1
|
Danowska M, Stefanowicz M, Strączkowski M. The expression of NFAT family genes in subcutaneous adipose tissue before and after weight loss in obese individuals. Nutr Metab Cardiovasc Dis 2024; 34:2455-2463. [PMID: 39069466 DOI: 10.1016/j.numecd.2024.06.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Revised: 06/12/2024] [Accepted: 06/13/2024] [Indexed: 07/30/2024]
Abstract
BACKGROUND AND AIMS Adipose tissue (AT) serves as a vital energy storage site and plays a pivotal role in metabolic regulation, exhibiting a high response to insulin. Impairment in this response may closely associate with obesity, and NFAT (nuclear factor of activated T cells) family genes may be involved in the process. However, human data linking NFAT and AT remains elusive. The aim of this study was to assess the expression of NFAT family genes and markers of adipogenesis in subcutaneous adipose tissue (SAT) among normal-weight and overweight/obese individuals before and after weight loss, in relation to insulin sensitivity. METHODS AND RESULTS The study included 45 participants, 15 normal-weight (control group) and 30 overweight or obese, who underwent a 12-week dietary intervention (DI) program. Before and after the program hyperinsulinemic-euglycemic clamp and SAT biopsy were conducted. Before DI, a positive correlations was observed in the expression of NFATc1, NFATc4, and NFAT5 with insulin sensitivity. The expression of NFAT family genes and markers of adipogenesis in SAT was lower in individuals with overweight or obesity compared to normal-weight. Additionally, a positive correlation was noted between NFAT family genes and adipogenesis markers both before and after weight loss. Following the DI program, there was an increase in the expression of NFATc3, NFATc4, and NFAT5 in SAT. CONCLUSION Decreased SAT expression of NFAT genes in obesity is partly reversed in response to weight loss. NFAT genes in SAT are associated with insulin sensitivity and adipogenesis. Registration number for clinical trial: NCT01393210.
Collapse
Affiliation(s)
- Magdalena Danowska
- Department of Prophylaxis of Metabolic Diseases, Institute of Animal Reproduction and Food Research, Polish Academy of Sciences, Olsztyn, Poland
| | - Magdalena Stefanowicz
- Department of Metabolic Diseases, Medical University of Bialystok, Bialystok, Poland
| | - Marek Strączkowski
- Department of Prophylaxis of Metabolic Diseases, Institute of Animal Reproduction and Food Research, Polish Academy of Sciences, Olsztyn, Poland.
| |
Collapse
|
2
|
Ma K, Zhang Y, Zhao J, Zhou L, Li M. Endoplasmic reticulum stress: bridging inflammation and obesity-associated adipose tissue. Front Immunol 2024; 15:1381227. [PMID: 38638434 PMCID: PMC11024263 DOI: 10.3389/fimmu.2024.1381227] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2024] [Accepted: 03/25/2024] [Indexed: 04/20/2024] Open
Abstract
Obesity presents a significant global health challenge, increasing the susceptibility to chronic conditions such as diabetes, cardiovascular disease, and hypertension. Within the context of obesity, lipid metabolism, adipose tissue formation, and inflammation are intricately linked to endoplasmic reticulum stress (ERS). ERS modulates metabolism, insulin signaling, inflammation, as well as cell proliferation and death through the unfolded protein response (UPR) pathway. Serving as a crucial nexus, ERS bridges the functionality of adipose tissue and the inflammatory response. In this review, we comprehensively elucidate the mechanisms by which ERS impacts adipose tissue function and inflammation in obesity, aiming to offer insights into targeting ERS for ameliorating metabolic dysregulation in obesity-associated chronic diseases such as hyperlipidemia, hypertension, fatty liver, and type 2 diabetes.
Collapse
Affiliation(s)
| | | | | | | | - Min Li
- Institute of Metabolic Diseases, Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| |
Collapse
|
3
|
Li LY, Liu SF, Zhuang JL, Li MM, Huang ZP, Chen YH, Chen XR, Chen CN, Lin S, Ye LC. Recent research progress on metabolic syndrome and risk of Parkinson's disease. Rev Neurosci 2023; 34:719-735. [PMID: 36450297 DOI: 10.1515/revneuro-2022-0093] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Accepted: 11/06/2022] [Indexed: 10/05/2023]
Abstract
Parkinson's disease (PD) is one of the most widespread neurodegenerative diseases. PD is associated with progressive loss of substantia nigra dopaminergic neurons, including various motor symptoms (e.g., bradykinesia, rigidity, and resting tremor), as well as non-motor symptoms (e.g., cognitive impairment, constipation, fatigue, sleep disturbance, and depression). PD involves multiple biological processes, including mitochondrial or lysosomal dysfunction, oxidative stress, insulin resistance, and neuroinflammation. Metabolic syndrome (MetS), a collection of numerous connected cerebral cardiovascular conditions, is a common and growing public health problem associated with many chronic diseases worldwide. MetS components include central/abdominal obesity, systemic hypertension, diabetes, and atherogenic dyslipidemia. MetS and PD share multiple pathophysiological processes, including insulin resistance, oxidative stress, and chronic inflammation. In recent years, MetS has been linked to an increased risk of PD, according to studies; however, the specific mechanism remains unclear. Researchers also found that some related metabolic therapies are potential therapeutic strategies to prevent and improve PD. This article reviews the epidemiological relationship between components of MetS and the risk of PD and discusses the potentially relevant mechanisms and recent progress of MetS as a risk factor for PD. Furthermore, we conclude that MetS-related therapies are beneficial for the prevention and treatment of PD.
Collapse
Affiliation(s)
- Lin-Yi Li
- Department of Neurology, The Second Affiliated Hospital, Fujian Medical University, No. 34 North Zhongshan Road, Quanzhou 362000, Fujian Province, China
| | - Shu-Fen Liu
- Department of Neurology, The Second Affiliated Hospital, Fujian Medical University, No. 34 North Zhongshan Road, Quanzhou 362000, Fujian Province, China
| | - Jian-Long Zhuang
- Prenatal Diagnosis Center, Quanzhou Women's and Children's Hospital, Quanzhou 362000, China
| | - Mi-Mi Li
- Department of Neurology, The Second Affiliated Hospital, Fujian Medical University, No. 34 North Zhongshan Road, Quanzhou 362000, Fujian Province, China
| | - Zheng-Ping Huang
- Department of Neurology, The Second Affiliated Hospital, Fujian Medical University, No. 34 North Zhongshan Road, Quanzhou 362000, Fujian Province, China
| | - Yan-Hong Chen
- Department of Neurology, Shishi General Hospital, Quanzhou 362000, Fujian Province, China
| | - Xiang-Rong Chen
- Department of Neurosurgery, The Second Affiliated Hospital, Fujian Medical University, Quanzhou 362000, Fujian Province, China
| | - Chun-Nuan Chen
- Department of Neurology, The Second Affiliated Hospital, Fujian Medical University, No. 34 North Zhongshan Road, Quanzhou 362000, Fujian Province, China
| | - Shu Lin
- Centre of Neurological and Metabolic Research, The Second Affiliated Hospital of Fujian Medical University, Quanzhou 362000, Fujian Province, China
- Group of Neuroendocrinology, Garvan Institute of Medical Research, 384 Victoria St, Sydney, NSW, Australia
| | - Li-Chao Ye
- Department of Neurology, The Second Affiliated Hospital, Fujian Medical University, No. 34 North Zhongshan Road, Quanzhou 362000, Fujian Province, China
| |
Collapse
|
4
|
Kim D, Memili A, Chen HH, Highland HM, Polikowsky HG, Anwar MY, Laing ST, Lee M, McCormick JB, Fisher-Hoch SP, Below JE, North KE, Gutierrez AD. Sex-specific associations between adipokine profiles and carotid-intima media thickness in the Cameron County Hispanic Cohort (CCHC). Cardiovasc Diabetol 2023; 22:231. [PMID: 37653519 PMCID: PMC10472619 DOI: 10.1186/s12933-023-01968-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/22/2023] [Accepted: 08/16/2023] [Indexed: 09/02/2023] Open
Abstract
BACKGROUND Adipokines are hormones secreted from adipose tissue and are associated with cardiometabolic diseases (CMD). Functional differences between adipokines (leptin, adiponectin, and resistin) are known, but inconsistently reported associations with CMD and lack of studies in Hispanic populations are research gaps. We investigated the relationship between subclinical atherosclerosis and multiple adipokine measures. METHODS Cross-sectional data from the Cameron County Hispanic Cohort (N = 624; mean age = 50; Female = 70.8%) were utilized to assess associations between adipokines [continuous measures of adiponectin, leptin, resistin, leptin-to-adiponectin ratio (LAR), and adiponectin-resistin index (ARI)] and early atherosclerosis [carotid-intima media thickness (cIMT)]. We adjusted for sex, age, body mass index (BMI), smoking status, cytokines, fasting blood glucose levels, blood pressure, lipid levels, and medication usage in the fully adjusted linear regression model. We conducted sexes-combined and sex-stratified analyses to account for sex-specificity and additionally tested whether stratification of participants by their metabolic status (metabolically elevated risk for CMD as defined by having two or more of the following conditions: hypertension, dyslipidemia, insulin resistance, and inflammation vs. not) influenced the relationship between adipokines and cIMT. RESULTS In the fully adjusted analyses, adiponectin, leptin, and LAR displayed significant interaction by sex (p < 0.1). Male-specific associations were between cIMT and LAR [β(SE) = 0.060 (0.016), p = 2.52 × 10-4], and female-specific associations were between cIMT and adiponectin [β(SE) = 0.010 (0.005), p = 0.043] and ARI [β(SE) = - 0.011 (0.005), p = 0.036]. When stratified by metabolic health status, the male-specific positive association between LAR and cIMT was more evident among the metabolically healthy group [β(SE) = 0.127 (0.015), p = 4.70 × 10-10] (p for interaction by metabolic health < 0.1). However, the female-specific associations between adiponectin and cIMT and ARI and cIMT were observed only among the metabolically elevated risk group [β(SE) = 0.014 (0.005), p = 0.012 for adiponectin; β(SE) = - 0.015 (0.006), p = 0.013 for ARI; p for interaction by metabolic health < 0.1]. CONCLUSION Associations between adipokines and cIMT were sex-specific, and metabolic health status influenced the relationships between adipokines and cIMT. These heterogeneities by sex and metabolic health affirm the complex relationships between adipokines and atherosclerosis.
Collapse
Affiliation(s)
- Daeeun Kim
- Department of Epidemiology, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Aylin Memili
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, USA
| | - Hung-Hsin Chen
- Vanderbilt Genetics Institute, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Heather M Highland
- Department of Epidemiology, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Hannah G Polikowsky
- Vanderbilt Genetics Institute, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Mohammad Yaser Anwar
- Department of Epidemiology, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Susan T Laing
- Department of Internal Medicine, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Miryoung Lee
- Department of Epidemiology, Human Genetics and Environmental Sciences, The University of Texas Health Science Center at Houston School of Public Health, Brownsville Regional Campus, Brownsville, TX, USA
| | - Joseph B McCormick
- Department of Epidemiology, Human Genetics and Environmental Sciences, The University of Texas Health Science Center at Houston School of Public Health, Brownsville Regional Campus, Brownsville, TX, USA
| | - Susan P Fisher-Hoch
- Department of Epidemiology, Human Genetics and Environmental Sciences, The University of Texas Health Science Center at Houston School of Public Health, Brownsville Regional Campus, Brownsville, TX, USA
| | - Jennifer E Below
- Vanderbilt Genetics Institute, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Kari E North
- Department of Epidemiology, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Absalon D Gutierrez
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, The University of Texas Health Science Center at Houston McGovern Medical School, Houston, TX, USA.
| |
Collapse
|
5
|
Glassman I, Le N, Asif A, Goulding A, Alcantara CA, Vu A, Chorbajian A, Mirhosseini M, Singh M, Venketaraman V. The Role of Obesity in Breast Cancer Pathogenesis. Cells 2023; 12:2061. [PMID: 37626871 PMCID: PMC10453206 DOI: 10.3390/cells12162061] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Revised: 08/03/2023] [Accepted: 08/11/2023] [Indexed: 08/27/2023] Open
Abstract
Research has shown that obesity increases the risk for type 2 diabetes mellitus (Type 2 DM) by promoting insulin resistance, increases serum estrogen levels by the upregulation of aromatase, and promotes the release of reactive oxygen species (ROS) by macrophages. Increased circulating glucose has been shown to activate mammalian target of rapamycin (mTOR), a significant signaling pathway in breast cancer pathogenesis. Estrogen plays an instrumental role in estrogen-receptor-positive breast cancers. The role of ROS in breast cancer warrants continued investigation, in relation to both pathogenesis and treatment of breast cancer. We aim to review the role of obesity in breast cancer pathogenesis and novel therapies mediating obesity-associated breast cancer development. We explore the association between body mass index (BMI) and breast cancer incidence and the mechanisms by which oxidative stress modulates breast cancer pathogenesis. We discuss the role of glutathione, a ubiquitous antioxidant, in breast cancer therapy. Lastly, we review breast cancer therapies targeting mTOR signaling, leptin signaling, blood sugar reduction, and novel immunotherapy targets.
Collapse
Affiliation(s)
- Ira Glassman
- College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, CA 91766, USA (N.L.); (A.A.); (C.A.A.); (M.M.)
| | - Nghia Le
- College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, CA 91766, USA (N.L.); (A.A.); (C.A.A.); (M.M.)
| | - Aamna Asif
- College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, CA 91766, USA (N.L.); (A.A.); (C.A.A.); (M.M.)
| | - Anabel Goulding
- College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, CA 91766, USA (N.L.); (A.A.); (C.A.A.); (M.M.)
| | - Cheldon Ann Alcantara
- College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, CA 91766, USA (N.L.); (A.A.); (C.A.A.); (M.M.)
| | - Annie Vu
- College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, CA 91766, USA (N.L.); (A.A.); (C.A.A.); (M.M.)
| | - Abraham Chorbajian
- College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, CA 91766, USA (N.L.); (A.A.); (C.A.A.); (M.M.)
| | - Mercedeh Mirhosseini
- College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, CA 91766, USA (N.L.); (A.A.); (C.A.A.); (M.M.)
| | - Manpreet Singh
- Corona Regional Medical Center, Department of Emergency Medicine, Corona, CA 92882, USA
| | - Vishwanath Venketaraman
- College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, CA 91766, USA (N.L.); (A.A.); (C.A.A.); (M.M.)
| |
Collapse
|
6
|
Herrerías-González F, Yeramian A, Baena-Fustegueras JA, Bueno M, Fleitas C, de la Fuente M, Serrano JCE, Granado-Serrano A, Santamaría M, Yeramian N, Zorzano-Martínez M, Mora C, Lecube A. PKN1 Kinase: A Key Player in Adipocyte Differentiation and Glucose Metabolism. Nutrients 2023; 15:nu15102414. [PMID: 37242297 DOI: 10.3390/nu15102414] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2023] [Revised: 05/12/2023] [Accepted: 05/17/2023] [Indexed: 05/28/2023] Open
Abstract
Adipocyte dysfunction is the driver of obesity and correlates with insulin resistance and the onset of type 2 diabetes. Protein kinase N1 (PKN1) is a serine/threonine kinase that has been shown to contribute to Glut4 translocation to the membrane and glucose transport. Here, we evaluated the role of PKN1 in glucose metabolism under insulin-resistant conditions in primary visceral adipose tissue (VAT) from 31 patients with obesity and in murine 3T3-L1 adipocytes. In addition, in vitro studies in human VAT samples and mouse adipocytes were conducted to investigate the role of PKN1 in the adipogenic maturation process and glucose homeostasis control. We show that insulin-resistant adipocytes present a decrease in PKN1 activation levels compared to nondiabetic control counterparts. We further show that PKN1 controls the adipogenesis process and glucose metabolism. PKN1-silenced adipocytes present a decrease in both differentiation process and glucose uptake, with a concomitant decrease in the expression levels of adipogenic markers, such as PPARγ, FABP4, adiponectin and CEBPα. Altogether, these results point to PKN1 as a regulator of key signaling pathways involved in adipocyte differentiation and as an emerging player of adipocyte insulin responsiveness. These findings may provide new therapeutic approaches for the management of insulin resistance in type 2 diabetes.
