collapse
Affiliation(s)
- Nannan Ding
- Department of Nephrology, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, China
| | - Bing Liu
- Department of Nephrology, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, China
| | - Jiaguang Song
- Department of Ultrasonography, Shandong Provincial Hospital Affiliated to Shandong University, China
| | - Shougang Bao
- Department of Ultrasonography, Shandong Medical Imaging Research Institute, Shandong University, China
| | - Junhui Zhen
- Department of Pathology, Medical School of Shandong University, Jinan, China
| | - Zhimei Lv
- Department of Nephrology, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, China.
| | - Rong Wang
- Department of Nephrology, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, China.
| |
Collapse
|
34
|
Manna P, Jain SK. Obesity, Oxidative Stress, Adipose Tissue Dysfunction, and the Associated Health Risks: Causes and Therapeutic Strategies. Metab Syndr Relat Disord 2016; 13:423-44. [PMID: 26569333 DOI: 10.1089/met.2015.0095] [Citation(s) in RCA: 671] [Impact Index Per Article: 74.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Obesity is gaining acceptance as a serious primary health burden that impairs the quality of life because of its associated complications, including diabetes, cardiovascular diseases, cancer, asthma, sleep disorders, hepatic dysfunction, renal dysfunction, and infertility. It is a complex metabolic disorder with a multifactorial origin. Growing evidence suggests that oxidative stress plays a role as the critical factor linking obesity with its associated complications. Obesity per se can induce systemic oxidative stress through various biochemical mechanisms, such as superoxide generation from NADPH oxidases, oxidative phosphorylation, glyceraldehyde auto-oxidation, protein kinase C activation, and polyol and hexosamine pathways. Other factors that also contribute to oxidative stress in obesity include hyperleptinemia, low antioxidant defense, chronic inflammation, and postprandial reactive oxygen species generation. In addition, recent studies suggest that adipose tissue plays a critical role in regulating the pathophysiological mechanisms of obesity and its related co-morbidities. To establish an adequate platform for the prevention of obesity and its associated health risks, understanding the factors that contribute to the cause of obesity is necessary. The most current list of obesity determinants includes genetic factors, dietary intake, physical activity, environmental and socioeconomic factors, eating disorders, and societal influences. On the basis of the currently identified predominant determinants of obesity, a broad range of strategies have been recommended to reduce the prevalence of obesity, such as regular physical activity, ad libitum food intake limiting to certain micronutrients, increased dietary intake of fruits and vegetables, and meal replacements. This review aims to highlight recent findings regarding the role of oxidative stress in the pathogenesis of obesity and its associated risk factors, the role of dysfunctional adipose tissue in development of these risk factors, and potential strategies to regulate body weight loss/gain for better health benefits.
Collapse
Affiliation(s)
- Prasenjit Manna
- Department of Pediatrics, LSU Health Sciences Center , Shreveport, Louisiana
| | - Sushil K Jain
- Department of Pediatrics, LSU Health Sciences Center , Shreveport, Louisiana
| |
Collapse
|
35
|
Dinh Cat AN, Friederich-Persson M, White A, Touyz RM. Adipocytes, aldosterone and obesity-related hypertension. J Mol Endocrinol 2016; 57:F7-F21. [PMID: 27357931 DOI: 10.1530/jme-16-0025] [Citation(s) in RCA: 49] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/29/2016] [Accepted: 05/09/2016] [Indexed: 12/15/2022]
Abstract
Understanding the mechanisms linking obesity with hypertension is important in the current obesity epidemic as it may improve therapeutic interventions. Plasma aldosterone levels are positively correlated with body mass index and weight loss in obese patients is reported to be accompanied by decreased aldosterone levels. This suggests a relationship between adipose tissue and the production/secretion of aldosterone. Aldosterone is synthesized principally by the adrenal glands, but its production may be regulated by many factors, including factors secreted by adipocytes. In addition, studies have reported local synthesis of aldosterone in extra-adrenal tissues, including adipose tissue. Experimental studies have highlighted a role for adipocyte-secreted aldosterone in the pathogenesis of obesity-related cardiovascular complications via the mineralocorticoid receptor. This review focuses on how aldosterone secretion may be influenced by adipose tissue and the importance of these mechanisms in the context of obesity-related hypertension.
Collapse
Affiliation(s)
- Aurelie Nguyen Dinh Cat
- Institute of Cardiovascular and Medical SciencesBHF Glasgow Cardiovascular Research Centre, University of Glasgow, Glasgow, UK
| | - Malou Friederich-Persson
- Institute of Cardiovascular and Medical SciencesBHF Glasgow Cardiovascular Research Centre, University of Glasgow, Glasgow, UK
| | - Anna White
- Institute of Cardiovascular and Medical SciencesBHF Glasgow Cardiovascular Research Centre, University of Glasgow, Glasgow, UK
| | - Rhian M Touyz
- Institute of Cardiovascular and Medical SciencesBHF Glasgow Cardiovascular Research Centre, University of Glasgow, Glasgow, UK
| |
Collapse
|
36
|
Abstract
Cardiovascular disease (CVD) and cancer are the 2 leading causes of death worldwide. Although commonly thought of as 2 separate disease entities, CVD and cancer possess various similarities and possible interactions, including a number of similar risk factors (eg, obesity, diabetes mellitus), suggesting a shared biology for which there is emerging evidence. Although chronic inflammation is an indispensable feature of the pathogenesis and progression of both CVD and cancer, additional mechanisms can be found at their intersection. Therapeutic advances, despite improving longevity, have increased the overlap between these diseases, with millions of cancer survivors now at risk of developing CVD. Cardiac risk factors have a major impact on subsequent treatment-related cardiotoxicity. In this review, we explore the risk factors common to both CVD and cancer, highlighting the major epidemiological studies and potential biological mechanisms that account for them.