Collapse
Affiliation(s)
- Fernando Herrerías-González
- Experimental Surgery Research Group, General and Digestive Surgery Department, Arnau de Vilanova University Hospital, University of Lleida, 25716 Lleida, Spain
- Institut de Recerca Biomèdica Lleida (IRB-LLeida), 25198 Lleida, Spain
| | - Andrée Yeramian
- Institut de Recerca Biomèdica Lleida (IRB-LLeida), 25198 Lleida, Spain
- Department of Experimental Medicine, University of Lleida, 25198 Lleida, Spain
| | - Juan Antonio Baena-Fustegueras
- Experimental Surgery Research Group, General and Digestive Surgery Department, Arnau de Vilanova University Hospital, University of Lleida, 25716 Lleida, Spain
- Institut de Recerca Biomèdica Lleida (IRB-LLeida), 25198 Lleida, Spain
| | - Marta Bueno
- Institut de Recerca Biomèdica Lleida (IRB-LLeida), 25198 Lleida, Spain
- Obesity, Diabetes and Metabolism (ODIM) Research Group, Endocrinology and Nutrition Department, Arnau de Vilanova University Hospital, University of Lleida, 25716 Lleida, Spain
| | - Catherine Fleitas
- Biobank Unit, Hospital Universitari Arnau de Vilanova, IRB-Lleida, 25198 Lleida, Spain
| | - Maricruz de la Fuente
- Experimental Surgery Research Group, General and Digestive Surgery Department, Arnau de Vilanova University Hospital, University of Lleida, 25716 Lleida, Spain
- Institut de Recerca Biomèdica Lleida (IRB-LLeida), 25198 Lleida, Spain
| | - José C E Serrano
- Institut de Recerca Biomèdica Lleida (IRB-LLeida), 25198 Lleida, Spain
- Department of Experimental Medicine, University of Lleida, 25198 Lleida, Spain
| | - Ana Granado-Serrano
- Institut de Recerca Biomèdica Lleida (IRB-LLeida), 25198 Lleida, Spain
- Department of Experimental Medicine, University of Lleida, 25198 Lleida, Spain
| | - Maite Santamaría
- Experimental Surgery Research Group, General and Digestive Surgery Department, Arnau de Vilanova University Hospital, University of Lleida, 25716 Lleida, Spain
- Institut de Recerca Biomèdica Lleida (IRB-LLeida), 25198 Lleida, Spain
| | - Nadine Yeramian
- Department of Biotechnology and Food Science, Faculty of Science, University of Burgos, 09001 Burgos, Spain
| | - Marta Zorzano-Martínez
- Institut de Recerca Biomèdica Lleida (IRB-LLeida), 25198 Lleida, Spain
- Obesity, Diabetes and Metabolism (ODIM) Research Group, Endocrinology and Nutrition Department, Arnau de Vilanova University Hospital, University of Lleida, 25716 Lleida, Spain
| | - Conchi Mora
- Institut de Recerca Biomèdica Lleida (IRB-LLeida), 25198 Lleida, Spain
- Immunology Unit, Department of Experimental Medicine, Faculty of Medicine, University of Lleida, 25716 Lleida, Spain
| | - Albert Lecube
- Institut de Recerca Biomèdica Lleida (IRB-LLeida), 25198 Lleida, Spain
- Obesity, Diabetes and Metabolism (ODIM) Research Group, Endocrinology and Nutrition Department, Arnau de Vilanova University Hospital, University of Lleida, 25716 Lleida, Spain
| |
Collapse
|
7
|
Abstract
Non-alcoholic fatty liver disease (NAFLD) is a challenging disease caused by multiple factors, which may partly explain why it still remains an orphan of adequate therapies. This review highlights the interaction between oxidative stress (OS) and disturbed lipid metabolism. Several reactive oxygen species generators, including those produced in the gastrointestinal tract, contribute to the lipotoxic hepatic (and extrahepatic) damage by fatty acids and a great variety of their biologically active metabolites in a “multiple parallel-hit model”. This leads to inflammation and fibrogenesis and contributes to NAFLD progression. The alterations of the oxidant/antioxidant balance affect also metabolism-related organelles, leading to lipid peroxidation, mitochondrial dysfunction, and endoplasmic reticulum stress. This OS-induced damage is at least partially counteracted by the physiological antioxidant response. Therefore, modulation of this defense system emerges as an interesting target to prevent NAFLD development and progression. For instance, probiotics, prebiotics, diet, and fecal microbiota transplantation represent new therapeutic approaches targeting the gut microbiota dysbiosis. The OS and its counter-regulation are under the influence of individual genetic and epigenetic factors as well. In the near future, precision medicine taking into consideration genetic or environmental epigenetic risk factors, coupled with new OS biomarkers, will likely assist in noninvasive diagnosis and monitoring of NAFLD progression and in further personalizing treatments.
Collapse
|
8
|
Abstract
Two decades of research have established that Nuclear Factor-κB (NF-κB) signaling plays a critical role in reprogramming the fat cell transcriptome towards inflammation in response to overnutrition and metabolic stress. Several groups have suggested that inhibition of NF-κB signaling could have metabolic benefits for obesity-associated adipose tissue inflammation. However, two significant problems arise with this approach. The first is how to deliver general NF-κB inhibitors into adipocytes without allowing these compounds to disrupt normal functioning in cells of the immune system. The second issue is that general inhibition of canonical NF-κB signaling in adipocytes will likely lead to a massive increase in adipocyte apoptosis under conditions of metabolic stress, leading full circle into a secondary inflammation (However, this problem may not be true for non-canonical NF-κB signaling.). This review will focus on the research that has examined canonical and non-canonical NF-κB signaling in adipocytes, focusing on genetic studies that examine loss-of-function of NF-κB specifically in fat cells. Although the development of general inhibitors of canonical NF-κB signaling seems unlikely to succeed in alleviating adipose tissue inflammation in humans, the door remains open for more targeted therapeutics. In principle, these would include compounds that interrogate NF-κB DNA binding, protein-protein interactions, or post-translational modifications that partition NF-κB activity towards some genes and away from others in adipocytes. I also discuss the possibility for inhibitors of non-canonical NF-κB signaling to realize success in mitigating fat cell dysfunction in obesity. To plant the seeds for such approaches, much biochemical “digging” in adipocytes remains; this includes identifying—in an unbiased manner–NF-κB direct and indirect targets, genomic DNA binding sites for all five NF-κB subunits, NF-κB protein-protein interactions, and post-translational modifications of NF-κB in fat cells.
Collapse
|
9
|
Jadhav RA, Maiya GA, Hombali A, Umakanth S, Shivashankar KN. Effect of physical activity promotion on adiponectin, leptin and other inflammatory markers in prediabetes: a systematic review and meta-analysis of randomized controlled trials. Acta Diabetol 2021; 58:419-429. [PMID: 33211181 PMCID: PMC8053655 DOI: 10.1007/s00592-020-01626-1] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/01/2020] [Accepted: 10/21/2020] [Indexed: 12/16/2022]
Abstract
AIMS Inflammatory stage in prediabetes is associated with increase in level of adipokines and pro-inflammatory cytokines. Physical activity promotion considered as a first-line therapeutic strategy to treat prediabetes. We have conducted the systematic review and meta-analysis to strengthen the evidence on the impact of physical activity promotion on inflammatory markers in prediabetes. METHODS Studies were identified using electronic search and manual search techniques by choosing keywords for prediabetes, physical activity and inflammatory marker. Randomized controlled trials on individuals diagnosed with prediabetes and provided intervention in the form of physical activity were included in this review. Adiponectin, leptin, C-reactive protein, interleukin-6 and tumour necrosis factor-α were the considered outcome measures. RESULTS Our search retrieved 1,688 citations, 31 full-text articles assessed for eligibility of inclusion. Nine studies satisfied the pre-specified criteria for inclusion. Meta-analysis found that physical activity with or without dietary or lifestyle modification reduces level of leptin (MD-2.11 ng/mL, 95% CI -3.81 - -0.42) and interleukin-6 (MD -0.15 pg/mL, 95% CI -0.25--0.04). It has no effect on level of adiponectin (MD 0.26 µg/mL, 95% CI -0.42- 0.93), C-reactive protein (MD -0.05 mg/L, 95% CI -0.33-0.23) and tumour necrosis factor-α (MD 0.67 pg/mL, 95% CI -2.56-3.89). CONCLUSIONS This review suggests that physical activity promotion with dietary and lifestyle modification may reduce the level of leptin and interleukin-6 but are uncertain if there is any effect on levels of adiponectin, C-reactive protein and tumour necrosis factor-α in the individuals with prediabetes.
Collapse
Affiliation(s)
- Radhika Aditya Jadhav
- Department of Physiotherapy, Centre for Diabetic Foot Care and Research, Manipal College of Health Professions, Manipal Academy of Higher Education, Manipal, 576104, Karnataka, India
| | - G Arun Maiya
- Department of Physiotherapy, Centre for Diabetic Foot Care and Research, Manipal College of Health Professions, Manipal Academy of Higher Education, Manipal, 576104, Karnataka, India.
| | - Aditi Hombali
- Independent Systematic Reviewer, Luton, Bedfordshire, UK
| | - Shashikiran Umakanth
- Department of Medicine, Melaka Manipal Medical College, Manipal Academy of Higher Education, Manipal, 576104, Karnataka, India
| | - K N Shivashankar
- Department of Medicine, Kasturba Medical College, Manipal Academy of Higher Education, Manipal, 576104, Karnataka, India
| |
Collapse
|
10
|
Penna E, Pizzella A, Cimmino F, Trinchese G, Cavaliere G, Catapano A, Allocca I, Chun JT, Campanozzi A, Messina G, Precenzano F, Lanzara V, Messina A, Monda V, Monda M, Perrone-Capano C, Mollica MP, Crispino M. Neurodevelopmental Disorders: Effect of High-Fat Diet on Synaptic Plasticity and Mitochondrial Functions. Brain Sci 2020; 10:brainsci10110805. [PMID: 33142719 PMCID: PMC7694125 DOI: 10.3390/brainsci10110805] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2020] [Revised: 10/28/2020] [Accepted: 10/29/2020] [Indexed: 02/07/2023] Open
Abstract
Neurodevelopmental disorders (NDDs) include diverse neuropathologies characterized by abnormal brain development leading to impaired cognition, communication and social skills. A common feature of NDDs is defective synaptic plasticity, but the underlying molecular mechanisms are only partially known. Several studies have indicated that people’s lifestyles such as diet pattern and physical exercise have significant influence on synaptic plasticity of the brain. Indeed, it has been reported that a high-fat diet (HFD, with 30–50% fat content), which leads to systemic low-grade inflammation, has also a detrimental effect on synaptic efficiency. Interestingly, metabolic alterations associated with obesity in pregnant woman may represent a risk factor for NDDs in the offspring. In this review, we have discussed the potential molecular mechanisms linking the HFD-induced metabolic dysfunctions to altered synaptic plasticity underlying NDDs, with a special emphasis on the roles played by synaptic protein synthesis and mitochondrial functions.
Collapse
Affiliation(s)
- Eduardo Penna
- Department of Biology, University of Naples Federico II, 80126 Naples, Italy; (E.P.); (A.P.); (F.C.); (G.T.); (G.C.); (A.C.); (I.A.); (M.C.)
| | - Amelia Pizzella
- Department of Biology, University of Naples Federico II, 80126 Naples, Italy; (E.P.); (A.P.); (F.C.); (G.T.); (G.C.); (A.C.); (I.A.); (M.C.)
| | - Fabiano Cimmino
- Department of Biology, University of Naples Federico II, 80126 Naples, Italy; (E.P.); (A.P.); (F.C.); (G.T.); (G.C.); (A.C.); (I.A.); (M.C.)
| | - Giovanna Trinchese
- Department of Biology, University of Naples Federico II, 80126 Naples, Italy; (E.P.); (A.P.); (F.C.); (G.T.); (G.C.); (A.C.); (I.A.); (M.C.)
| | - Gina Cavaliere
- Department of Biology, University of Naples Federico II, 80126 Naples, Italy; (E.P.); (A.P.); (F.C.); (G.T.); (G.C.); (A.C.); (I.A.); (M.C.)
| | - Angela Catapano
- Department of Biology, University of Naples Federico II, 80126 Naples, Italy; (E.P.); (A.P.); (F.C.); (G.T.); (G.C.); (A.C.); (I.A.); (M.C.)
- Department of Pharmacy, University of Naples Federico II, 80131 Naples, Italy;
| | - Ivana Allocca
- Department of Biology, University of Naples Federico II, 80126 Naples, Italy; (E.P.); (A.P.); (F.C.); (G.T.); (G.C.); (A.C.); (I.A.); (M.C.)
| | - Jong Tai Chun
- Department of Biology and Evolution of Marine Organisms, Stazione Zoologica Anton Dohrn, 80121 Naples, Italy;
| | - Angelo Campanozzi
- Department of Medical and Surgical Sciences, University of Foggia, 71122 Foggia, Italy;
| | - Giovanni Messina
- Department of Clinical and Experimental Medicine, University of Foggia, 71122 Foggia, Italy;
| | - Francesco Precenzano
- Department of Mental Health, Physical and Preventive Medicine, Clinic of Child and Adolescent Neuropsychiatry, Università degli Studi della Campania “Luigi Vanvitelli”, 80138 Naples, Italy; (F.P.); (V.L.)
| | - Valentina Lanzara
- Department of Mental Health, Physical and Preventive Medicine, Clinic of Child and Adolescent Neuropsychiatry, Università degli Studi della Campania “Luigi Vanvitelli”, 80138 Naples, Italy; (F.P.); (V.L.)
| | - Antonietta Messina
- Department of Experimental Medicine, Section of Human Physiology and Unit of Dietetics and Sports Medicine, Università degli Studi della Campania “Luigi Vanvitelli”, 80138 Naples, Italy; (A.M.); (M.M.)
| | - Vincenzo Monda
- Department of Experimental Medicine, Università degli Studi della Campania “Luigi Vanvitelli”, 81100 Caserta, Italy;
| | - Marcellino Monda
- Department of Experimental Medicine, Section of Human Physiology and Unit of Dietetics and Sports Medicine, Università degli Studi della Campania “Luigi Vanvitelli”, 80138 Naples, Italy; (A.M.); (M.M.)
| | - Carla Perrone-Capano
- Department of Pharmacy, University of Naples Federico II, 80131 Naples, Italy;
- Institute of Genetics and Biophysics “Adriano Buzzati Traverso”, CNR, 80131 Naples, Italy
| | - Maria Pina Mollica
- Department of Biology, University of Naples Federico II, 80126 Naples, Italy; (E.P.); (A.P.); (F.C.); (G.T.); (G.C.); (A.C.); (I.A.); (M.C.)