Collapse
Affiliation(s)
- Ryan J Koene
- From Department of Medicine, Division of Cardiovascular Medicine, University of Minnesota, Minneapolis (R.J.K., S.H.K.); Department of Epidemiology and Community Health, University of Minnesota, Minneapolis (A.E.P.); Department of Medicine, Division of Oncology, University of Minnesota, Minneapolis (A.B.); and Masonic Cancer Center, University of Minnesota, Minneapolis (A.E.P.).
| | - Anna E Prizment
- From Department of Medicine, Division of Cardiovascular Medicine, University of Minnesota, Minneapolis (R.J.K., S.H.K.); Department of Epidemiology and Community Health, University of Minnesota, Minneapolis (A.E.P.); Department of Medicine, Division of Oncology, University of Minnesota, Minneapolis (A.B.); and Masonic Cancer Center, University of Minnesota, Minneapolis (A.E.P.)
| | - Anne Blaes
- From Department of Medicine, Division of Cardiovascular Medicine, University of Minnesota, Minneapolis (R.J.K., S.H.K.); Department of Epidemiology and Community Health, University of Minnesota, Minneapolis (A.E.P.); Department of Medicine, Division of Oncology, University of Minnesota, Minneapolis (A.B.); and Masonic Cancer Center, University of Minnesota, Minneapolis (A.E.P.)
| | - Suma H Konety
- From Department of Medicine, Division of Cardiovascular Medicine, University of Minnesota, Minneapolis (R.J.K., S.H.K.); Department of Epidemiology and Community Health, University of Minnesota, Minneapolis (A.E.P.); Department of Medicine, Division of Oncology, University of Minnesota, Minneapolis (A.B.); and Masonic Cancer Center, University of Minnesota, Minneapolis (A.E.P.)
| |
Collapse
|
37
|
Ilkhanizadeh B, Shirpoor A, Khadem Ansari MH, Nemati S, Rasmi Y. Protective Effects of Ginger (Zingiber officinale) Extract against Diabetes-Induced Heart Abnormality in Rats. Diabetes Metab J 2016; 40:46-53. [PMID: 26912155 PMCID: PMC4768050 DOI: 10.4093/dmj.2016.40.1.46] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/02/2015] [Accepted: 10/12/2015] [Indexed: 11/14/2022] Open
Abstract
BACKGROUND Diabetic cardiomyopathy is an important causal factor in morbidity and mortality among diabetic patients, and currently, no effective means are available to reverse its pathological progress. The purpose of the present study was to investigate the effect of ginger extract on apolipoproteins (apo) A and B, hyperhomocysteinemia, cathepsin G and leptin changes, as well as cardiac fibrosis and heart muscle cell proliferation under hyperglycemic conditions in vivo. METHODS Twenty-four male Wistar rats were divided into three groups, namely: control, non-treated diabetic, and ginger extract-treated diabetic groups. The ginger extract-treated diabetic group received a 50 mg daily dose of ginger extract intragastrically for 6 weeks. RESULTS The results revealed concurrent significant increases in plasma C-reactive protein (CRP), homocysteine (Hcy), cathepsin G and apoB levels and decreases in apoA and leptin levels in the non-treated diabetic group compared to the control group. Moreover, heart structural changes, including fibrosis and heart muscle cell proliferation, were observed in non-treated diabetic rats compared to the control rats. Significant amelioration of changes in the heart structure together with restoration of the elevated levels of Hcy and CRP, leptin, cathepsin G, and apoA and B were found in the ginger extract-treated diabetic group compared to the non-treated diabetic group. CONCLUSION The findings indicated that ginger extract significantly reduces heart structural abnormalities in diabetic rats and that these effects might be associated with improvements in serum apo, leptin, cathepsin G, and Hcy levels and with the antioxidant properties of ginger extract.
Collapse
Affiliation(s)
- Behrouz Ilkhanizadeh
- Department of Pathology, Urmia University of Medical Sciences Faculty of Medicine, Urmia, Iran
| | - Alireza Shirpoor
- Department of Physiology, Urmia University of Medical Sciences Faculty of Medicine, Urmia, Iran.
| | | | - Samira Nemati
- Department of Physiology, Urmia University of Medical Sciences Faculty of Medicine, Urmia, Iran
| | - Yusef Rasmi
- Department of Biochemistry, Urmia University of Medical Sciences Faculty of Medicine, Urmia, Iran
| |
Collapse
|
38
|
Turner RT, Dube M, Branscum AJ, Wong CP, Olson DA, Zhong X, Kweh MF, Larkin IV, Wronski TJ, Rosen CJ, Kalra SP, Iwaniec UT. Hypothalamic leptin gene therapy reduces body weight without accelerating age-related bone loss. J Endocrinol 2015; 227:129-41. [PMID: 26487675 PMCID: PMC4917201 DOI: 10.1530/joe-15-0280] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 09/14/2015] [Indexed: 02/04/2023]
Abstract
Excessive weight gain in adults is associated with a variety of negative health outcomes. Unfortunately, dieting, exercise, and pharmacological interventions have had limited long-term success in weight control and can result in detrimental side effects, including accelerating age-related cancellous bone loss. We investigated the efficacy of using hypothalamic leptin gene therapy as an alternative method for reducing weight in skeletally-mature (9 months old) female rats and determined the impact of leptin-induced weight loss on bone mass, density, and microarchitecture, and serum biomarkers of bone turnover (CTx and osteocalcin). Rats were implanted with cannulae in the 3rd ventricle of the hypothalamus and injected with either recombinant adeno-associated virus encoding the gene for rat leptin (rAAV-Leptin, n=7) or a control vector encoding green fluorescent protein (rAAV-GFP, n=10) and sacrificed 18 weeks later. A baseline control group (n=7) was sacrificed at vector administration. rAAV-Leptin-treated rats lost weight (-4±2%) while rAAV-GFP-treated rats gained weight (14±2%) during the study. At study termination, rAAV-Leptin-treated rats weighed 17% less than rAAV-GFP-treated rats and had lower abdominal white adipose tissue weight (-80%), serum leptin (-77%), and serum IGF1 (-34%). Cancellous bone volume fraction in distal femur metaphysis and epiphysis, and in lumbar vertebra tended to be lower (P<0.1) in rAAV-GFP-treated rats (13.5 months old) compared to baseline control rats (9 months old). Significant differences in cancellous bone or biomarkers of bone turnover were not detected between rAAV-Leptin and rAAV-GFP rats. In summary, rAAV-Leptin-treated rats maintained a lower body weight compared to baseline and rAAV-GFP-treated rats with minimal effects on bone mass, density, microarchitecture, or biochemical markers of bone turnover.