- Correspondence: ; Tel.: +39-081-679990; Fax: +39-081-679233
| | - Marianna Crispino
- Department of Biology, University of Naples Federico II, 80126 Naples, Italy; (E.P.); (A.P.); (F.C.); (G.T.); (G.C.); (A.C.); (I.A.); (M.C.)
| |
Collapse
|
11
|
Interplay between Peripheral and Central Inflammation in Obesity-Promoted Disorders: The Impact on Synaptic Mitochondrial Functions. Int J Mol Sci 2020; 21:ijms21175964. [PMID: 32825115 PMCID: PMC7504224 DOI: 10.3390/ijms21175964] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2020] [Revised: 08/12/2020] [Accepted: 08/17/2020] [Indexed: 12/14/2022] Open
Abstract
The metabolic dysfunctions induced by high fat diet (HFD) consumption are not limited to organs involved in energy metabolism but cause also a chronic low-grade systemic inflammation that affects the whole body including the central nervous system. The brain has been considered for a long time to be protected from systemic inflammation by the blood–brain barrier, but more recent data indicated an association between obesity and neurodegeneration. Moreover, obesity-related consequences, such as insulin and leptin resistance, mitochondrial dysfunction and reactive oxygen species (ROS) production, may anticipate and accelerate the physiological aging processes characterized by systemic inflammation and higher susceptibility to neurological disorders. Here, we discussed the link between obesity-related metabolic dysfunctions and neuroinflammation, with particular attention to molecules regulating the interplay between energetic impairment and altered synaptic plasticity, for instance AMP-activated protein kinase (AMPK) and Brain-derived neurotrophic factor (BDNF). The effects of HFD-induced neuroinflammation on neuronal plasticity may be mediated by altered brain mitochondrial functions. Since mitochondria play a key role in synaptic areas, providing energy to support synaptic plasticity and controlling ROS production, the negative effects of HFD may be more pronounced in synapses. In conclusion, it will be emphasized how HFD-induced metabolic alterations, systemic inflammation, oxidative stress, neuroinflammation and impaired brain plasticity are tightly interconnected processes, implicated in the pathogenesis of neurological diseases.
Collapse
|
12
|
Li Y, Ma Q, Li P, Wang J, Wang M, Fan Y, Wang T, Wang C, Wang T, Zhao B. Proteomics reveals different pathological processes of adipose tissue, liver, and skeletal muscle under insulin resistance. J Cell Physiol 2020; 235:6441-6461. [PMID: 32115712 DOI: 10.1002/jcp.29658] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2019] [Accepted: 02/12/2020] [Indexed: 12/17/2022]
Abstract
Type 2 diabetes mellitus is the most common type of diabetes, and insulin resistance (IR) is its core pathological mechanism. Proteomics is an ingenious and promising Omics technology that can comprehensively describe the global protein expression profiling of body or specific tissue, and is widely applied to the study of molecular mechanisms of diseases. In this paper, we focused on insulin target organs: adipose tissue, liver, and skeletal muscle, and analyzed the different pathological processes of IR in these three tissues based on proteomics research. By literature studies, we proposed that the main pathological processes of IR among target organs were diverse, which showed unique characteristics and focuses. We further summarized the differential proteins in target organs which were verified to be related to IR, and discussed the proteins that may play key roles in the emphasized pathological processes, aiming at discovering potentially specific differential proteins of IR, and providing new ideas for pathological mechanism research of IR.
Collapse
Affiliation(s)
- Yaqi Li
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Quantao Ma
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Pengfei Li
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Jingkang Wang
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Min Wang
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Yuanyuan Fan
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Tieshan Wang
- Beijing Research Institute of Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Chunguo Wang
- Beijing Research Institute of Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Ting Wang
- Beijing Research Institute of Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Baosheng Zhao
- Beijing Research Institute of Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| |
Collapse
|
13
|
Hoshino S, Kobayashi M, Tagawa R, Konno R, Abe T, Furuya K, Miura K, Wakasawa H, Okita N, Sudo Y, Mizunoe Y, Nakagawa Y, Nakamura T, Kawabe H, Higami Y. WWP1 knockout in mice exacerbates obesity-related phenotypes in white adipose tissue but improves whole-body glucose metabolism. FEBS Open Bio 2020; 10:306-315. [PMID: 31965758 PMCID: PMC7050250 DOI: 10.1002/2211-5463.12795] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2019] [Revised: 12/27/2019] [Accepted: 01/16/2020] [Indexed: 12/31/2022] Open
Abstract
White adipose tissue (WAT) is important for maintenance of homeostasis, because it stores energy and secretes adipokines. The WAT of obese people demonstrates mitochondrial dysfunction, accompanied by oxidative stress, which leads to insulin resistance. WW domain-containing E3 ubiquitin protein ligase 1 (WWP1) is a member of the HECT-type E3 family of ubiquitin ligases and is associated with several diseases. Recently, we demonstrated that WWP1 is induced specifically in the WAT of obese mice, where it protects against oxidative stress. Here, we investigated the function of WWP1 in WAT of obese mice by analyzing the phenotype of Wwp1 knockout (KO) mice fed a high-fat diet. The levels of oxidative stress markers were higher in obese WAT from Wwp1 KO mice. Moreover, Wwp1 KO mice had lower activity of citrate synthase, a mitochondrial enzyme. We also measured AKT phosphorylation in obese WAT and found lower levels in Wwp1 KO mice. However, plasma insulin level was low and glucose level was unchanged in obese Wwp1 KO mice. Moreover, both glucose tolerance test and insulin tolerance test were improved in obese Wwp1 KO mice. These findings indicate that WWP1 participates in the antioxidative response and mitochondrial function in WAT, but knockdown of WWP1 improves whole-body glucose metabolism.
Collapse
Affiliation(s)
- Shunsuke Hoshino
- Laboratory of Molecular Pathology and Metabolic Disease, Faculty of Pharmaceutical Sciences, Tokyo University of Science, Noda, Japan
| | - Masaki Kobayashi
- Laboratory of Molecular Pathology and Metabolic Disease, Faculty of Pharmaceutical Sciences, Tokyo University of Science, Noda, Japan
| | - Ryoma Tagawa
- Laboratory of Molecular Pathology and Metabolic Disease, Faculty of Pharmaceutical Sciences, Tokyo University of Science, Noda, Japan
| | - Ryutaro Konno
- Laboratory of Molecular Pathology and Metabolic Disease, Faculty of Pharmaceutical Sciences, Tokyo University of Science, Noda, Japan
| | - Takuro Abe
- Laboratory of Molecular Pathology and Metabolic Disease, Faculty of Pharmaceutical Sciences, Tokyo University of Science, Noda, Japan
| | - Kazuhiro Furuya
- Laboratory of Molecular Pathology and Metabolic Disease, Faculty of Pharmaceutical Sciences, Tokyo University of Science, Noda, Japan
| | - Kumi Miura
- Laboratory of Molecular Pathology and Metabolic Disease, Faculty of Pharmaceutical Sciences, Tokyo University of Science, Noda, Japan
| | - Hiroki Wakasawa
- Laboratory of Molecular Pathology and Metabolic Disease, Faculty of Pharmaceutical Sciences, Tokyo University of Science, Noda, Japan
| | - Naoyuki Okita
- Division of Pathological Biochemistry, Faculty of Pharmaceutical Sciences, Sanyo-Onoda City University, Sanyo-onoda, Japan
| | - Yuka Sudo
- Laboratory of Molecular Pathology and Metabolic Disease, Faculty of Pharmaceutical Sciences, Tokyo University of Science, Noda, Japan
| | - Yuhei Mizunoe
- Department of Internal Medicine (Endocrinology and Metabolism), Faculty of Medicine, University of Tsukuba, Tsukuba, Japan
| | - Yoshimi Nakagawa
- Department of Internal Medicine (Endocrinology and Metabolism), Faculty of Medicine, University of Tsukuba, Tsukuba, Japan
| | - Takeshi Nakamura
- Division of Biosignaling, Research Institute for Biomedical Sciences, Tokyo University of Science, Noda, Japan
| | - Hiroshi Kawabe
- Department of Molecular Neurobiology, Max Planck Institute of Experimental Medicine, Göttingen, Germany.,Division of Pathogenic Signaling, Department of Biochemistry and Molecular Biology, Kobe University Graduate School of Medicine, Chuo-ku, Kobe, Japan.,Department of Gerontology, Laboratory of Molecular Life Science, Institute of Biomedical Research and Innovation, Foundation for Biomedical Research and Innovation at Kobe, Japan
| | - Yoshikazu Higami
- Laboratory of Molecular Pathology and Metabolic Disease, Faculty of Pharmaceutical Sciences, Tokyo University of Science, Noda, Japan
| |
Collapse
|
14
|
Anti-obesity effect of cocoa proteins (Theobroma cacao L.) variety “Criollo” and the expression of genes related to the dysfunction of white adipose tissue in high-fat diet-induced obese rats. J Funct Foods 2019. [DOI: 10.1016/j.jff.2019.103519] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
|
15
|
Saleh J, Al-Maqbali M, Abdel-Hadi D. Role of Complement and Complement-Related Adipokines in Regulation of Energy Metabolism and Fat Storage. Compr Physiol 2019; 9:1411-1429. [PMID: 31688967 DOI: 10.1002/cphy.c170037] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
Adipose tissue releases many cytokines and inflammatory factors described as adipokines. In obesity, adipokines released from expanding adipose tissue are implicated in disease progression and metabolic dysfunction. However, mechanisms controlling the progression of adiposity and metabolic complications are not fully understood. It has been suggested that expanding fat mass and sustained release of inflammatory adipokines in adipose tissue lead to hypoxia, oxidative stress, apoptosis, and cellular damage. These changes trigger an immune response involving infiltration of adipose tissue with immune cells, complement activation and generation of factors involved in opsonization and clearance of damaged cells. Abundant evidence now indicates that adipose tissue is an active secretory source of complement and complement-related adipokines that, in addition to their inflammatory role, contribute to the regulation of metabolic function. This article highlights advances in knowledge regarding the role of these adipokines in energy regulation of adipose tissue through modulating lipogenic and lipolytic pathways. Several adipokines will be discussed including adipsin, Factor H, properdin, C3a, Acylation-Stimulating Protein, C1q/TNF-related proteins, and response gene to complement-32 (RGC-32). Interactions between these factors will be described considering their immune-metabolic roles in the adipose tissue microenvironment and their potential contribution to progression of adiposity and metabolic dysfunction. The differential expression and the role of complement factors in gender-related fat partitioning will also be addressed. Identifying lipogenic adipokines and their specific autocrine/paracrine roles may provide means for adipose-tissue-targeted therapeutic interventions that may disrupt the vicious circle of adiposity and disease progression. © 2019 American Physiological Society. Compr Physiol 9:1411-1429, 2019.
Collapse
Affiliation(s)
- Jumana Saleh
- Biochemistry Department, College of Medicine & Health Sciences, Sultan Qaboos University, Muscat, Oman
| | - Muna Al-Maqbali
- Biochemistry Department, College of Medicine, Sultan Qaboos University, Muscat, Oman
| | | |
Collapse
|
16
|
Chen Y, Lu W, Jin Z, Yu J, Shi B. Carbenoxolone ameliorates hepatic lipid metabolism and inflammation in obese mice induced by high fat diet via regulating the JAK2/STAT3 signaling pathway. Int Immunopharmacol 2019; 74:105498. [DOI: 10.1016/j.intimp.2019.03.011] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2019] [Revised: 02/11/2019] [Accepted: 03/05/2019] [Indexed: 02/06/2023]
|
17
|
Šrámková V, Koc M, Krauzová E, Kračmerová J, Šiklová M, Elkalaf M, Langin D, Štich V, Rossmeislová L. Expression of lipogenic markers is decreased in subcutaneous adipose tissue and adipocytes of older women and is negatively linked to GDF15 expression. J Physiol Biochem 2019; 75:253-262. [PMID: 30912009 DOI: 10.1007/s13105-019-00676-6] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2018] [Accepted: 02/10/2019] [Indexed: 12/25/2022]
Abstract
In aging, the capacity of subcutaneous adipose tissue (SAT) to store lipids decreases and this results in metabolically unfavorable fat redistribution. Triggers of this age-related SAT dysfunction may include cellular senescence or endoplasmic reticulum (ER) stress. Therefore, we compared lipogenic capacity of SAT between young and older women and investigated its relation to senescence and ER stress markers. Samples of SAT and corresponding SAT-derived primary preadipocytes were obtained from two groups of women differing in age (36 vs. 72 years, n = 15 each) but matched for fat mass. mRNA levels of selected genes (lipogenesis: ACACA, FASN, SCD1, DGAT2, ELOVL6; senescence: p16, p21, NOX4, GDF15; ER stress-ATF4, XBP1s, PERK, HSPA5, GADD34, HYOU1, CHOP, EDEM1, DNAJC3) were assessed by qPCR, protein levels of GDF15 by ELISA, and mitochondrial function by the Seahorse Analyzer. Compared to the young, SAT and in vitro differentiated adipocytes from older women exhibited reduced mRNA expression of lipogenic enzymes. Out of analyzed senescence and ER stress markers, the only gene, whose expression correlated negatively with the expression of lipogenic enzymes in both SAT and adipocytes, was GDF15, a marker of not only senescence but also mitochondrial dysfunction. In line with this, inhibition of mitochondrial ATP synthase in adipocytes strongly upregulated GDF15 while reduced expression of lipogenic enzymes. Moreover, adipocytes from older women had a tendency for diminished mitochondrial capacity. Thus, a reduced lipogenic capacity of adipocytes in aged SAT appears to be linked to mitochondrial dysfunction rather than to ER stress or accumulation of senescent cells.
Collapse
Affiliation(s)
- Veronika Šrámková
- Department of Pathophysiology, Third Faculty of Medicine, Charles University, Prague, Czech Republic.,Franco-Czech Laboratory for Clinical Research on Obesity, Third Faculty of Medicine, Prague, Czech Republic
| | - Michal Koc
- Department of Pathophysiology, Third Faculty of Medicine, Charles University, Prague, Czech Republic.,Franco-Czech Laboratory for Clinical Research on Obesity, Third Faculty of Medicine, Prague, Czech Republic
| | - Eva Krauzová
- Department of Pathophysiology, Third Faculty of Medicine, Charles University, Prague, Czech Republic.,Franco-Czech Laboratory for Clinical Research on Obesity, Third Faculty of Medicine, Prague, Czech Republic.,Second Department of Internal Medicine, University Hospital Kralovske Vinohrady, Prague, Czech Republic
| | - Jana Kračmerová
- Department of Pathophysiology, Third Faculty of Medicine, Charles University, Prague, Czech Republic.,Franco-Czech Laboratory for Clinical Research on Obesity, Third Faculty of Medicine, Prague, Czech Republic
| | - Michaela Šiklová
- Department of Pathophysiology, Third Faculty of Medicine, Charles University, Prague, Czech Republic.,Franco-Czech Laboratory for Clinical Research on Obesity, Third Faculty of Medicine, Prague, Czech Republic
| | - Moustafa Elkalaf
- Department of Biochemistry, Cell and Molecular Biology, Third Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Dominique Langin
- Franco-Czech Laboratory for Clinical Research on Obesity, Third Faculty of Medicine, Prague, Czech Republic.,INSERM, UMR1048, Institute of Metabolic and Cardiovascular Diseases, Toulouse, France.,Paul Sabatier University, Toulouse, France.,Department of Clinical Biochemistry, Toulouse University Hospitals, Toulouse, France
| | - Vladimír Štich
- Department of Pathophysiology, Third Faculty of Medicine, Charles University, Prague, Czech Republic.,Franco-Czech Laboratory for Clinical Research on Obesity, Third Faculty of Medicine, Prague, Czech Republic.,Second Department of Internal Medicine, University Hospital Kralovske Vinohrady, Prague, Czech Republic
| | - Lenka Rossmeislová
- Department of Pathophysiology, Third Faculty of Medicine, Charles University, Prague, Czech Republic. .,Franco-Czech Laboratory for Clinical Research on Obesity, Third Faculty of Medicine, Prague, Czech Republic.
| |
Collapse
|
18
|
Baig S, Parvaresh Rizi E, Chia C, Shabeer M, Aung N, Loh TP, Magkos F, Vidal-Puig A, Seet RCS, Khoo CM, Toh SA. Genes Involved in Oxidative Stress Pathways Are Differentially Expressed in Circulating Mononuclear Cells Derived From Obese Insulin-Resistant and Lean Insulin-Sensitive Individuals Following a Single Mixed-Meal Challenge. Front Endocrinol (Lausanne) 2019; 10:256. [PMID: 31068904 PMCID: PMC6491694 DOI: 10.3389/fendo.2019.00256] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/26/2018] [Accepted: 04/02/2019] [Indexed: 01/11/2023] Open
Abstract
Background: Oxidative stress induced by nutritional overload has been linked to the pathogenesis of insulin resistance, which is associated with metabolic syndrome, obesity, type 2 diabetes and diabetic vascular complications. Postprandial changes in expression of oxidative stress pathway genes in obese vs. lean individuals, following intake of different types of meals varying in macronutrient composition have not been characterized to date. Here we aimed to test whether/how oxidative stress responses in obese vs. lean individuals are modulated by meal composition. Methods: High-carbohydrate (HC), high-fat (HF), or high-protein (HP) liquid mixed meals were administered to study subjects (lean insulin-sensitive, n = 9 and obese insulin-resistant, n = 9). Plasma levels of glucose and insulin, lipid profile, urinary F2-isoprostanes (F2-IsoP), and expression levels of genes of oxidative stress pathways were assessed in mononuclear cells (MNC) derived from fresh peripheral blood, at baseline and up to 6-h postprandial states. Differences in these parameters were compared between insulin-sensitive/resistant groups undergoing aforementioned meal challenges. Results: Obese individuals exhibited increased pro-oxidant (i.e., CYBB and CYBA) and anti-oxidant (i.e., TXN RD1) gene expression in the postprandial state, compared with lean subjects, regardless of meal type (P interaction for group × time < 0.05). By contrast, lean subjects had higher expression of NCF-4 gene (pro-oxidant) after HC meal and SOD1 gene (anti-oxidant) after HC and HF meals (P interaction for group × meal < 0.05). There was an increase in postprandial level of urinary F2-IsoP in the obese (P < 0.05) but not lean group. Conclusions: These findings may represent an adaptive oxidative response to mitigate increased stress induced by acute nutritional excess. Further, the results suggest an increased predisposition of obese subjects to oxidative stress. Chronic nutritional excess resulting in increases in body weight and adiposity might lead to decompensation leading to worsening insulin resistance and its sequel. Insights from this study could impact on nutritional recommendations for obese subjects at high-risk of cardiovascular diseases.