Collapse
Affiliation(s)
- Russell T Turner
- Skeletal Biology LaboratorySchool of Biological and Population Health Sciences, Oregon State University, Corvallis, Oregon 97331, USACenter for Healthy Aging ResearchOregon State University, Corvallis, Oregon, USADepartment of NeuroscienceMcKnight Brain Institute, University of Florida, Gainesville, Florida, USABiostatisticsSchool of Biological and Population Health Sciences, Oregon State University, Corvallis, Oregon, USADepartment of Physiological SciencesUniversity of Florida, Gainesville, Florida, USADepartment of Large Animal Clinical SciencesUniversity of Florida, Gainesville, Florida, USAMaine Medical Center Research InstituteScarborough, Maine, USA Skeletal Biology LaboratorySchool of Biological and Population Health Sciences, Oregon State University, Corvallis, Oregon 97331, USACenter for Healthy Aging ResearchOregon State University, Corvallis, Oregon, USADepartment of NeuroscienceMcKnight Brain Institute, University of Florida, Gainesville, Florida, USABiostatisticsSchool of Biological and Population Health Sciences, Oregon State University, Corvallis, Oregon, USADepartment of Physiological SciencesUniversity of Florida, Gainesville, Florida, USADepartment of Large Animal Clinical SciencesUniversity of Florida, Gainesville, Florida, USAMaine Medical Center Research InstituteScarborough, Maine, USA
| | - Michael Dube
- Skeletal Biology LaboratorySchool of Biological and Population Health Sciences, Oregon State University, Corvallis, Oregon 97331, USACenter for Healthy Aging ResearchOregon State University, Corvallis, Oregon, USADepartment of NeuroscienceMcKnight Brain Institute, University of Florida, Gainesville, Florida, USABiostatisticsSchool of Biological and Population Health Sciences, Oregon State University, Corvallis, Oregon, USADepartment of Physiological SciencesUniversity of Florida, Gainesville, Florida, USADepartment of Large Animal Clinical SciencesUniversity of Florida, Gainesville, Florida, USAMaine Medical Center Research InstituteScarborough, Maine, USA
| | - Adam J Branscum
- Skeletal Biology LaboratorySchool of Biological and Population Health Sciences, Oregon State University, Corvallis, Oregon 97331, USACenter for Healthy Aging ResearchOregon State University, Corvallis, Oregon, USADepartment of NeuroscienceMcKnight Brain Institute, University of Florida, Gainesville, Florida, USABiostatisticsSchool of Biological and Population Health Sciences, Oregon State University, Corvallis, Oregon, USADepartment of Physiological SciencesUniversity of Florida, Gainesville, Florida, USADepartment of Large Animal Clinical SciencesUniversity of Florida, Gainesville, Florida, USAMaine Medical Center Research InstituteScarborough, Maine, USA
| | - Carmen P Wong
- Skeletal Biology LaboratorySchool of Biological and Population Health Sciences, Oregon State University, Corvallis, Oregon 97331, USACenter for Healthy Aging ResearchOregon State University, Corvallis, Oregon, USADepartment of NeuroscienceMcKnight Brain Institute, University of Florida, Gainesville, Florida, USABiostatisticsSchool of Biological and Population Health Sciences, Oregon State University, Corvallis, Oregon, USADepartment of Physiological SciencesUniversity of Florida, Gainesville, Florida, USADepartment of Large Animal Clinical SciencesUniversity of Florida, Gainesville, Florida, USAMaine Medical Center Research InstituteScarborough, Maine, USA
| | - Dawn A Olson
- Skeletal Biology LaboratorySchool of Biological and Population Health Sciences, Oregon State University, Corvallis, Oregon 97331, USACenter for Healthy Aging ResearchOregon State University, Corvallis, Oregon, USADepartment of NeuroscienceMcKnight Brain Institute, University of Florida, Gainesville, Florida, USABiostatisticsSchool of Biological and Population Health Sciences, Oregon State University, Corvallis, Oregon, USADepartment of Physiological SciencesUniversity of Florida, Gainesville, Florida, USADepartment of Large Animal Clinical SciencesUniversity of Florida, Gainesville, Florida, USAMaine Medical Center Research InstituteScarborough, Maine, USA
| | - Xiaoying Zhong
- Skeletal Biology LaboratorySchool of Biological and Population Health Sciences, Oregon State University, Corvallis, Oregon 97331, USACenter for Healthy Aging ResearchOregon State University, Corvallis, Oregon, USADepartment of NeuroscienceMcKnight Brain Institute, University of Florida, Gainesville, Florida, USABiostatisticsSchool of Biological and Population Health Sciences, Oregon State University, Corvallis, Oregon, USADepartment of Physiological SciencesUniversity of Florida, Gainesville, Florida, USADepartment of Large Animal Clinical SciencesUniversity of Florida, Gainesville, Florida, USAMaine Medical Center Research InstituteScarborough, Maine, USA
| | - Mercedes F Kweh
- Skeletal Biology LaboratorySchool of Biological and Population Health Sciences, Oregon State University, Corvallis, Oregon 97331, USACenter for Healthy Aging ResearchOregon State University, Corvallis, Oregon, USADepartment of NeuroscienceMcKnight Brain Institute, University of Florida, Gainesville, Florida, USABiostatisticsSchool of Biological and Population Health Sciences, Oregon State University, Corvallis, Oregon, USADepartment of Physiological SciencesUniversity of Florida, Gainesville, Florida, USADepartment of Large Animal Clinical SciencesUniversity of Florida, Gainesville, Florida, USAMaine Medical Center Research InstituteScarborough, Maine, USA
| | - Iske V Larkin