Collapse
Affiliation(s)
- Sonia Baig
- Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Ehsan Parvaresh Rizi
- Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Department of Medicine, National University Health System, Singapore, Singapore
| | - Chelsea Chia
- Trinity College Dublin, University of Dublin, Dublin, Ireland
| | - Muhammad Shabeer
- Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Nweni Aung
- Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Tze Ping Loh
- Department of Laboratory Medicine, National University Health System, Singapore, Singapore
| | - Faidon Magkos
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Singapore Institute of Clinical Sciences (SICS), ASTAR, Singapore, Singapore
| | - Antonio Vidal-Puig
- MRC MDU, Metabolic Research Laboratories, University of Cambridge, Cambridge, United Kingdom
| | - Raymond C. S. Seet
- Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Department of Medicine, National University Health System, Singapore, Singapore
| | - Chin Meng Khoo
- Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Department of Medicine, National University Health System, Singapore, Singapore
| | - Sue-Anne Toh
- Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Department of Medicine, National University Health System, Singapore, Singapore
- Programme in Cardiovascular and Metabolic Disorders, Duke-National University of Singapore Graduate Medical School, Singapore, Singapore
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
- *Correspondence: Sue-Anne Toh
| |
Collapse
|
19
|
Maslov LN, Naryzhnaya NV, Boshchenko AA, Popov SV, Ivanov VV, Oeltgen PR. Is oxidative stress of adipocytes a cause or a consequence of the metabolic syndrome? JOURNAL OF CLINICAL AND TRANSLATIONAL ENDOCRINOLOGY 2018; 15:1-5. [PMID: 30479968 PMCID: PMC6240632 DOI: 10.1016/j.jcte.2018.11.001] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/30/2018] [Revised: 11/02/2018] [Accepted: 11/08/2018] [Indexed: 01/17/2023]
Abstract
Metabolic syndrome is accompanied by oxidative stress in animals and humans. The main source of ROS in experimental metabolic syndrome is NADPH oxidase and possibly adipocyte mitochondria. It is now documented that oxidative stress induces insulin resistance of adipocytes and increases secretion of leptin, MCP-1, IL-6, and TNF-α by adipocytes. It was established that oxidative stress induces a decrease in adiponectin production by adipocytes. It has also been shown that obesity itself can induce oxidative stress. Oxidative stress can cause an alteration of intracellular signaling in adipocytes that apparently leads to the formation of insulin resistance of adipocytes. Chronic stress, glucocorticoids, mineralocorticoids, angiotensin-II, TNF-α also play an important role in the pathogenesis of oxidative stress of adipocytes. Oxidative stress is not only a consequence of metabolic syndrome, but also a reason and a foundational link in the pathogenesis of the metabolic syndrome.
Collapse
Affiliation(s)
- Leonid N Maslov
- Cardiology Research Institute, Tomsk National Research Medical Centre, Russian Academy of Sciences, Tomsk, Russia
| | - Natalia V Naryzhnaya
- Cardiology Research Institute, Tomsk National Research Medical Centre, Russian Academy of Sciences, Tomsk, Russia
| | - Alla A Boshchenko
- Cardiology Research Institute, Tomsk National Research Medical Centre, Russian Academy of Sciences, Tomsk, Russia
| | - Sergey V Popov
- Cardiology Research Institute, Tomsk National Research Medical Centre, Russian Academy of Sciences, Tomsk, Russia
| | | | - Peter R Oeltgen
- Department of Pathology, University of Kentucky College of Medicine, Lexington, KY, USA
| |
Collapse
|
20
|
Vitamin D receptor ligands attenuate the inflammatory profile of IL-1β-stimulated human white preadipocytes via modulating the NF-κB and unfolded protein response pathways. Biochem Biophys Res Commun 2018; 503:1049-1056. [DOI: 10.1016/j.bbrc.2018.06.115] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2018] [Accepted: 06/20/2018] [Indexed: 01/31/2023]
|
21
|
Serum adipokine levels and insulin resistance in the first trimester of pregnancy in adolescents and their relationship with neonatal weight. BIOMEDICA 2018; 38:427-436. [PMID: 30335248 DOI: 10.7705/biomedica.v38i4.4035] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/15/2017] [Revised: 03/26/2018] [Indexed: 11/21/2022]
Abstract
Introduction: The approach to the physiology of pregnancy based on adipokine behavior and the homeostasis-insulin resistance (HOMA-IR) model, along with their relationship to neonatal weight, has been poorly studied in adolescent pregnant women.
Objective: To determine possible correlations between adipokines –leptin and adiponectin– and HOMA-IR in pregnant women aged 14 to 17 years, and first-trimester body mass index (BMI) and neonatal weight.
Materials and methods: In the weeks 11 to 14 of gestation, the biochemical variables leptin, adiponectin, glycemia and insulin were measured and HOMA-IR was calculated. Maternal and neonatal anthropometric variables were obtained. Statistical analysis was performed with Pearson correlation and the p value.
Results: We noticed a positive correlation of serum leptin levels with HOMA-IR in the first trimester of gestation (r=0.5, p≤0.000) and a negative correlation between adiponectin and HOMA-IR (r=-0.4; p=0.017), along with positive correlations between BMI and leptin, insulin and HOMA-IR (r=0.83 and p <0.000, r=0.56 and p≤0.000; r=0.54 and p≤0.000, respectively). In adolescent non-obese mothers with no history of dyslipidemia, there was a positive correlation between HOMA-IR and neonatal weight (r=0.43, p=0.012).
Conclusions: Leptin and HOMA-IR showed a positive correlation, while adiponectin and HOMA-IR showed a negative correlation. Leptin and HOMA-IR were positively correlated with BMI. HOMA-IR correlated with the weight of neonates of non-obese adolescents without dyslipidemia.
Collapse
|
22
|
Shioya-Yamada M, Shimada K, Nishitani-Yokoyama M, Sai E, Takeno K, Tamura Y, Watada H, Kawamori R, Daida H, Kawai S. Association Between Visceral Fat Accumulation and Exercise Tolerance in Non-Obese Subjects Without Diabetes. J Clin Med Res 2018; 10:630-635. [PMID: 29977420 PMCID: PMC6031249 DOI: 10.14740/jocmr3403w] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2018] [Accepted: 06/04/2018] [Indexed: 12/21/2022] Open
Abstract
Background We examined the associations between visceral fat accumulation, presence of the components of metabolic syndrome (MetS), and exercise tolerance in non-obese subjects without diabetes. Methods Seventy-four non-obese, non-diabetic Japanese men were enrolled. The subjects were divided into the following two groups: non-obese subjects without any MetS risk factors (n = 38, Group A) and non-obese subjects with one or two MetS risk factors (n = 36, Group B). Anthropometric and metabolic parameters were measured. The response of heart rate (HR) and blood pressure (BP), and exercise tolerance were also evaluated with a cardiopulmonary exercise test using a bicycle ergometer. Results The body mass index, abdominal circumference, visceral fat area, and homeostasis model assessment-insulin resistance, were significantly higher, while levels of anaerobic threshold and maximal oxygen uptake were significantly lower in Group B than in Group A. The levels of resting HR, resting BP, and BP at maximal exercise were significantly higher in Group B than in Group A. There were no significant differences in the HR at maximal exercise as well as the HR and BP after exercise between the two groups. The visceral fat area was significantly and negatively correlated with exercise tolerance. Multivariate linear regression analyses demonstrated that visceral fat area, but not abdominal circumference, was significantly and independently associated with maximal oxygen uptake. Conclusions These data suggest that the visceral fat area is a significant determinant for exercise tolerance even in non-obese subjects without diabetes.
Collapse
Affiliation(s)
- Miki Shioya-Yamada
- Juntendo University Graduate School of Health and Sports Science, Chiba, Japan
| | - Kazunori Shimada
- Juntendo University Graduate School of Health and Sports Science, Chiba, Japan.,Department of Cardiovascular Medicine, Juntendo University Graduate School of Medicine, Tokyo, Japan.,Sportology Center, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Miho Nishitani-Yokoyama
- Department of Cardiovascular Medicine, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Eiryu Sai
- Department of Cardiovascular Medicine, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Kageumi Takeno
- Department of Metabolism and Endocrinology, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Yoshifumi Tamura
- Sportology Center, Juntendo University Graduate School of Medicine, Tokyo, Japan.,Department of Metabolism and Endocrinology, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Hirotaka Watada
- Sportology Center, Juntendo University Graduate School of Medicine, Tokyo, Japan.,Department of Metabolism and Endocrinology, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Ryuzo Kawamori
- Sportology Center, Juntendo University Graduate School of Medicine, Tokyo, Japan.,Department of Metabolism and Endocrinology, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Hiroyuki Daida
- Department of Cardiovascular Medicine, Juntendo University Graduate School of Medicine, Tokyo, Japan.,Sportology Center, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Sachio Kawai
- Juntendo University Graduate School of Health and Sports Science, Chiba, Japan
| |
Collapse
|
23
|
Gourronc FA, Robertson LW, Klingelhutz AJ. A delayed proinflammatory response of human preadipocytes to PCB126 is dependent on the aryl hydrocarbon receptor. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2018; 25:16481-16492. [PMID: 28699004 PMCID: PMC5764822 DOI: 10.1007/s11356-017-9676-z] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/16/2017] [Accepted: 06/27/2017] [Indexed: 05/10/2023]
Abstract
Inflammation in adipose tissue is recognized as a causative factor in the development of type II diabetes. Adipocyte hypertrophy as well as bacterial and environmental factors have been implicated in causing inflammation in mature adipocytes. Exposure to persistent organic pollutants such as polychlorinated biphenyls (PCBs) has been associated with the development of type II diabetes. We show here that PCB126, a dioxin-like PCB, activates a robust proinflammatory state in fat cell precursors (preadipocytes). The response was found to be dependent on aryl hydrocarbon receptor (AhR) activation, although induction of the response was delayed compared to upregulation of CYP1A1, a classic AhR-responsive gene. Treatment of preadipocytes with a nuclear factor kappa-light-chain-enhancer of activated B cell (NF-κB) inhibitor partially attenuated the PCB126-induced inflammatory response and partly, but not completely, ameliorated disruption of adipogenesis caused by PCB126. Our results indicate a role for PCB126 in mediating an inflammatory response through AhR in preadipocytes that interferes with adipogenesis.
Collapse
Affiliation(s)
- Francoise A Gourronc
- Department of Microbiology and Immunology, Carver College of Medicine, University of Iowa, Iowa City, IA, 52242, USA
| | - Larry W Robertson
- Department of Occupational and Environmental Health, College of Public Health, University of Iowa, Iowa City, IA, 52242, USA
| | - Aloysius J Klingelhutz
- Department of Microbiology and Immunology, Carver College of Medicine, University of Iowa, Iowa City, IA, 52242, USA.
- Department of Microbiology and Immunology, Carver College of Medicine, The University of Iowa, 2202 MERF, 375 Newton Road, Iowa City, IA, 52242, USA.
| |
Collapse
|
24
|
Garcia-Contreras C, Vazquez-Gomez M, Torres-Rovira L, Gonzalez J, Porrini E, Gonzalez-Colaço M, Isabel B, Astiz S, Gonzalez-Bulnes A. Characterization of Ageing- and Diet-Related Swine Models of Sarcopenia and Sarcopenic Obesity. Int J Mol Sci 2018. [PMID: 29534532 PMCID: PMC5877684 DOI: 10.3390/ijms19030823] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Sarcopenia and sarcopenic obesity are currently considered major global threats for health and well-being. However, there is a lack of adequate preclinical models for their study. The present trial evaluated the suitability of aged swine by determining changes in adiposity, fatty acids composition, antioxidant status and lipid peroxidation, development of metabolic disturbances and structural changes in tissues and organs. Iberian sows with clinical evidence of aging-related sarcopenia were fed a standard diet fulfilling their maintenance requirements or an obesogenic diet for 100 days. Aging and sarcopenia were related to increased lipid accumulation and cellular dysfunction at both adipose tissue and non-adipose ectopic tissues (liver and pancreas). Obesity concomitant to sarcopenia aggravates the condition by increasing visceral adiposity and causing dyslipidemia, insulin resistance and lipotoxicity in non-adipose tissues. These results support that the Iberian swine model represents certain features of sarcopenia and sarcopenic obesity in humans, paving the way for future research on physiopathology of these conditions and possible therapeutic targets.
Collapse
Affiliation(s)
| | - Marta Vazquez-Gomez
- Faculty of Veterinary Sciences, Universidad Complutense de Madrid, 28040 Madrid, Spain.
| | | | - Jorge Gonzalez
- Micros Veterinaria, Campus de Vegazana, 24007 Leon, Spain.
| | - Esteban Porrini
- Institute of Biomedical Technology (ITB), Universidad de La Laguna, 38200 Tenerife, Spain.
| | - Magali Gonzalez-Colaço
- Central Unit of Clinical Research and Clinical Assays (UCICEC), Universitary Hospital of Canary Island, 28010 Tenerife, Spain.
| | - Beatriz Isabel
- Faculty of Veterinary Sciences, Universidad Complutense de Madrid, 28040 Madrid, Spain.
| | - Susana Astiz
- Comparative Physiology Group, SGIT-INIA, 28040 Madrid, Spain.
| | - Antonio Gonzalez-Bulnes
- Comparative Physiology Group, SGIT-INIA, 28040 Madrid, Spain.
- Faculty of Veterinary Sciences, Universidad Complutense de Madrid, 28040 Madrid, Spain.
| |
Collapse
|
25
|
Cuffe H, Liu M, Key CCC, Boudyguina E, Sawyer JK, Weckerle A, Bashore A, Fried SK, Chung S, Parks JS. Targeted Deletion of Adipocyte Abca1 (ATP-Binding Cassette Transporter A1) Impairs Diet-Induced Obesity. Arterioscler Thromb Vasc Biol 2018; 38:733-743. [PMID: 29348118 DOI: 10.1161/atvbaha.117.309880] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2017] [Accepted: 01/01/2018] [Indexed: 01/23/2023]
Abstract
OBJECTIVE Adipose tissue cholesterol increases with adipocyte triglyceride content and size during development of obesity. However, how adipocyte cholesterol affects adipocyte function is poorly understood. The aim of this study was to evaluate the role of the cellular cholesterol exporter, Abca1 (ATP-binding cassette transporter A1), on adipose tissue function during diet-induced obesity. APPROACH AND RESULTS Adiponectin Cre recombinase transgenic mice were crossed with Abca1flox/flox mice to generate ASKO (adipocyte-specific Abca1 knockout) mice. Control and ASKO mice were then fed a high-fat, high-cholesterol (45% calories as fat and 0.2% cholesterol) diet for 16 weeks. Compared with control mice, ASKO mice had a 2-fold increase in adipocyte plasma membrane cholesterol content and significantly lower body weight, epididymal fat pad weight, and adipocyte size. ASKO versus control adipose tissue had decreased PPARγ (peroxisome proliferator-activated receptor γ) and CCAAT/enhancer-binding protein expression, nuclear SREBP1 (sterol regulatory element-binding protein 1) protein, lipogenesis, and triglyceride accretion but similar Akt activation after acute insulin stimulation. Acute siRNA-mediated Abca1 silencing during 3T3L1 adipocyte differentiation reduced adipocyte Abca1 and PPARγ protein expression and triglyceride content. Systemic stimulated triglyceride lipolysis and glucose homeostasis were similar between control and ASKO mice. CONCLUSIONS Adipocyte Abca1 is a key regulator of adipocyte lipogenesis and lipid accretion, likely because of increased adipose tissue membrane cholesterol, resulting in decreased activation of lipogenic transcription factors PPARγ and SREBP1.
Collapse
Affiliation(s)
- Helen Cuffe
- From the Section on Molecular Medicine, Department of Internal Medicine (H.C., M.L., C.-C.C.K., E.B., J.K.S., A.W., A.B., J.S.P.) and Department of Biochemistry (J.S.P.), Wake Forest School of Medicine, Winston-Salem, NC; Diabetes, Obesity and Metabolism Institute, Icahn School of Medicine at Mount Sinai, New York (S.K.F.); and Department of Nutrition and Health Sciences, University of Nebraska, Lincoln, NE (S.C.)
| | - Mingxia Liu
- From the Section on Molecular Medicine, Department of Internal Medicine (H.C., M.L., C.-C.C.K., E.B., J.K.S., A.W., A.B., J.S.P.) and Department of Biochemistry (J.S.P.), Wake Forest School of Medicine, Winston-Salem, NC; Diabetes, Obesity and Metabolism Institute, Icahn School of Medicine at Mount Sinai, New York (S.K.F.); and Department of Nutrition and Health Sciences, University of Nebraska, Lincoln, NE (S.C.)
| | - Chia-Chi C Key
- From the Section on Molecular Medicine, Department of Internal Medicine (H.C., M.L., C.-C.C.K., E.B., J.K.S., A.W., A.B., J.S.P.) and Department of Biochemistry (J.S.P.), Wake Forest School of Medicine, Winston-Salem, NC; Diabetes, Obesity and Metabolism Institute, Icahn School of Medicine at Mount Sinai, New York (S.K.F.); and Department of Nutrition and Health Sciences, University of Nebraska, Lincoln, NE (S.C.)
| | - Elena Boudyguina
- From the Section on Molecular Medicine, Department of Internal Medicine (H.C., M.L., C.-C.C.K., E.B., J.K.S., A.W., A.B., J.S.P.) and Department of Biochemistry (J.S.P.), Wake Forest School of Medicine, Winston-Salem, NC; Diabetes, Obesity and Metabolism Institute, Icahn School of Medicine at Mount Sinai, New York (S.K.F.); and Department of Nutrition and Health Sciences, University of Nebraska, Lincoln, NE (S.C.)
| | - Janet K Sawyer
- From the Section on Molecular Medicine, Department of Internal Medicine (H.C., M.L., C.-C.C.K., E.B., J.K.S., A.W., A.B., J.S.P.) and Department of Biochemistry (J.S.P.), Wake Forest School of Medicine, Winston-Salem, NC; Diabetes, Obesity and Metabolism Institute, Icahn School of Medicine at Mount Sinai, New York (S.K.F.); and Department of Nutrition and Health Sciences, University of Nebraska, Lincoln, NE (S.C.)
| | - Allison Weckerle
- From the Section on Molecular Medicine, Department of Internal Medicine (H.C., M.L., C.-C.C.K., E.B., J.K.S., A.W., A.B., J.S.P.) and Department of Biochemistry (J.S.P.), Wake Forest School of Medicine, Winston-Salem, NC; Diabetes, Obesity and Metabolism Institute, Icahn School of Medicine at Mount Sinai, New York (S.K.F.); and Department of Nutrition and Health Sciences, University of Nebraska, Lincoln, NE (S.C.)
| | - Alexander Bashore
- From the Section on Molecular Medicine, Department of Internal Medicine (H.C., M.L., C.-C.C.K., E.B., J.K.S., A.W., A.B., J.S.P.) and Department of Biochemistry (J.S.P.), Wake Forest School of Medicine, Winston-Salem, NC; Diabetes, Obesity and Metabolism Institute, Icahn School of Medicine at Mount Sinai, New York (S.K.F.); and Department of Nutrition and Health Sciences, University of Nebraska, Lincoln, NE (S.C.)
| | - Susan K Fried
- From the Section on Molecular Medicine, Department of Internal Medicine (H.C., M.L., C.-C.C.K., E.B., J.K.S., A.W., A.B., J.S.P.) and Department of Biochemistry (J.S.P.), Wake Forest School of Medicine, Winston-Salem, NC; Diabetes, Obesity and Metabolism Institute, Icahn School of Medicine at Mount Sinai, New York (S.K.F.); and Department of Nutrition and Health Sciences, University of Nebraska, Lincoln, NE (S.C.)
| | - Soonkyu Chung
- From the Section on Molecular Medicine, Department of Internal Medicine (H.C., M.L., C.-C.C.K., E.B., J.K.S., A.W., A.B., J.S.P.) and Department of Biochemistry (J.S.P.), Wake Forest School of Medicine, Winston-Salem, NC; Diabetes, Obesity and Metabolism Institute, Icahn School of Medicine at Mount Sinai, New York (S.K.F.); and Department of Nutrition and Health Sciences, University of Nebraska, Lincoln, NE (S.C.)
| | - John S Parks
- From the Section on Molecular Medicine, Department of Internal Medicine (H.C., M.L., C.-C.C.K., E.B., J.K.S., A.W., A.B., J.S.P.) and Department of Biochemistry (J.S.P.), Wake Forest School of Medicine, Winston-Salem, NC; Diabetes, Obesity and Metabolism Institute, Icahn School of Medicine at Mount Sinai, New York (S.K.F.); and Department of Nutrition and Health Sciences, University of Nebraska, Lincoln, NE (S.C.).
| |
Collapse
|
26
|
Du G, Sun J, Zhang Y. Perfluorooctanoic acid impaired glucose homeostasis through affecting adipose AKT pathway. Cytotechnology 2018; 70:479-487. [PMID: 29335808 DOI: 10.1007/s10616-017-0164-6] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2017] [Accepted: 10/31/2017] [Indexed: 12/16/2022] Open
Abstract
Perfluorooctanoic acid (PFOA) is commonly applied in manufactured products, and its potential health risk is concerned greatly. Increasing evidences have indicated PFOA-induced liver dysfunction. However, detailed molecular mechanism has not been completely identified. In this study, we aimed to investigate the mechanical association between PFOA exposure and AKT pathway in white adipose tissue. As results, PFOA-treated mice showed increased blood glucose and insulin levels, and induced insulin resistance. In addition, serum levels of leptin and adiponectin in PFOA-treated mice were elevated. As shown in histological examination, increased cell death counts in PFOA-treated adipose were observed, as well as ultrastructural impairment in adipose cells was found. Further, immunohistochemical stains exhibited GLUT4, p-AKT positive cells were down-regulated in PFOA-treated adipose, while PTEN immune-labeled cells were reduced. In validated data, RT-PCR assay suggested adipose AKT mRNA was down-regulated in PFOA-treated mice, and PTEN mRNA was increased. Western blot data showed that intracellular PTEN protein level in PFOA-treated adipose was up-regulated, while phosphorylation of AKT, GSK3β levels were lowered dose-dependently. Taken together, the present findings indicate that PFOA impaired glucose homeostasis via negatively regulating AKT pathway in white adipose tissue.
Collapse
Affiliation(s)
- Gang Du
- Department of Endocrinology, Affiliated Hospital of Weifang Medical University, No 2428 Yuhe Road of Kuiwen District, Weifang, 261031, Shandong, China
| | - Jinhong Sun
- Department of Endocrinology, Affiliated Hospital of Weifang Medical University, No 2428 Yuhe Road of Kuiwen District, Weifang, 261031, Shandong, China
| | - Yang Zhang
- Department of Endocrinology, Affiliated Hospital of Weifang Medical University, No 2428 Yuhe Road of Kuiwen District, Weifang, 261031, Shandong, China.
| |
Collapse
|
27
|
Klingelhutz AJ, Gourronc FA, Chaly A, Wadkins DA, Burand AJ, Markan KR, Idiga SO, Wu M, Potthoff MJ, Ankrum JA. Scaffold-free generation of uniform adipose spheroids for metabolism research and drug discovery. Sci Rep 2018; 8:523. [PMID: 29323267 PMCID: PMC5765134 DOI: 10.1038/s41598-017-19024-z] [Citation(s) in RCA: 88] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2017] [Accepted: 12/20/2017] [Indexed: 02/06/2023] Open
Abstract
Adipose tissue dysfunction is critical to the development of type II diabetes and other metabolic diseases. While monolayer cell culture has been useful for studying fat biology, 2D culture often does not reflect the complexity of fat tissue. Animal models are also problematic in that they are expensive, time consuming, and may not completely recapitulate human biology because of species variation. To address these problems, we have developed a scaffold-free method to generate 3D adipose spheroids from primary or immortal human or mouse pre-adipocytes. Pre-adipocytes self-organize into spheroids in hanging drops and upon transfer to low attachment plates, can be maintained in long-term cultures. Upon exposure to differentiation cues, the cells mature into adipocytes, accumulating large lipid droplets that expand with time. The 3D spheroids express and secrete higher levels of adiponectin compared to 2D culture and respond to stress, either culture-related or toxin-associated, by secreting pro-inflammatory adipokines. In addition, 3D spheroids derived from brown adipose tissue (BAT) retain expression of BAT markers better than 2D cultures derived from the same tissue. Thus, this model can be used to study both the maturation of pre-adipocytes or the function of mature adipocytes in a 3D culture environment.
Collapse
Affiliation(s)
- Aloysius J Klingelhutz
- University of Iowa Fraternal Order of Eagles Diabetes Research Center, 169 Newton Rd, Iowa City, IA, 52242, USA. .,Department of Microbiology, Carver College of Medicine, University of Iowa, Iowa City, IA, 52242, USA.
| | - Francoise A Gourronc
- Department of Microbiology, Carver College of Medicine, University of Iowa, Iowa City, IA, 52242, USA
| | - Anna Chaly
- Department of Microbiology, Carver College of Medicine, University of Iowa, Iowa City, IA, 52242, USA
| | - David A Wadkins
- University of Iowa Fraternal Order of Eagles Diabetes Research Center, 169 Newton Rd, Iowa City, IA, 52242, USA.,Department of Biomedical Engineering, University of Iowa, Iowa City, IA, 52242, USA
| | - Anthony J Burand
- University of Iowa Fraternal Order of Eagles Diabetes Research Center, 169 Newton Rd, Iowa City, IA, 52242, USA.,Department of Biomedical Engineering, University of Iowa, Iowa City, IA, 52242, USA
| | - Kathleen R Markan
- University of Iowa Fraternal Order of Eagles Diabetes Research Center, 169 Newton Rd, Iowa City, IA, 52242, USA.,Department of Pharmacology, Carver College of Medicine, University of Iowa, Iowa City, IA, 52242, USA
| | - Sharon O Idiga
- University of Iowa Fraternal Order of Eagles Diabetes Research Center, 169 Newton Rd, Iowa City, IA, 52242, USA.,Department of Pharmacology, Carver College of Medicine, University of Iowa, Iowa City, IA, 52242, USA
| | - Meng Wu
- Department of Biochemistry, Carver College of Medicine, University of Iowa, Iowa City, IA, 52242, USA.,Division of Medicinal and Natural Products Chemistry, Department of Pharmaceutical Sciences and Experimental Therapeutics, College of Pharmacy, University of Iowa, 115 S. Grand Ave, Iowa City, IA, 52242, USA.,High Throughput Screening Core Facility at University of Iowa (UIHTS), University of Iowa, 115 S. Grand Ave, Iowa City, IA, 52242, USA
| | - Matthew J Potthoff
- University of Iowa Fraternal Order of Eagles Diabetes Research Center, 169 Newton Rd, Iowa City, IA, 52242, USA.,Department of Pharmacology, Carver College of Medicine, University of Iowa, Iowa City, IA, 52242, USA
| | - James A Ankrum
- University of Iowa Fraternal Order of Eagles Diabetes Research Center, 169 Newton Rd, Iowa City, IA, 52242, USA. .,Department of Biomedical Engineering, University of Iowa, Iowa City, IA, 52242, USA.
| |
Collapse
|
28
|
Gertow J, Ng CZ, Mamede Branca RM, Werngren O, Du L, Kjellqvist S, Hemmingsson P, Bruchfeld A, MacLaughlin H, Eriksson P, Axelsson J, Fisher RM. Altered Protein Composition of Subcutaneous Adipose Tissue in Chronic Kidney Disease. Kidney Int Rep 2017; 2:1208-1218. [PMID: 29270529 PMCID: PMC5733748 DOI: 10.1016/j.ekir.2017.07.007] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2017] [Revised: 06/30/2017] [Accepted: 07/24/2017] [Indexed: 12/26/2022] Open
Abstract
Introduction Loss of renal function is associated with high mortality from cardiovascular disease (CVD). Patients with chronic kidney disease (CKD) have altered circulating adipokine and nonesterified fatty acid concentrations and insulin resistance, which are features of disturbed adipose tissue metabolism. Because dysfunctional adipose tissue contributes to the development of CVD, we hypothesize that adipose tissue dysfunctionality in patients with CKD could explain, at least in part, their high rates of CVD. Therefore we characterized adipose tissue from patients with CKD, in comparison to healthy controls, to search for signs of dysfunctionality. Methods Biopsy samples of subcutaneous adipose tissue from 16 CKD patients and 11 healthy controls were analyzed for inflammation, fibrosis, and adipocyte size. Protein composition was assessed using 2-dimensional gel proteomics combined with multivariate analysis. Results Adipose tissue of CKD patients contained significantly more CD68-positive cells, but collagen content did not differ. Adipocyte size was significantly smaller in CKD patients. Proteomic analysis of adipose tissue revealed significant differences in the expression of certain proteins between the groups. Proteins whose expression differed the most were α-1-microglobulin/bikunin precursor (AMBP, higher in CKD) and vimentin (lower in CKD). Vimentin is a lipid droplet−associated protein, and changes in its expression may impair fatty acid storage/mobilization in adipose tissue, whereas high levels of AMBP may reflect oxidative stress. Discussion These findings demonstrate that adipose tissue of CKD patients shows signs of inflammation and disturbed functionality, thus potentially contributing to the unfavorable metabolic profile and increased risk of CVD in these patients.