- Skeletal Biology LaboratorySchool of Biological and Population Health Sciences, Oregon State University, Corvallis, Oregon 97331, USACenter for Healthy Aging ResearchOregon State University, Corvallis, Oregon, USADepartment of NeuroscienceMcKnight Brain Institute, University of Florida, Gainesville, Florida, USABiostatisticsSchool of Biological and Population Health Sciences, Oregon State University, Corvallis, Oregon, USADepartment of Physiological SciencesUniversity of Florida, Gainesville, Florida, USADepartment of Large Animal Clinical SciencesUniversity of Florida, Gainesville, Florida, USAMaine Medical Center Research InstituteScarborough, Maine, USA
| | - Thomas J Wronski
- Skeletal Biology LaboratorySchool of Biological and Population Health Sciences, Oregon State University, Corvallis, Oregon 97331, USACenter for Healthy Aging ResearchOregon State University, Corvallis, Oregon, USADepartment of NeuroscienceMcKnight Brain Institute, University of Florida, Gainesville, Florida, USABiostatisticsSchool of Biological and Population Health Sciences, Oregon State University, Corvallis, Oregon, USADepartment of Physiological SciencesUniversity of Florida, Gainesville, Florida, USADepartment of Large Animal Clinical SciencesUniversity of Florida, Gainesville, Florida, USAMaine Medical Center Research InstituteScarborough, Maine, USA
| | - Clifford J Rosen
- Skeletal Biology LaboratorySchool of Biological and Population Health Sciences, Oregon State University, Corvallis, Oregon 97331, USACenter for Healthy Aging ResearchOregon State University, Corvallis, Oregon, USADepartment of NeuroscienceMcKnight Brain Institute, University of Florida, Gainesville, Florida, USABiostatisticsSchool of Biological and Population Health Sciences, Oregon State University, Corvallis, Oregon, USADepartment of Physiological SciencesUniversity of Florida, Gainesville, Florida, USADepartment of Large Animal Clinical SciencesUniversity of Florida, Gainesville, Florida, USAMaine Medical Center Research InstituteScarborough, Maine, USA
| | - Satya P Kalra
- Skeletal Biology LaboratorySchool of Biological and Population Health Sciences, Oregon State University, Corvallis, Oregon 97331, USACenter for Healthy Aging ResearchOregon State University, Corvallis, Oregon, USADepartment of NeuroscienceMcKnight Brain Institute, University of Florida, Gainesville, Florida, USABiostatisticsSchool of Biological and Population Health Sciences, Oregon State University, Corvallis, Oregon, USADepartment of Physiological SciencesUniversity of Florida, Gainesville, Florida, USADepartment of Large Animal Clinical SciencesUniversity of Florida, Gainesville, Florida, USAMaine Medical Center Research InstituteScarborough, Maine, USA
| | - Urszula T Iwaniec
- Skeletal Biology LaboratorySchool of Biological and Population Health Sciences, Oregon State University, Corvallis, Oregon 97331, USACenter for Healthy Aging ResearchOregon State University, Corvallis, Oregon, USADepartment of NeuroscienceMcKnight Brain Institute, University of Florida, Gainesville, Florida, USABiostatisticsSchool of Biological and Population Health Sciences, Oregon State University, Corvallis, Oregon, USADepartment of Physiological SciencesUniversity of Florida, Gainesville, Florida, USADepartment of Large Animal Clinical SciencesUniversity of Florida, Gainesville, Florida, USAMaine Medical Center Research InstituteScarborough, Maine, USA Skeletal Biology LaboratorySchool of Biological and Population Health Sciences, Oregon State University, Corvallis, Oregon 97331, USACenter for Healthy Aging ResearchOregon State University, Corvallis, Oregon, USADepartment of NeuroscienceMcKnight Brain Institute, University of Florida, Gainesville, Florida, USABiostatisticsSchool of Biological and Population Health Sciences, Oregon State University, Corvallis, Oregon, USADepartment of Physiological SciencesUniversity of Florida, Gainesville, Florida, USADepartment of Large Animal Clinical SciencesUniversity of Florida, Gainesville, Florida, USAMaine Medical Center Research InstituteScarborough, Maine, USA
| |
Collapse
|
39
|
Xie B, Zhang C, Kang K, Jiang S. miR-599 Inhibits Vascular Smooth Muscle Cells Proliferation and Migration by Targeting TGFB2. PLoS One 2015; 10:e0141512. [PMID: 26551255 PMCID: PMC4638351 DOI: 10.1371/journal.pone.0141512] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2015] [Accepted: 10/08/2015] [Indexed: 02/02/2023] Open
Abstract
Aberrant proliferation and migration of vascular smooth muscle cells (VSMCs) play a crucial role in the pathogenesis of cardiovascular diseases including coronary heart disease, restenosis and atherosclerosis. MicroRNAs are a class of small, non-coding and endogenous RNAs that play critical roles in VSMCs function. In this study, we showed that PDGF-bb, as a stimulant, promoted VSMCs proliferation and suppressed the expression of miR-599. Moreover, overexpression of miR-599 inhibited VSMCs proliferation and also suppressed the PCNA and ki-67 expression. In addition, we demonstrated that ectopic expression of miR-599 repressed the VSMCs migration. We also showed that miR-599 inhibited type I collagen, type V collagen and proteoglycan expression. Furthermore, we identified TGFb2 as a direct target gene of miR-599 in VSMCs. Overexpression of TGFb2 reversed miR-599-induced inhibition of VSMCs proliferation and type I collagen, type V collagen and proteoglycan expression. In conclusion, our findings suggest miR-599 plays a crucial role in controlling VSMCs proliferation and matrix gene expression by regulating TGFb2 expression.
Collapse
Affiliation(s)
- Baodong Xie
- Department of Cardiovascular Surgery, the Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Chunfeng Zhang
- Department of Cardiovascular Surgery, the Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Kai Kang
- Department of Cardiovascular Surgery, the Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Shulin Jiang
- Department of Cardiovascular Surgery, the Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| |
Collapse
|
40
|
Chen XX, Yang T. Roles of leptin in bone metabolism and bone diseases. J Bone Miner Metab 2015; 33:474-85. [PMID: 25777984 DOI: 10.1007/s00774-014-0569-7] [Citation(s) in RCA: 65] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/24/2013] [Accepted: 01/16/2014] [Indexed: 02/05/2023]
Abstract
Adipose tissue has been more accepted as an active contributor to whole body homeostasis, rather than just a fat depot, since leptin, a 16 kDa protein, was discovered as the product of the obese gene in 1994. With more and more studies conducted on this hormone, it has been shown that there is a close relationship between adipose tissue and bone, which have important effects on each other. Bone is the source of many hormones, such as osteocalcin, that can affect energy metabolism and then the anabolism or catabolism of fat tissue. In contrast, the adipose tissue synthesizes and releases a series of adipokines, which are involved in bone metabolism through direct or indirect effects on bone formation and resorption. Interestingly, leptin, one of the most important cytokines derived from fat tissue, seems to account for the largest part of effects on bone, through direct or indirect involvement in bone remodeling and by playing a significant role in many bone diseases, such as osteoporosis, osteoarthritis, rheumatic arthritis, bone tumors and even fractures. In this review, we will discuss the progress in leptin research, particularly focusing on the roles of leptin in bone diseases.
Collapse
Affiliation(s)
- Xu Xu Chen
- Department of Orthopedic Surgery, West China Hospital, Sichuan University, 37# Guo Xue Xiang, Chengdu, 610041, Sichuan, People's Republic of China
| | | |
Collapse
|
41
|
Lee JH, Yoo JY, You YA, Kwon WS, Lee SM, Pang MG, Kim YJ. Proteomic analysis of fetal programming-related obesity markers. Proteomics 2015; 15:2669-77. [PMID: 25886259 DOI: 10.1002/pmic.201400359] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2014] [Revised: 02/16/2015] [Accepted: 04/14/2015] [Indexed: 12/14/2022]
Abstract
The objectives of this study were to analyze fetal programming in rat brain using proteomic analysis and to identify fetal programming-related obesity markers. Sprague-Dawley rats were divided into four feeding groups: (i) the Ad Libitum (AdLib)/AdLib group was given a normal diet during pregnancy and the lactation period; (ii) the AdLib/maternal food restriction group (FR) was subjected to 50% FR during the lactation period; (iii) the FR/AdLib group was subjected to 50% FR during pregnancy; and (iv) the FR/FR group was subjected to 50% FR during pregnancy and the lactation period. Offspring from each group were sacrificed at 3 weeks of age and whole brains were dissected. To obtain a maximum number of protein markers related to obesity, 2DE and Pathway Studio bioinformatics analysis were performed. The identities of the markers among the selected and candidate proteins were confirmed by Western blotting and immunohistochemistry. Proteomic and bioinformatics analyses revealed that expression of ubiquitin carboxy-terminal hydrolase L1 (UCHL1) and Secernin 1 (SCRN1) were significantly different in the FR/AdLib group compared with the AdLib/AdLib group for both male and female offspring. These findings suggest that UCHL1 and SCRN1 may be used as fetal programming-related obesity markers.