Collapse
Affiliation(s)
- Joanna Gertow
- Cardiovascular Medicine Unit, Department of Medicine Solna, Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Chang Zhi Ng
- Cardiovascular Medicine Unit, Department of Medicine Solna, Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Rui Miguel Mamede Branca
- Clinical Proteomics Mass Spectrometry, Department of Oncology-Pathology, Science For Life Laboratory and Karolinska Institutet, Stockholm, Sweden
| | - Olivera Werngren
- Cardiovascular Medicine Unit, Department of Medicine Solna, Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Lei Du
- Cardiovascular Medicine Unit, Department of Medicine Solna, Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Sanela Kjellqvist
- Department of Biochemistry and Biophysics, Science for Life Laboratory, Stockholm University, Stockholm, Sweden
| | - Peter Hemmingsson
- Department of Clinical Science, Intervention and Technology (CLINTEC), Karolinska Institutet, Stockholm, Sweden
| | - Annette Bruchfeld
- Department of Clinical Science, Intervention and Technology (CLINTEC), Karolinska Institutet, Stockholm, Sweden
| | - Helen MacLaughlin
- Division of Diabetes and Nutritional Sciences, King’s College London and King’s College Hospital, London, United Kingdom
| | - Per Eriksson
- Cardiovascular Medicine Unit, Department of Medicine Solna, Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Jonas Axelsson
- Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| | - Rachel M. Fisher
- Cardiovascular Medicine Unit, Department of Medicine Solna, Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
- Correspondence: Rachel M. Fisher, Cardiovascular Medicine Unit, Department of Medicine Solna, Karolinska Institutet, Center for Molecular Medicine, Karolinska University Hospital (L8:03), 171 76 Stockholm, Sweden.Cardiovascular Medicine UnitDepartment of Medicine SolnaKarolinska InstitutetCenter for Molecular MedicineKarolinska University Hospital (L8:03)171 76 StockholmSweden
| |
Collapse
|
29
|
Spahis S, Borys JM, Levy E. Metabolic Syndrome as a Multifaceted Risk Factor for Oxidative Stress. Antioxid Redox Signal 2017; 26:445-461. [PMID: 27302002 DOI: 10.1089/ars.2016.6756] [Citation(s) in RCA: 84] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
SIGNIFICANCE Metabolic syndrome (MetS) is associated with a greater risk of diabetes and cardiovascular diseases. It is estimated that this multifactorial condition affects 20%-30% of the world's population. A detailed understanding of MetS mechanisms is crucial for the development of effective prevention strategies and adequate intervention tools that could curb its increasing prevalence and limit its comorbidities, particularly in younger age groups. With advances in basic redox biology, oxidative stress (OxS) involvement in the complex pathophysiology of MetS has become widely accepted. Nevertheless, its clear association with and causative effects on MetS require further elucidation. Recent Advances: Although a better understanding of the causes, risks, and effects of MetS is essential, studies suggest that oxidant/antioxidant imbalance is a key contributor to this condition. OxS is now understood to be a major underlying mechanism for mitochondrial dysfunction, ectopic lipid accumulation, and gut microbiota impairment. CRITICAL ISSUES Further studies, particularly in the field of translational research, are clearly required to understand and control the production of reactive oxygen species (ROS) levels, especially in the mitochondria, since the various therapeutic trials conducted to date have not targeted this major ROS-generating system, aimed to delay MetS onset, or prevent its progression. FUTURE DIRECTIONS Multiple relevant markers need to be identified to clarify the role of ROS in the etiology of MetS. Future clinical trials should provide important proof of concept for the effectiveness of antioxidants as useful therapeutic approaches to simultaneously counteract mitochondrial OxS, alleviate MetS symptoms, and prevent complications. Antioxid. Redox Signal. 26, 445-461.
Collapse
Affiliation(s)
- Schohraya Spahis
- 1 Research Center , Ste-Justine MUHC, Montreal, Canada .,2 Department of Nutrition, Université de Montréal , Montreal, Canada
| | | | - Emile Levy
- 1 Research Center , Ste-Justine MUHC, Montreal, Canada .,2 Department of Nutrition, Université de Montréal , Montreal, Canada .,3 EPODE International Network , Paris, France
| |
Collapse
|
30
|
von Frankenberg AD, Marina A, Song X, Callahan HS, Kratz M, Utzschneider KM. A high-fat, high-saturated fat diet decreases insulin sensitivity without changing intra-abdominal fat in weight-stable overweight and obese adults. Eur J Nutr 2017; 56:431-443. [PMID: 26615402 PMCID: PMC5291812 DOI: 10.1007/s00394-015-1108-6] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2015] [Accepted: 11/16/2015] [Indexed: 01/22/2023]
Abstract
PURPOSE We sought to determine the effects of dietary fat on insulin sensitivity and whether changes in insulin sensitivity were explained by changes in abdominal fat distribution or very low-density lipoprotein (VLDL) fatty acid composition. METHODS Overweight/obese adults with normal glucose tolerance consumed a control diet (35 % fat/12 % saturated fat/47 % carbohydrate) for 10 days, followed by a 4-week low-fat diet (LFD, n = 10: 20 % fat/8 % saturated fat/62 % carbohydrate) or high-fat diet (HFD, n = 10: 55 % fat/25 % saturated fat/27 % carbohydrate). All foods and their eucaloric energy content were provided. Insulin sensitivity was measured by labeled hyperinsulinemic-euglycemic clamps, abdominal fat distribution by MRI, and fasting VLDL fatty acids by gas chromatography. RESULTS The rate of glucose disposal (Rd) during low- and high-dose insulin decreased on the HFD but remained unchanged on the LFD (Rd-low: LFD: 0.12 ± 0.11 vs. HFD: -0.37 ± 0.15 mmol/min, mean ± SE, p < 0.01; Rd-high: LFD: 0.11 ± 0.37 vs. HFD: -0.71 ± 0.26 mmol/min, p = 0.08). Hepatic insulin sensitivity did not change. Changes in subcutaneous fat were positively associated with changes in insulin sensitivity on the LFD (r = 0.78, p < 0.01) with a trend on the HFD (r = 0.60, p = 0.07), whereas there was no association with intra-abdominal fat. The LFD led to an increase in VLDL palmitic (16:0), stearic (18:0), and palmitoleic (16:1n7c) acids, while no changes were observed on the HFD. Changes in VLDL n-6 docosapentaenoic acid (22:5n6) were strongly associated with changes in insulin sensitivity on both diets (LFD: r = -0.77; p < 0.01; HFD: r = -0.71; p = 0.02). CONCLUSIONS A diet very high in fat and saturated fat adversely affects insulin sensitivity and thereby might contribute to the development of type 2 diabetes. CLINICALTRIALS. GOV IDENTIFIER NCT00930371.
Collapse
Affiliation(s)
- Anize D von Frankenberg
- Division of Metabolism, Endocrinology and Nutrition, Department of Medicine, University of Washington, 1959 NE Pacific Street, Seattle, WA, 98195, USA.
- Post-Graduate Endocrinology Program, School of Medicine, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil.
- Endocrinology, VA Puget Sound Health Care System, 1660 S Columbian Way (151), Seattle, WA, 98108, USA.
| | - Anna Marina
- Division of Metabolism, Endocrinology and Nutrition, Department of Medicine, University of Washington, 1959 NE Pacific Street, Seattle, WA, 98195, USA
| | - Xiaoling Song
- Division of Public Health Sciences, Fred Hutchinson Cancer Research Center, 1100 Fairview Ave N, Seattle, WA, 98109, USA
- Department of Epidemiology, University of Washington, 1959 NE Pacific Street, Seattle, WA, 98195, USA
| | - Holly S Callahan
- School of Medicine, University of Washington, 1959 NE Pacific Street, Seattle, WA, 98195, USA
| | - Mario Kratz
- Division of Metabolism, Endocrinology and Nutrition, Department of Medicine, University of Washington, 1959 NE Pacific Street, Seattle, WA, 98195, USA
- Division of Public Health Sciences, Fred Hutchinson Cancer Research Center, 1100 Fairview Ave N, Seattle, WA, 98109, USA
- Department of Epidemiology, University of Washington, 1959 NE Pacific Street, Seattle, WA, 98195, USA
| | - Kristina M Utzschneider
- Division of Metabolism, Endocrinology and Nutrition, Department of Medicine, University of Washington, 1959 NE Pacific Street, Seattle, WA, 98195, USA
- Endocrinology, VA Puget Sound Health Care System, 1660 S Columbian Way (151), Seattle, WA, 98108, USA
| |
Collapse
|
31
|
Rabadán-Chávez GM, Reyes-Maldonado E, Quevedo-Corona L, Paniagua-Castro N, Escalona-Cardoso G, Jaramillo-Flores ME. The prothrombotic state associated with obesity-induced hypertension is reduced by cocoa and its main flavanols. Food Funct 2016; 7:4880-4888. [DOI: 10.1039/c6fo01165a] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Cocoa flavanols could ameliorate cardiovascular health in obese patients.
Collapse
Affiliation(s)
- G. M. Rabadán-Chávez
- Escuela Nacional de Ciencias Biológicas
- Instituto Politécnico Nacional-Wilfrido Massiew s/n esq
- Manuel Stampa
- Unidad Profesional Adolfo López Mateos
- C.P. 07738 Ciudad de México
| | - E. Reyes-Maldonado
- Escuela Nacional de Ciencias Biológicas
- Instituto Politécnico Nacional-Wilfrido Massiew s/n esq
- Manuel Stampa
- Unidad Profesional Adolfo López Mateos
- C.P. 07738 Ciudad de México
| | - L. Quevedo-Corona
- Escuela Nacional de Ciencias Biológicas
- Instituto Politécnico Nacional-Wilfrido Massiew s/n esq
- Manuel Stampa
- Unidad Profesional Adolfo López Mateos
- C.P. 07738 Ciudad de México
| | - N. Paniagua-Castro
- Escuela Nacional de Ciencias Biológicas
- Instituto Politécnico Nacional-Wilfrido Massiew s/n esq
- Manuel Stampa
- Unidad Profesional Adolfo López Mateos
- C.P. 07738 Ciudad de México
| | - G. Escalona-Cardoso
- Escuela Nacional de Ciencias Biológicas
- Instituto Politécnico Nacional-Wilfrido Massiew s/n esq
- Manuel Stampa
- Unidad Profesional Adolfo López Mateos
- C.P. 07738 Ciudad de México
| | - M. E. Jaramillo-Flores
- Escuela Nacional de Ciencias Biológicas
- Instituto Politécnico Nacional-Wilfrido Massiew s/n esq
- Manuel Stampa
- Unidad Profesional Adolfo López Mateos
- C.P. 07738 Ciudad de México
| |
Collapse
|
32
|
Lazra Y, Falach A, Frenkel L, Rozenberg K, Sampson S, Rosenzweig T. Autocrine/paracrine function of globular adiponectin: inhibition of lipid metabolism and inflammatory response in 3T3-L1 adipocytes. J Cell Biochem 2015; 116:754-66. [PMID: 25491932 DOI: 10.1002/jcb.25031] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2014] [Accepted: 12/04/2014] [Indexed: 12/15/2022]
Abstract
Adiponectin is an adipose-derived hormone, with beneficial effects on insulin sensitivity and inflammation. The aim of this study was to clarify the autocrine/paracrine effects of globular adiponectin (gAd) administrated during differentiation on the function of the mature adipocytes. Experiments were performed on 3T3-L1 preadipocytes treated with gAd (10 nM), starting at an early stage of differentiation. gAd treatment during differentiation was without effect on mRNA expression of adiponectin and AdipoR2, but increased AdipoR1 expression. PPARgamma, perillipin and FABP4 mRNA expressions were lower in gAd-treated adipocytes, accompanied by a reduction in lipid accumulation. While mRNA expression of HSL was not affected by gAd compared to untreated adipocytes, both ATGL and FAS were reduced, indicating that gAd regulates both lipolysis and lipogenesis. PPARα, ACOX2 and UCPs mRNA expressions were not affected by gAd, indicating that the reduction in lipid content is not attributed to an increase in fatty-acid oxidation. In accord with the lower PPARγ expression, there was reduced Glut4 mRNA expression; however, insulin-induced PKB phosphorylation was enhanced by gAd, and glucose uptake was not altered. To investigate the effect of gAd on LPS-induced inflammatory response, cells were treated with gAd either during differentiation or 24 h before induction of inflammation in differentiated adipocytes. LPS-induced inflammatory response, as indicated by increase in IL6 and MCP-1 mRNA expression. gAd given through differentiation was much more effective in abrogating LPS-dependent cytokines production than gAd given to the mature adipocyte. In conclusion, elevated gAd at differentiation of 3T3-L1 cells leads to reduced lipid content, reduced lipid metabolism and high resistance to inflammation.
Collapse
Affiliation(s)
- Yulia Lazra
- Departments of Molecular Biology and Nutrition, Ariel University, Ariel, 40700, Israel; Mina and Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Ramat-Gan, Israel
| | | | | | | | | | | |
Collapse
|
33
|
Geng T, Sutter A, Harland MD, Law BA, Ross JS, Lewin D, Palanisamy A, Russo SB, Chavin KD, Cowart LA. SphK1 mediates hepatic inflammation in a mouse model of NASH induced by high saturated fat feeding and initiates proinflammatory signaling in hepatocytes. J Lipid Res 2015; 56:2359-71. [PMID: 26482537 DOI: 10.1194/jlr.m063511] [Citation(s) in RCA: 65] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2015] [Indexed: 11/20/2022] Open
Abstract
Steatohepatitis occurs in up to 20% of patients with fatty liver disease and leads to its primary disease outcomes, including fibrosis, cirrhosis, and increased risk of hepatocellular carcinoma. Mechanisms that mediate this inflammation are of major interest. We previously showed that overload of saturated fatty acids, such as that which occurs with metabolic syndrome, induced sphingosine kinase 1 (SphK1), an enzyme that generates sphingosine-1-phosphate (S1P). While data suggest beneficial roles for S1P in some contexts, we hypothesized that it may promote hepatic inflammation in the context of obesity. Consistent with this, we observed 2-fold elevation of this enzyme in livers from humans with nonalcoholic fatty liver disease and also in mice with high saturated fat feeding, which recapitulated the human disease. Mice exhibited activation of NFκB, elevated cytokine production, and immune cell infiltration. Importantly, SphK1-null mice were protected from these outcomes. Studies in cultured cells demonstrated saturated fatty acid induction of SphK1 message, protein, and activity, and also a requirement of the enzyme for NFκB signaling and increased mRNA encoding TNFα and MCP1. Moreover, saturated fat-induced NFκB signaling and elevation of TNFα and MCP1 mRNA in HepG2 cells was blocked by targeted knockdown of S1P receptor 1, supporting a role for this lipid signaling pathway in inflammation in nonalcoholic fatty liver disease.