Collapse
Affiliation(s)
- Ji Hye Lee
- Department of Obstetrics and Gynecology, Ewha Womans University, Seoul, South Korea
| | - Jae Young Yoo
- Department of Obstetrics and Gynecology, Ewha Womans University, Seoul, South Korea
| | - Young-Ah You
- Department of Obstetrics and Gynecology, Ewha Womans University, Seoul, South Korea
| | - Woo-Sung Kwon
- Department of Animal Science and Technology, Chung-Ang University, Anseong, Gyeonggi-Do 456-756, South Korea
| | - Sang Mi Lee
- Department of Obstetrics and Gynecology, Ewha Womans University, Seoul, South Korea
| | - Myung-Geol Pang
- Department of Animal Science and Technology, Chung-Ang University, Anseong, Gyeonggi-Do 456-756, South Korea
| | - Young Ju Kim
- Department of Obstetrics and Gynecology, Ewha Womans University, Seoul, South Korea
| |
Collapse
|
42
|
Tahergorabi Z, Khazaei M. Leptin and its cardiovascular effects: Focus on angiogenesis. Adv Biomed Res 2015; 4:79. [PMID: 26015905 PMCID: PMC4434486 DOI: 10.4103/2277-9175.156526] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2014] [Accepted: 07/20/2014] [Indexed: 12/24/2022] Open
Abstract
Leptin is an endocrine hormone synthesized by adipocytes. It plays a key role in the energy homeostasis in central and peripheral tissues and has additional roles are attributed to it, such as the regulation of reproduction, immune function, bone homeostasis, and angiogenesis. The plasma concentration of leptin significantly increases in obese individuals. In the present review, we give an introduction concerning leptin, its receptors, signaling pathways, and its effect on cardiovascular system, especially on angiogenesis.
Collapse
Affiliation(s)
- Zoya Tahergorabi
- Department of Physiology and Pharmacology, Birjand University of Medical Sciences, Birjand, Iran
| | - Majid Khazaei
- Department of Physiology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
43
|
Jiang S, Song K, Feng S, Shi YB. Association between serum leptin levels and peritoneal dialysis: A meta-analysis. Exp Ther Med 2015; 10:300-308. [PMID: 26170953 DOI: 10.3892/etm.2015.2441] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2014] [Accepted: 02/13/2015] [Indexed: 01/11/2023] Open
Abstract
There is limited information available with regard to the association between serum leptin levels, or other adipokines, and serum lipid levels and insulin sensitivity in patients undergoing peritoneal dialysis (PD). Thus, the aim of the present study was to perform a meta-analysis investigating this association. Potential relevant studies were identified through searching the following databases: MEDLINE, Science Citation Index, Cochrane Library, PubMed, Embase, CINAHL, Chinese Biomedical, Chinese Journal Full-Text and Weipu Journal. Statistical analyses were calculated using version 12.0 STATA software. In total, 21 case-control studies comprising 1,187 subjects (574 patients and 613 controls) were collected for the meta-analysis. The results identified a statistically significant difference in the serum levels of leptin when comparing the PD patients with the healthy controls [controls vs. cases, standardized mean difference (SMD), 2.09; 95% confidence interval (CI), 1.58-2.59; P<0.001]. Furthermore, ethnicity-subgroup analysis indicated that the PD patients of Asian and Caucasian populations were associated with increased serum levels of leptin (Asian population, SMD, 2.05; 95% CI, 1.48-2.62; P<0.001; Caucasian population, SMD, 2.19; 95% CI, 1.19-3.18; P<0.001). Therefore, serum leptin levels may be used as a prognostic marker for PD.
Collapse
Affiliation(s)
- Shan Jiang
- Department of Nephrology, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215000, P.R. China
| | - Kai Song
- Department of Nephrology, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215000, P.R. China
| | - Sheng Feng
- Department of Nephrology, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215000, P.R. China
| | - Yong-Bin Shi
- Department of Nephrology, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215000, P.R. China
| |
Collapse
|
44
|
20 years of leptin: Role of leptin in cardiomyocyte physiology and physiopathology. Life Sci 2015; 140:10-8. [PMID: 25748420 DOI: 10.1016/j.lfs.2015.02.016] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2015] [Accepted: 02/14/2015] [Indexed: 02/08/2023]
Abstract
Since the discovery of leptin in 1994 by Zhang et al., there have been a number of reports showing its implication in the development of a wide range of cardiovascular diseases. However, there exists some controversy about how leptin can induce or preserve cardiovascular function, as different authors have found contradictory results about leptin beneficial or detrimental effects in leptin deficient/resistant murine models and in wild type tissue and cardiomyocytes. Here, we will focus on the main discoveries about the leptin functions at cardiac level within the last two decades, focusing on its role in cardiac metabolism, remodeling and contractile function.
Collapse
|
45
|
Endrighi R, Hamer M, Hackett RA, Carvalho LA, Jackson SE, Wardle J, Steptoe A. Effect of short-term weight loss on mental stress-induced cardiovascular and pro-inflammatory responses in women. Stress 2015; 18:602-6. [PMID: 26181102 PMCID: PMC4732430 DOI: 10.3109/10253890.2015.1064889] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Epidemiologic evidence links psychosocial stress with obesity but experimental studies examining the mechanisms that mediates the effect of stress on adiposity are scarce. The aim of this study was to investigate whether changes in adiposity following minimal weight loss affect heightened stress responses in women, and examine the role of the adipokine leptin in driving inflammatory responses. Twenty-three overweight or obese, but otherwise healthy, women (M age = 30.41 ± 8.0 years; BMI = 31.9 ± 4.1 kg/m(2)) completed standardized acute mental stress before and after a 9-week calorie restriction program designed to modify adiposity levels. Cardiovascular (blood pressure and heart rate) and inflammatory cytokines (leptin and interleukin-6; IL-6) responses to mental stress were assessed several times between baseline and a 45-min post-stress recovery period. There were modest changes in adiposity measures while the adipokine leptin was markedly reduced (-27%) after the intervention. Blood pressure reactivity was attenuated (-3.38 ± 1.39 mmHg) and heart rate recovery was improved (2.07 ± 0.96 Bpm) after weight loss. Blood pressure responses were inversely associated with changes in waist to hip ratio post intervention. Decreased levels of circulating leptin following weight loss were inversely associated with the IL-6 inflammatory response to stress (r = -0.47). We offered preliminary evidence suggesting that modest changes in adiposity following a brief caloric restriction program may yield beneficial effect on cardiovascular stress responses. In addition, reductions in basal leptin activity might be important in blunting pro-inflammatory responses. Large randomized trials of the effect of adiposity on autonomic responses are thus warranted.