Collapse
Affiliation(s)
- Tuoyu Geng
- Departments of Biochemistry and Molecular Biology, Medical University of South Carolina, Charleston, SC 29425
| | - Alton Sutter
- Surgery, Medical University of South Carolina, Charleston, SC 29425
| | - Michael D Harland
- Departments of Biochemistry and Molecular Biology, Medical University of South Carolina, Charleston, SC 29425
| | - Brittany A Law
- Departments of Biochemistry and Molecular Biology, Medical University of South Carolina, Charleston, SC 29425 Ralph H. Johnson Veterans Affairs Medical Center, Charleston, SC 29403
| | - Jessica S Ross
- Departments of Biochemistry and Molecular Biology, Medical University of South Carolina, Charleston, SC 29425
| | - David Lewin
- Pathology and Laboratory Medicine, Medical University of South Carolina, Charleston, SC 29425
| | - Arun Palanisamy
- Surgery, Medical University of South Carolina, Charleston, SC 29425
| | - Sarah B Russo
- Departments of Biochemistry and Molecular Biology, Medical University of South Carolina, Charleston, SC 29425
| | - Kenneth D Chavin
- Surgery, Medical University of South Carolina, Charleston, SC 29425
| | - L Ashley Cowart
- Departments of Biochemistry and Molecular Biology, Medical University of South Carolina, Charleston, SC 29425 Ralph H. Johnson Veterans Affairs Medical Center, Charleston, SC 29403
| |
Collapse
|
34
|
Xu P, Fischer-Posovszky P, Bischof J, Radermacher P, Wabitsch M, Henne-Bruns D, Wolf AM, Hillenbrand A, Knippschild U. Gene expression levels of Casein kinase 1 (CK1) isoforms are correlated to adiponectin levels in adipose tissue of morbid obese patients and site-specific phosphorylation mediated by CK1 influences multimerization of adiponectin. Mol Cell Endocrinol 2015; 406:87-101. [PMID: 25724478 DOI: 10.1016/j.mce.2015.02.010] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/15/2014] [Revised: 01/29/2015] [Accepted: 02/11/2015] [Indexed: 01/03/2023]
Abstract
White adipose tissue has now been recognized as an important endocrine organ secreting bioactive molecules termed adipocytokines. In obesity, anti-inflammatory adipocytokines like adiponectin are decreased while pro-inflammatory factors are over-produced. These changes contribute to the development of insulin resistance and obesity-associated diseases. Since members of the casein kinase 1 (CK1) family are involved in the regulation of various signaling pathways we ask here whether they are able to modulate the functions of adiponectin. We show that CK1δ and ε are expressed in adipose tissue and that the expression of CK1 isoforms correlates with that of adiponectin. Furthermore, adiponectin co-immunoprecipitates with CK1δ and CK1ε and is phosphorylated by CK1δ at serine 174 and threonine 235, thereby influencing the formation of adiponectin oligomeric complexes. Furthermore, inhibition of CK1δ in human adipocytes by IC261 leads to an increase in basal and insulin-stimulated glucose uptake. In summary, our data indicate that site-specific phosphorylation of adiponectin, especially at sites targeted by CK1δ in vitro, provides an additional regulatory mechanism for modulating adiponectin complex formation and function.
Collapse
Affiliation(s)
- Pengfei Xu
- Department of General and Visceral Surgery, University of Ulm, Albert-Einstein-Allee 23, 89081 Ulm, Germany
| | - Pamela Fischer-Posovszky
- Divison of Pediatric Endocrinology and Diabetes, Department of Pediatrics and Adolescent Medicine, University of Ulm, Eythstrasse 24, 89075 Ulm, Germany
| | - Joachim Bischof
- Department of General and Visceral Surgery, University of Ulm, Albert-Einstein-Allee 23, 89081 Ulm, Germany
| | - Peter Radermacher
- Institute of Pathophysiology and Process Development in Anesthesia, University of Ulm, Helmholtzstrasse 8/1, 89081 Ulm, Germany
| | - Martin Wabitsch
- Divison of Pediatric Endocrinology and Diabetes, Department of Pediatrics and Adolescent Medicine, University of Ulm, Eythstrasse 24, 89075 Ulm, Germany
| | - Doris Henne-Bruns
- Department of General and Visceral Surgery, University of Ulm, Albert-Einstein-Allee 23, 89081 Ulm, Germany
| | - Anna-Maria Wolf
- Department of General and Visceral Surgery, University of Ulm, Albert-Einstein-Allee 23, 89081 Ulm, Germany
| | - Andreas Hillenbrand
- Department of General and Visceral Surgery, University of Ulm, Albert-Einstein-Allee 23, 89081 Ulm, Germany
| | - Uwe Knippschild
- Department of General and Visceral Surgery, University of Ulm, Albert-Einstein-Allee 23, 89081 Ulm, Germany.
| |
Collapse
|
35
|
The effect of N-stearoylethanolamine on cholesterol content, fatty acid composition and protein carbonylation level in rats with alimentary obesity-induced insulin resistance. UKRAINIAN BIOCHEMICAL JOURNAL 2014. [DOI: 10.15407/ubj86.06.119] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
|
36
|
Peinado JR, Diaz-Ruiz A, Frühbeck G, Malagon MM. Mitochondria in metabolic disease: getting clues from proteomic studies. Proteomics 2014; 14:452-66. [PMID: 24339000 DOI: 10.1002/pmic.201300376] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2013] [Revised: 11/08/2013] [Accepted: 11/21/2013] [Indexed: 01/11/2023]
Abstract
Mitochondria play a key role as major regulators of cellular energy homeostasis, but in the context of mitochondrial dysfunction, mitochondria may generate reactive oxidative species and induce cellular apoptosis. Indeed, altered mitochondrial status has been linked to the pathogenesis of several metabolic disorders and specially disorders related to insulin resistance, such as obesity, type 2 diabetes, and other comorbidities comprising the metabolic syndrome. In the present review, we summarize information from various mitochondrial proteomic studies of insulin-sensitive tissues under different metabolic states. To that end, we first focus our attention on the pancreas, as mitochondrial malfunction has been shown to contribute to beta cell failure and impaired insulin release. Furthermore, proteomic studies of mitochondria obtained from liver, muscle, and adipose tissue are summarized, as these tissues constitute the primary insulin target metabolic tissues. Since recent advances in proteomic techniques have exposed the importance of PTMs in the development of metabolic disease, we also present information on specific PTMs that may directly affect mitochondria during the pathogenesis of metabolic disease. Specifically, mitochondrial protein acetylation, phosphorylation, and other PTMs related to oxidative damage, such as nitrosylation and carbonylation, are discussed.
Collapse
Affiliation(s)
- Juan R Peinado
- Department of Medical Sciences, Faculty of Medicine, Ciudad Real, Spain
| | | | | | | |
Collapse
|
37
|
Gadupudi G, Gourronc FA, Ludewig G, Robertson LW, Klingelhutz AJ. PCB126 inhibits adipogenesis of human preadipocytes. Toxicol In Vitro 2014; 29:132-41. [PMID: 25304490 DOI: 10.1016/j.tiv.2014.09.015] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2014] [Revised: 08/28/2014] [Accepted: 09/25/2014] [Indexed: 12/19/2022]
Abstract
Emerging evidence indicates that persistent organic pollutants (POPs), including polychlorinated biphenyls (PCBs), are involved in the development of diabetes. Dysfunctional adipocytes play a significant role in initiating insulin resistance. Preadipocytes make up a large portion of adipose tissue and are necessary for the generation of functional mature adipocytes through adipogenesis. PCB126 is a dioxin-like PCB and a potent aryl hydrocarbon receptor (AhR) agonist. We hypothesized that PCB126 may be involved in the development of diabetes through disruption of adipogenesis. Using a newly developed human preadipocyte cell line called NPAD (Normal PreADipocytes), we found that exposure of preadipocytes to PCB126 resulted in significant reduction in their subsequent ability to fully differentiate into adipocytes, more so than when the cells were exposed to PCB126 during differentiation. Reduction in differentiation by PCB126 was associated with downregulation of transcript levels of a key adipocyte transcription factor, PPARγ, and late adipocyte differentiation genes. An AhR antagonist, CH223191, blocked this effect. These studies indicate that preadipocytes are particularly sensitive to the effects of PCB126 and suggest that AhR activation inhibits PPARγ transcription and subsequent adipogenesis. Our results validate the NPAD cell line as a useful model for studying the effects of POPs on adipogenesis.
Collapse
Affiliation(s)
- Gopi Gadupudi
- Interdisciplinary Graduate Program in Human Toxicology, The University of Iowa, Iowa City, IA 52242, United States; Department of Occupational & Environmental Health, The University of Iowa, Iowa City, IA 52242, United States
| | - Francoise A Gourronc
- Department of Microbiology, The University of Iowa, Iowa City, IA 52242, United States
| | - Gabriele Ludewig
- Interdisciplinary Graduate Program in Human Toxicology, The University of Iowa, Iowa City, IA 52242, United States; Department of Occupational & Environmental Health, The University of Iowa, Iowa City, IA 52242, United States
| | - Larry W Robertson
- Interdisciplinary Graduate Program in Human Toxicology, The University of Iowa, Iowa City, IA 52242, United States; Department of Occupational & Environmental Health, The University of Iowa, Iowa City, IA 52242, United States
| | | |
Collapse
|
38
|
Alteration of NCoR corepressor splicing in mice causes increased body weight and hepatosteatosis without glucose intolerance. Mol Cell Biol 2014; 34:4104-14. [PMID: 25182530 DOI: 10.1128/mcb.00554-14] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Alternative mRNA splicing is an important means of diversifying function in higher eukaryotes. Notably, both NCoR and SMRT corepressors are subject to alternative mRNA splicing, yielding a series of distinct corepressor variants with highly divergent functions. Normal adipogenesis is associated with a switch in corepressor splicing from NCoRω to NCoRδ, which appears to help regulate this differentiation process. We report here that mimicking this development switch in mice by a splice-specific whole-animal ablation of NCoRω is very different from a whole-animal or tissue-specific total NCoR knockout and produces significantly enhanced weight gain on a high-fat diet. Surprisingly, NCoRω(-/-) mice are protected against diet-induced glucose intolerance despite enhanced adiposity and the presence of multiple additional, prodiabetic phenotypic changes. Our results indicate that the change in NCoR splicing during normal development both helps drive normal adipocyte differentiation and plays a key role in determining a metabolically appropriate storage of excess calories. We also conclude that whole-gene "knockouts" fail to reveal how important gene products are customized, tailored, and adapted through alternative mRNA splicing and thus do not reveal all the functions of the protein products of that gene.
Collapse
|
39
|
Chabot F, Caron A, Laplante M, St-Pierre DH. Interrelationships between ghrelin, insulin and glucose homeostasis: Physiological relevance. World J Diabetes 2014; 5:328-341. [PMID: 24936254 PMCID: PMC4058737 DOI: 10.4239/wjd.v5.i3.328] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/09/2013] [Accepted: 05/08/2014] [Indexed: 02/05/2023] Open
Abstract
Ghrelin is a 28 amino acid peptide mainly derived from the oxyntic gland of the stomach. Both acylated (AG) and unacylated (UAG) forms of ghrelin are found in the circulation. Initially, AG was considered as the only bioactive form of ghrelin. However, recent advances indicate that both AG and UAG exert distinct and common effects in organisms. Soon after its discovery, ghrelin was shown to promote appetite and adiposity in animal and human models. In response to these anabolic effects, an impressive number of elements have suggested the influence of ghrelin on the regulation of metabolic functions and the development of obesity-related disorders. However, due to the complexity of its biochemical nature and the physiological processes it governs, some of the effects of ghrelin are still debated in the literature. Evidence suggests that ghrelin influences glucose homeostasis through the modulation of insulin secretion and insulin receptor signaling. On the other hand, insulin was also shown to influence circulating levels of ghrelin. Here, we review the relationship between ghrelin and insulin and we describe the impact of this interaction on the modulation of glucose homeostasis.
Collapse
|
40
|
Hage Hassan R, Bourron O, Hajduch E. Defect of insulin signal in peripheral tissues: Important role of ceramide. World J Diabetes 2014; 5:244-257. [PMID: 24936246 PMCID: PMC4058729 DOI: 10.4239/wjd.v5.i3.244] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/12/2013] [Revised: 01/29/2014] [Accepted: 05/08/2014] [Indexed: 02/05/2023] Open
Abstract
In healthy people, balance between glucose production and its utilization is precisely controlled. When circulating glucose reaches a critical threshold level, pancreatic β cells secrete insulin that has two major actions: to lower circulating glucose levels by facilitating its uptake mainly into skeletal muscle while inhibiting its production by the liver. Interestingly, dietary triglycerides are the main source of fatty acids to fulfill energy needs of oxidative tissues. Normally, the unconsumed fraction of excess of fatty acids is stored in lipid droplets that are localized in adipocytes to provide energy during fasting periods. Thus, adipose tissue acts as a trap for fatty acid excess liberated from plasma triglycerides. When the buffering action of adipose tissue to store fatty acids is impaired, fatty acids that build up in other tissues are metabolized as sphingolipid derivatives such as ceramides. Several studies suggest that ceramides are among the most active lipid second messengers to inhibit the insulin signaling pathway and this review describes the major role played by ceramide accumulation in the development of insulin resistance of peripherals tissues through the targeting of specific proteins of the insulin signaling pathway.
Collapse
|
41
|
Association between haptoglobin 2-2 genotype and coronary artery disease and its severity in a tunisian population. Biochem Genet 2014; 52:269-82. [PMID: 24535155 DOI: 10.1007/s10528-014-9646-9] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2013] [Accepted: 09/09/2013] [Indexed: 12/12/2022]
Abstract
Haptoglobin (Hp) polymorphism generates three common human genotypes (Hp1-1, Hp2-1, and Hp2-2), having functional differences, related to the risk of development of cardiovascular diseases. These functions are a consequence of hemoglobin binding that leads to the synthesis of an antioxidant like ferritin. We explored the association of Hp polymorphism with significant coronary stenosis (SCS) and its severity within 400 Tunisian patients, using genotyping, biochemical parameters, and the Gensini score. After adjustments for age and gender, Hp2-2 was associated with the highest ferritin but the lowest Hp concentrations. After adjustments for confounding parameters, the OR of SCS associated with Hp2-2 was 1.74 (95% CI 1.18-2.58; p = 0.005). This effect was enhanced within diabetics (OR 1.90, 95% CI 1.11-3.24; p = 0.018), obese subjects (OR 1.98, 95% CI 1.10-4.86; p = 0.034), and smokers (OR 4.17, 95% CI 1.54-1.29; p = 0.005). The Hp2-2 genotype is associated with an increase in SCS especially in diabetics, the obese, and smokers.
Collapse
|
42
|
DeMarsilis AJ, Walji TA, Maedeker JA, Stoka KV, Kozel BA, Mecham RP, Wagenseil JE, Craft CS. Elastin Insufficiency Predisposes Mice to Impaired Glucose Metabolism. J Mol Genet Med 2014; 8. [PMID: 26167199 PMCID: PMC4497575 DOI: 10.4172/1747-0862.1000129] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
Abstract
Williams-Beuren syndrome is the consequence of a large contiguous-gene deletion on the seventh human chromosome that includes the elastin gene. Elastin is an extracellular matrix protein responsible for the cardiovascular abnormalities associated with Williams’s syndrome, including hypertension and aortic stenosis. A high percentage of individuals with Williams’s syndrome also have impaired glucose tolerance, independent of traditional risk factors for diabetes. Here, we show that murine adipose tissue does assemble elastic fibers; however, isolated elastin insufficiency (Eln+/−) in mice does not independently influence glucose metabolism or tissue lipid accumulation. Similarly, isolated ApoE deficiency (ApoE−/−), a model of hyperlipidemia and atherosclerosis, does not impair insulin sensitivity. However, Eln+/−; ApoE−/− double mutant mice exhibit notable hyperglycemia, adipocyte hypertrophy, inflammation of adipose tissue, and ectopic lipid accumulation in liver tissue. Further, Eln+/−; ApoE−/− mutants have significant impairment of insulin sensitivity by insulin tolerance testing, independent of body weight or diet, suggesting that elastin insufficiency predisposes to metabolic disease in susceptible individuals.
Collapse
Affiliation(s)
- Antea J DeMarsilis
- Department of Cell Biology & Physiology, Washington University in St. Louis, MO, USA
| | - Tezin A Walji
- Department of Cell Biology & Physiology, Washington University in St. Louis, MO, USA
| | - Justine A Maedeker
- Department of Mechanical Engineering and Materials Science, Washington University in St. Louis, MO, USA
| | - Kellie V Stoka
- Department of Mechanical Engineering and Materials Science, Washington University in St. Louis, MO, USA
| | - Beth A Kozel
- Department of Pediatrics,, Washington University in St. Louis, MO, USA
| | - Robert P Mecham
- Department of Cell Biology & Physiology, Washington University in St. Louis, MO, USA
| | - Jessica E Wagenseil
- Department of Mechanical Engineering and Materials Science, Washington University in St. Louis, MO, USA
| | - Clarissa S Craft
- Department of Cell Biology & Physiology, Washington University in St. Louis, MO, USA
| |
Collapse
|
43
|
Xu D, Yin C, Wang S, Xiao Y. JAK-STAT in lipid metabolism of adipocytes. JAKSTAT 2013; 2:e27203. [PMID: 24498541 DOI: 10.4161/jkst.27203] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2013] [Revised: 11/13/2013] [Accepted: 11/14/2013] [Indexed: 11/19/2022] Open
Abstract
JAK-STAT signaling pathway plays an important role in the cells' development and homeostasis. Over the past decades, the studies have identified the role of the JAK-STAT pathway in cell proliferation and apoptosis. Here, we want to discuss that whether and how the JAK-STAT pathway affects the lipid metabolism of adipose tissue. A host of cytokines and hormones can regulate lipid metabolism through activating the JAK-STAT signaling pathway. Activated STATs can regulate lipid metabolism directly by influencing the expression of enzymes. We have summarized the relevant research and articles of JAK-STAT during the recent years. Within this review, we will introduce you the recent research and highlight the unresolved problems in understanding how JAK-STAT signaling pathway contribute to the lipid metabolism in mature adipocytes and preadipocytes. Dysregulation of the JAK-STAT pathway would lead to a multiple metabolism disorders and medicines for this signaling pathway maybe become a new idea for diseases such as metabolic syndrome, especially in children.
Collapse
Affiliation(s)
- Dong Xu
- Department of Pediatrics; Second Affiliated Hospital of Medical School of Xi'an; Jiaotong University; Xi'an, Shaanxi PR China
| | - Chunyan Yin
- Department of Pediatrics; Second Affiliated Hospital of Medical School of Xi'an; Jiaotong University; Xi'an, Shaanxi PR China
| | - Sisi Wang
- Department of Pediatrics; Second Affiliated Hospital of Medical School of Xi'an; Jiaotong University; Xi'an, Shaanxi PR China
| | - Yanfeng Xiao
- Department of Pediatrics; Second Affiliated Hospital of Medical School of Xi'an; Jiaotong University; Xi'an, Shaanxi PR China
| |
Collapse
|
44
|
Mališová L, Kováčová Z, Koc M, Kračmerová J, Štich V, Rossmeislová L. Ursodeoxycholic acid but not tauroursodeoxycholic acid inhibits proliferation and differentiation of human subcutaneous adipocytes. PLoS One 2013; 8:e82086. [PMID: 24312631 PMCID: PMC3849437 DOI: 10.1371/journal.pone.0082086] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2013] [Accepted: 10/30/2013] [Indexed: 12/21/2022] Open
Abstract
Stress of endoplasmic reticulum (ERS) is one of the molecular triggers of adipocyte dysfunction and chronic low inflammation accompanying obesity. ERS can be alleviated by chemical chaperones from the family of bile acids (BAs). Thus, two BAs currently used to treat cholestasis, ursodeoxycholic and tauroursodeoxycholic acid (UDCA and TUDCA), could potentially lessen adverse metabolic effects of obesity. Nevertheless, BAs effects on human adipose cells are mostly unknown. They could regulate gene expression through pathways different from their chaperone function, namely through activation of farnesoid X receptor (FXR) and TGR5, G-coupled receptor. Therefore, this study aimed to analyze effects of UDCA and TUDCA on human preadipocytes and differentiated adipocytes derived from paired samples of two distinct subcutaneous adipose tissue depots, abdominal and gluteal. While TUDCA did not alter proliferation of cells from either depot, UDCA exerted strong anti-proliferative effect. In differentiated adipocytes, acute exposition to neither TUDCA nor UDCA was able to reduce effect of ERS stressor tunicamycin. However, exposure of cells to UDCA during whole differentiation process decreased expression of ERS markers. At the same time however, UDCA profoundly inhibited adipogenic conversion of cells. UDCA abolished expression of PPARγ and lipogenic enzymes already in the early phases of adipogenesis. This anti-adipogenic effect of UDCA was not dependent on FXR or TGR5 activation, but could be related to ability of UDCA to sustain the activation of ERK1/2 previously linked with PPARγ inactivation. Finally, neither BAs did lower expression of chemokines inducible by TLR4 pathway, when UDCA enhanced their expression in gluteal adipocytes. Therefore while TUDCA has neutral effect on human preadipocytes and adipocytes, the therapeutic use of UDCA different from treating cholestatic diseases should be considered with caution because UDCA alters functions of human adipose cells.
Collapse
Affiliation(s)
- Lucia Mališová
- Department of Sport Medicine, Third Faculty of Medicine, Charles University in Prague, Prague, Czech Republic
- Franco-Czech Laboratory for Clinical Research on Obesity, Third Faculty of Medicine, Prague, Czech Republic
- INSERM, Toulouse, France
| | - Zuzana Kováčová
- Department of Sport Medicine, Third Faculty of Medicine, Charles University in Prague, Prague, Czech Republic
- Franco-Czech Laboratory for Clinical Research on Obesity, Third Faculty of Medicine, Prague, Czech Republic
- INSERM, Toulouse, France
| | - Michal Koc
- Department of Sport Medicine, Third Faculty of Medicine, Charles University in Prague, Prague, Czech Republic
- Franco-Czech Laboratory for Clinical Research on Obesity, Third Faculty of Medicine, Prague, Czech Republic
- INSERM, Toulouse, France
| | - Jana Kračmerová
- Department of Sport Medicine, Third Faculty of Medicine, Charles University in Prague, Prague, Czech Republic
- Franco-Czech Laboratory for Clinical Research on Obesity, Third Faculty of Medicine, Prague, Czech Republic
- INSERM, Toulouse, France
| | - Vladimír Štich
- Department of Sport Medicine, Third Faculty of Medicine, Charles University in Prague, Prague, Czech Republic
- Franco-Czech Laboratory for Clinical Research on Obesity, Third Faculty of Medicine, Prague, Czech Republic
- INSERM, Toulouse, France
| | - Lenka Rossmeislová
- Department of Sport Medicine, Third Faculty of Medicine, Charles University in Prague, Prague, Czech Republic
- Franco-Czech Laboratory for Clinical Research on Obesity, Third Faculty of Medicine, Prague, Czech Republic
- INSERM, Toulouse, France
- * E-mail:
| |
Collapse
|
45
|
The NS, Crandell JL, Lawrence JM, King IB, Dabelea D, Marcovina SM, D'Agostino RB, Norris JM, Pihoker C, Mayer-Davis EJ. Vitamin D in youth with Type 1 diabetes: prevalence of insufficiency and association with insulin resistance in the SEARCH Nutrition Ancillary Study. Diabet Med 2013; 30:1324-32. [PMID: 23909945 PMCID: PMC3822440 DOI: 10.1111/dme.12297] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 07/30/2013] [Indexed: 01/06/2023]
Abstract
AIMS To determine the prevalence of plasma vitamin D (25-dihydroxyvitamin D) insufficiency in individuals with Type 1 diabetes and to determine the cross-sectional and longitudinal associations of plasma vitamin D with insulin resistance. METHODS Participants from the SEARCH for Diabetes in Youth Study [n = 1426; mean age 11.2 years (sd 3.9)] had physician-diagnosed Type 1 diabetes [diabetes duration mean 10.2 months (sd 6.5)] with data available at baseline and follow-up (approximately 12 and 24 months after baseline). Insulin resistance was estimated using a validated equation. Cross-sectional and longitudinal multivariate logistic regression models were used to determine the association of plasma vitamin D with insulin resistance, adjusting for potential confounders. RESULTS Forty-nine per cent of individuals had plasma vitamin D < 50 nmol/l and 26% were insulin resistant. In cross-sectional multivariate analyses, participants who had higher plasma vitamin D (65 nmol/l) had lower odds of prevalent insulin resistance than participants with lower plasma vitamin D (25 nmol/l) (odds ratio 0.70, 95% CI 0.57-0.85). This association was attenuated after additional adjustment for BMI z-score, which could be a confounder or a mediator (odds ratio 0.81, 95% CI 0.64-1.03). In longitudinal multivariate analyses, individuals with higher plasma vitamin D at baseline had lower odds of incident insulin resistance, but this was not significant (odds ratio 0.85, 95% CI 0.63-1.14). CONCLUSIONS Vitamin D insufficiency is common in individuals with Type 1 diabetes and may increase risk for insulin resistance. Additional prospective studies are needed to determine the association between plasma vitamin D and insulin resistance, and to further examine the role of adiposity on this association.
Collapse
Affiliation(s)
- N S The
- Department of Health Sciences, Furman University, Greenville, SC
| | | | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Ishikawa K, Yokote K. [Cutting-edge of medicine; dysfunction of adipose tissue and insulin resistance]. ACTA ACUST UNITED AC 2013; 102:2691-8. [PMID: 24400553 DOI: 10.2169/naika.102.2691] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Affiliation(s)
- Ko Ishikawa
- Clinical Cell Biology and Medicine, Chiba University Graduate School of Medicine, Japan
| | - Koutaro Yokote
- Clinical Cell Biology and Medicine, Chiba University Graduate School of Medicine, Japan
| |
Collapse
|
47
|
Adams Jr JD. Preventive medicine and the traditional concept of living in balance. World J Pharmacol 2013; 2:73-77. [DOI: 10.5497/wjp.v2.i3.73] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/06/2013] [Accepted: 06/19/2013] [Indexed: 02/06/2023] Open
Abstract
Chronic diseases such as arthritis, heart disease and type 2 diabetes are becoming much more common. The cost of maintaining patients inflicted with these diseases increases yearly. These diseases were less common prior to 1970. This paper will consider several questions. How do toxic lifestyles contribute to these chronic diseases What is preventive medicine How can traditional healing help educate people about disease prevention What is the traditional concept of balance and how is it important in modern medicine The dangers of obesity are discussed in terms of inflammatory adipokine and inflammatory fat production. Mechanisms of disease causation or promotion are reviewed for heart disease, type 2 diabetes, arthritis and cancer. A preventive medicine approach to preventing or perhaps curing these diseases is given which involves treating toxic lifestyles and encouraging people to live in balance. The traditional concept of balance is explained in traditional Chinese medicine terms and in scientific terms. Yin and yang are cold and hot but can also be seen as agonist and antagonist. In addition, yin and yang can be seen as rest and exercise. When yin and yang are in balance, chi flows in the body. Chi is the flow of extracellular and intracellular signaling compounds and processes in the body. When the body is in balance, it can heal itself. The traditional concept of balance should be taught as a central principle of preventive medicine.
Collapse
|
48
|
Zhao D, Liu H. Adipose tissue dysfunction and the pathogenesis of metabolic syndrome. World J Hypertens 2013; 3:18-26. [DOI: 10.5494/wjh.v3.i3.18] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/27/2013] [Revised: 08/22/2013] [Accepted: 09/04/2013] [Indexed: 02/06/2023] Open
Abstract
Metabolic syndrome is a growing research area. The underlying mechanisms of metabolic syndrome are still not very clear. Insulin resistance, obesity, inflammation and oxidative stress may play an important role in the pathogenesis of metabolic syndrome. The role of adipose tissue dysfunction is emphasized during the development of obesity. Adipose tissue is identified as a complex endocrine organ and its metabolic functions extend well beyond the classical actions of thermoregulation and of storage and release of fatty acids. Chronic low-grade inflammation activated by the immune system in adipose tissue is a key contributing factor to type 2 diabetes mellitus and cardiovascular diseases. Visceral obesity results in cell autonomous impairment in insulin signaling that leads to insulin resistance. Chronic inflammation in adipose tissue has gained acceptance as a lead promoter of insulin resistance in obesity. Furthermore, obesity creates oxidative stress conditions in adipose tissue that not only correlates with insulin resistance but is also causative in its development. Oxidative stress may be a mechanistic link between several components of metabolic syndrome and cardiovascular diseases, through its role in inflammation and its ability to disrupt insulin-signaling. The study around adipose tissue dysfunction will help to understand the pathogenesis of metabolic syndrome and may bring effective therapy in treatment of metabolic syndrome related diseases. Therefore, this review mainly focuses on the roles of adipose tissue dysfunction in inflammation, insulin resistance, and oxidative stress in the pathogenesis of metabolic syndrome.
Collapse
|
49
|
Zheng J, Wang K, Jin P, Dong C, Yuan Q, Li Y, Yang Z. The association of adipose-derived dimethylarginine dimethylaminohydrolase-2 with insulin sensitivity in experimental type 2 diabetes mellitus. Acta Biochim Biophys Sin (Shanghai) 2013; 45:641-8. [PMID: 23702602 DOI: 10.1093/abbs/gmt058] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
Asymmetric dimethylarginine (ADMA) is an endogenous inhibitor of nitric oxide synthase (NOS), which can be hydrolyzed by dimethylarginine-dimethylaminohydrolase (DDAH). It has been reported that adipocytes can produce DDAH/ADMA, but its role remains unknown. In the present study, we examined the effects of adipocyte-derived DDAH/ADMA on insulin sensitivity using animal and cell models. Results showed that in adipose tissue of high fat diet-fed diabetic rats, as well as in high glucose (25 mM) plus insulin (100 nM)-treated 3T3-L1 adipocytes, expression levels of insulin receptor substance-1 (IRS-1), glucose transporter-4 (GLUT-4), and DDAH isoform-2 (DDAH-2) were down-regulated compared with control, although DDAH-1 expression showed no significant changes. We also observed that nitric oxide bioavailability, DDAH and NOS activities were subsequently decreased, while the local ADMA content was elevated in diabetic adipose tissue. Transfection of human DDAH-2 gene into high glucose- and insulin-treated 3T3-L1 adipocytes significantly ameliorated DDAH activity, reduced ADMA contents, and up-regulated the mRNA expression levels of IRS-1 and GLUT-4. These findings suggested that in the development of type 2 diabetes mellitus, local DDAH-2 in adipocytes might play an important role in regulating insulin sensitivity.
Collapse
Affiliation(s)
- Jie Zheng
- Department of Pharmacology, School of Pharmaceutical Sciences, Central South University, Changsha 410078, China
| | | | | | | | | | | | | |
Collapse
|
50
|
Schulte W, Bernhagen J, Bucala R. Cytokines in sepsis: potent immunoregulators and potential therapeutic targets--an updated view. Mediators Inflamm 2013; 2013:165974. [PMID: 23853427 PMCID: PMC3703895 DOI: 10.1155/2013/165974] [Citation(s) in RCA: 474] [Impact Index Per Article: 43.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2012] [Accepted: 05/22/2013] [Indexed: 12/11/2022] Open
Abstract
Sepsis and septic shock are among the leading causes of death in intensive care units worldwide. Numerous studies on their pathophysiology have revealed an imbalance in the inflammatory network leading to tissue damage, organ failure, and ultimately, death. Cytokines are important pleiotropic regulators of the immune response, which have a crucial role in the complex pathophysiology underlying sepsis. They have both pro- and anti-inflammatory functions and are capable of coordinating effective defense mechanisms against invading pathogens. On the other hand, cytokines may dysregulate the immune response and promote tissue-damaging inflammation. In this review, we address the current knowledge of the actions of pro- and anti-inflammatory cytokines in sepsis pathophysiology as well as how these cytokines and other important immunomodulating agents may be therapeutically targeted to improve the clinical outcome of sepsis.
Collapse
Affiliation(s)
- Wibke Schulte
- Department of Internal Medicine, Yale University School of Medicine, The Anlyan Center, S525, P.O. Box 208031, 300 Cedar Street, New Haven, CT 06520-8031, USA
- Institute of Biochemistry and Molecular Cell Biology, University Hospital of RWTH Aachen University, Pauwelsstraße 30, 52074 Aachen, Germany
| | - Jürgen Bernhagen
- Institute of Biochemistry and Molecular Cell Biology, University Hospital of RWTH Aachen University, Pauwelsstraße 30, 52074 Aachen, Germany
| | - Richard Bucala
- Department of Internal Medicine, Yale University School of Medicine, The Anlyan Center, S525, P.O. Box 208031, 300 Cedar Street, New Haven, CT 06520-8031, USA
| |
Collapse
|