Collapse
Affiliation(s)
- Romano Endrighi
- Department of Medical and Clinical Psychology, F. Edward Hébert School of Medicine, Uniformed Services University of the Health Sciences,
Bethesda,
MD,
USA
- Department of Epidemiology and Public Health (Psychobiology Group)
- Correspondence: Romano Endrighi,
USUHS, Bldg. 28 Rm. 113, 4301 Jones Bridge Rd.,
Bethesda,
MD 20814,
USA. Tel: +1 301 295 1530. Fax: +1 301 295 3034. E-mail: ;
| | - Mark Hamer
- Department of Epidemiology and Public Health (Psychobiology Group)
| | - Ruth A. Hackett
- Department of Epidemiology and Public Health (Psychobiology Group)
| | | | - Sarah E. Jackson
- Department of Epidemiology and Public Health (Health Behavior Research Center), Institute of Epidemiology and Health Care, University College London,
London,
UK
| | - Jane Wardle
- Department of Epidemiology and Public Health (Health Behavior Research Center), Institute of Epidemiology and Health Care, University College London,
London,
UK
| | - Andrew Steptoe
- Department of Epidemiology and Public Health (Psychobiology Group)
| |
Collapse
|
46
|
ACE2 activation by xanthenone prevents leptin-induced increases in blood pressure and proteinuria during pregnancy in Sprague-Dawley rats. Reprod Toxicol 2014; 49:155-61. [DOI: 10.1016/j.reprotox.2014.08.006] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2014] [Revised: 08/04/2014] [Accepted: 08/25/2014] [Indexed: 12/13/2022]
|
47
|
Li Z, Yu X, Liang J, Wu WKK, Yu J, Shen J. Leptin downregulates aggrecan through the p38-ADAMST pathway in human nucleus pulposus cells. PLoS One 2014; 9:e109595. [PMID: 25299465 PMCID: PMC4192299 DOI: 10.1371/journal.pone.0109595] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2014] [Accepted: 09/05/2014] [Indexed: 12/22/2022] Open
Abstract
The mechanistic basis of obesity-associated intervertebral disc degeneration (IDD) is unclear. Aberrant expression of aggrecan and its degrading enzymes ADAMTS-4 and ADAMTS-5 is implicated in the development of IDD. Here, we investigated the effect of leptin, a hormone with increased circulating levels in obesity, on the expression of aggrecan and ADAMTSs in primary human nucleus pulposus (NP) cells. Real-time PCR and Western blots showed that leptin increased the mRNA and protein expression of ADAMTS-4 and ADAMTS-5 and reduced the level of aggrecan in NP cells, accompanied by a prominent induction of p38 phosphorylation. Treatment of NP cells with SB203580 (a p38 inhibitor) abolished the regulation of aggrecan and ADAMTSs by leptin. Knockdown of ADAMTS-4 and ADAMTS-5 by siRNAs also attenuated the degradation of aggrecan in leptin-stimulated NP cells. To conclude, we demonstrated that leptin induces p38 to upregulate ADAMTSs and thereby promoting aggrecan degradation in human NP cells. These results provide a novel mechanistic insight into the molecular pathogenesis of obesity-associated IDD.
Collapse
Affiliation(s)
- Zheng Li
- Department of Orthopaedic Surgery, Peking Union Medical College Hospital, Peking Union Medical College, Beijing, China
| | - Xin Yu
- Department of Orthopaedic Surgery, Peking Union Medical College Hospital, Peking Union Medical College, Beijing, China
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Xicheng District, Beijing, China
| | - Jinqian Liang
- Department of Orthopaedic Surgery, Peking Union Medical College Hospital, Peking Union Medical College, Beijing, China
| | - William Ka Kei Wu
- Department of Anaesthesia and Intensive Care, The Chinese University of Hong Kong, Hong Kong
| | - Jun Yu
- Department of Anaesthesia and Intensive Care, The Chinese University of Hong Kong, Hong Kong
| | - Jianxiong Shen
- Department of Orthopaedic Surgery, Peking Union Medical College Hospital, Peking Union Medical College, Beijing, China
- * E-mail:
| |
Collapse
|
48
|
Burch AE, Allen MT. Stress task specific impairments of cardiovascular functioning in obese participants. Int J Psychophysiol 2014; 94:1-8. [PMID: 25017962 DOI: 10.1016/j.ijpsycho.2014.07.002] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2014] [Revised: 07/01/2014] [Accepted: 07/02/2014] [Indexed: 11/28/2022]
Abstract
The role that excess adipose tissue plays in chronic inflammation gives rise to its importance as an independent risk factor in cardiovascular dysfunction. Operationalizing chronic stress as obesity, we sought to explore the relationship between obesity, perceived stress and cardiovascular reactivity and recovery from laboratory stressors. Cardiovascular function was assessed using blood pressure and heart rate. Two stress tasks (mental arithmetic and cold pressor) were employed to examine potential differences between type of stress and cardiovascular response. Body mass index (BMI) was able to predict dysfunction in both cardiovascular reactivity and recovery. Participants with a higher BMI exhibited blunted systolic blood pressure and heart rate reactivity to the mental arithmetic task. In contrast, BMI has an incongruent effect on blood pressure reactivity to the cold pressor task that is dependent on the level of perceived stress. This suggests that in some instances the effect of BMI on cardiovascular response to acute stress may be moderated by perceived stress. Further, we found greater adiposity was related to delayed heart rate recovery following both stress tasks.
Collapse
Affiliation(s)
- Ashley E Burch
- Department of Psychology, University of Mississippi, USA.
| | | |
Collapse
|
49
|
Li H, Wang YP, Zhang LN, Tian G. Perivascular adipose tissue-derived leptin promotes vascular smooth muscle cell phenotypic switching via p38 mitogen-activated protein kinase in metabolic syndrome rats. Exp Biol Med (Maywood) 2014; 239:954-965. [PMID: 24719379 DOI: 10.1177/1535370214527903] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Perivascular adipose tissue (PVAT)-derived leptin is a detrimental adipocytokine and plays a critical role in the development of cardiovascular diseases in metabolic syndrome (MetS). During vascular remodeling, vascular smooth muscle cells (VSMCs) undergo phenotypic switching into a synthetic phenotype characterized by decreased expression of differentiation markers (smooth muscle myosin heavy chain, α-smooth muscle actin, and calponin) and increased proliferation. We aimed to determine whether PVAT-derived leptin influences VSMC phenotypic switching and to explore the underlying mechanisms in MetS rats. In vivo, 32 Wistar rats were divided into two groups that received either a normal diet (control rat) or a high-fat diet (MetS rats). After 16 weeks, rat aortas were stained using hematoxylin–eosin and imaged. VSMC differentiation markers and proliferating cell nuclear antigen (PCNA), PVAT-derived leptin, aortic leptin receptor (ObR), and p38 mitogen-activated protein kinase (MAPK) expression were detected. In vitro, aortic VSMCs were incubated with MetS rat PVAT conditioned medium (PVAT-CM) to mimic in vivo conditions and were pretreated with a p38 MAPK inhibitor (SB 203580) or leptin antagonist. Differentiation marker expression, including PCNA and p38 MAPK, was detected. MetS rats exhibited pronounced insulin resistance, hyperglycemia, hyperlipidemia, hypertension, obesity, and an associated increase in PVAT weight. VSMCs underwent phenotypic switching in MetS rat aorta and contributed to vascular remodeling. PVAT-derived leptin expression was higher in MetS rats than in control rats ( P < 0.01). ObRa expression and p38 MAPK phosphorylation were upregulated in MetS rat aorta. In vitro, VSMCs incubated with MetS rat PVAT-CM underwent phenotypic switching, associated with increased p38 MAPK phosphorylation. This VSMC phenotypic switching was inhibited by pretreatment with SB 203580 or a leptin antagonist. These results suggest that in MetS rats, PVAT-derived leptin promotes VSMC phenotypic switching via a p38 MAPK-dependent pathway to exacerbate vascular remodeling.
Collapse
Affiliation(s)
- Hao Li
- Department of Cardiovascular Medicine, the First Affiliated Hospital, Xi’an Jiaotong University College of Medicine, Xi’an, Shaanxi 710061, P.R. China
- Department of Critical Care Medicine, the First Affiliated Hospital, Xi’an Jiaotong University College of Medicine, Xi’an, Shaanxi 710061, P.R. China
| | - Ya-Ping Wang
- Geriatric Cardiology Department,Shaanxi Provincial People’s Hospital, Xi’an, Shaanxi 710061, P.R. China
| | - Li-Na Zhang
- Department of Cardiovascular Medicine, the First Affiliated Hospital, Xi’an Jiaotong University College of Medicine, Xi’an, Shaanxi 710061, P.R. China
| | - Gang Tian
- Department of Cardiovascular Medicine, the First Affiliated Hospital, Xi’an Jiaotong University College of Medicine, Xi’an, Shaanxi 710061, P.R. China
| |
Collapse
|
50
|
Ebert T, Focke D, Petroff D, Wurst U, Richter J, Bachmann A, Lössner U, Kralisch S, Kratzsch J, Beige J, Bast I, Anders M, Blüher M, Stumvoll M, Fasshauer M. Serum levels of the myokine irisin in relation to metabolic and renal function. Eur J Endocrinol 2014; 170:501-6. [PMID: 24399249 DOI: 10.1530/eje-13-1053] [Citation(s) in RCA: 93] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
OBJECTIVE Irisin has recently been introduced as a novel myokine which reverses visceral obesity and improves glucose metabolism in mice. However, regulation of irisin in humans in relation to renal and metabolic disease has not been comprehensively studied. DESIGN AND METHODS Serum irisin levels were quantified by ELISA and correlated with anthropometric and biochemical parameters of renal function, glucose and lipid metabolism, as well as inflammation, in 532 patients with stages 1-5 of chronic kidney disease (CKD). RESULTS Median serum irisin levels adjusted for age, gender, and BMI significantly decreased with increasing CKD stage and lowest concentrations were seen in patients with CKD stage 5. Furthermore, irisin concentrations were associated with facets of the metabolic syndrome including diastolic blood pressure, markers of impaired glucose tolerance, and dyslipidemia in univariate analysis. Moreover, markers of renal function, e.g. glomerular filtration rate, and insulin resistance, e.g. homeostasis model assessment of insulin resistance, remained independently associated with circulating irisin levels in robust multivariate analysis. CONCLUSIONS We show that irisin serum concentrations decrease with increasing CKD stage and are independently and positively predicted by renal function and insulin resistance. The physiological relevance of our findings, as well as the factors contributing to irisin regulation in humans, needs to be further defined in future experiments.
Collapse
Affiliation(s)
- Thomas Ebert
- Department of Endocrinology and Nephrology, University of Leipzig, Liebigstraße 20, 04103 Leipzig, Germany
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|